You are on page 1of 14

Clin Pharmacokinet

DOI 10.1007/s40262-013-0094-1

REVIEW ARTICLE

Pharmacokinetics and Pharmacodynamics of Anticoagulants


in Paediatric Patients
Donald L. Yee • Sarah H. O’Brien •

Guy Young

Ó Springer International Publishing Switzerland 2013

Abstract Given the rising incidence of thrombotic com- such as dabigatran, rivaroxaban and apixaban, hold prom-
plications in paediatric patients, understanding of the ise for future use in paediatrics but require further phar-
pharmacologic behaviour of anticoagulant drugs in chil- macologic study in infants, children and adolescents.
dren has gained importance. Significant developmental
differences between children and adults in the haemostatic
system and pharmacologic parameters for individual drugs 1 Introduction
highlight potentially unique aspects of anticoagulant
pharmacology in this special and vulnerable population. Thrombosis was once considered a rare disorder in children,
This review focuses on pharmacologic information rele- but multiple reports in recent years have confirmed its rising
vant to the dosing of unfractionated heparin, low molecular incidence [1–3]. Although venous thromboembolism (VTE)
weight heparin, warfarin, bivalirudin, argatroban and is the most commonly recognized manifestation of throm-
fondaparinux in paediatric patients. The bulk of clinical botic disease in paediatrics, pathologic thrombi can develop
experience with paediatric anticoagulation rests with the in the arteries as well, generally classified as either arterial
first three of these agents, each of which requires higher thrombosis or arterial ischemic stroke, depending on the site
bodyweight-based dosing for the youngest patients, com- of involvement. Each of these forms of thrombosis can
pared with adults, in order to achieve comparable phar- require treatment via anticoagulation given for several
macodynamic effects, likely related to an inverse weeks or months, according to current practice guidelines
correlation between age and bodyweight-normalized [4]. The major risk associated with anticoagulation is
clearance of these drugs. Whether extrapolation of thera- bleeding, which can have catastrophic consequences,
peutic ranges targeted for adult patients prescribed these including long-term neurologic sequelae and death. On the
agents is valid for children, however, is unknown and a other hand, the primary risk associated with inadequate
high priority for future research. Novel oral anticoagulants, anticoagulation of an existing thrombus is its growth,
recurrence or embolization, which can have similarly lasting
deleterious effects. Because optimal anticoagulant therapy
must occur within this narrow therapeutic window, the
D. L. Yee (&)
Texas Children’s Hospital, Baylor College of Medicine, pharmacologic behaviour of agents used in this setting
6701 Fannin Street, Suite 1580, Houston, TX 77030, USA requires precise understanding. Furthermore, the complex
e-mail: dlyee@txch.org array of haemostatic proteins regulating thrombus formation
and breakdown is highly age dependent, with plasma con-
S. H. O’Brien
Center for Innovation in Pediatric Practice, centrations often varying significantly between pre-mature
The Research Institute at Nationwide Children’s Hospital, infants, neonates, older infants, children, adolescents and
Columbus, OH, USA adults [5, 6] (see the next section). Finally, age-dependent
pharmacokinetic parameters such as the volume of distri-
G. Young
Children’s Hospital Los Angeles, University of Southern bution, drug elimination and drug-protein binding add a
California Keck School of Medicine, Los Angeles, CA, USA final layer of complexity to proper dosing and monitoring.
D. L. Yee et al.

Given these considerations, it is expected that in paedi- 2 Developmental Haemostasis


atric patients, anticoagulant pharmacokinetics and phar-
macodynamics would demonstrate unique, age-dependent The well-established paradigm of ‘developmental haemo-
characteristics compared with those in adults. Such differ- stasis’ holds significant implications for anticoagulant use
ences clearly hold clinical relevance for proper anticoagu- in paediatrics, given the dependence of anticoagulation on
lant management in children. Whereas other published critical interactions between these drugs and age-dependent
reviews have provided a more general, clinical perspective, plasma proteins, such as antithrombin, prothrombin and
including information on risk factors and epidemiology factor Xa [5, 6]. With respect to thrombosis, antithrombin
pertinent to thrombosis in children [7–9], this critical holds a central role as a natural anticoagulant, down-reg-
review focuses specifically on published pharmacokinetic ulating multiple procoagulant proteins but especially factor
and pharmacodynamic data on commercially available IIa (the activated form of prothrombin) and factor Xa. In
anticoagulants in paediatric patients. In compiling this the presence of heparin, binding of antithrombin to factors
review, a systematic review of the literature was performed IIa and Xa increases several hundred- to thousand-fold,
using PubMed to analyze pharmacokinetic and pharmaco- leading to accelerated deactivation of these procoagulant
dynamic studies of anticoagulant medications in paediatric molecules; antithrombin is thus also critical to the antico-
subjects. Each drug name with the ‘pharmacokinetic’ sub- agulant effect of pharmacologically administered heparin
heading attached was ‘or’ed with the drug name combined [5]. Full-term neonates have plasma concentrations of
with the term ‘pharmacodynamics’. The retrieval was then antithrombin that are only 50–75 % of adult levels, while
limited to ‘human’ and the ‘0–18 year old’ age group, and pre-term infants may have levels that are significantly
to the time period of 1988–2013. Reference lists of retrieved lower. Adult levels are typically not attained until
articles were searched for additional articles that were 6–12 months of age [6], necessitating exogenous admin-
included in this review. The background on each agent istration of antithrombin concentrate in some young infants
(Table 1) is briefly reviewed, followed by a summary and and in other children with serious medical conditions
critical analysis of relevant studies. receiving heparin anticoagulation [10].

Table 1 Basic properties of anticoagulants used in children


Anticoagulant Route of Dosing interval Half-life Antithrombin Antidote Comments
administration dependence

Heparin Intravenous Bolus followed 30 min–2 h Yes Protamine Higher bodyweight-based dosing is required
by continuous in neonates to achieve comparable anti-Xa
infusion levels; the optimal pharmacodynamic assay
for monitoring the drug effect in paediatrics
has not been determined
Enoxaparin Subcutaneous Every 12 h 6h Yes Protamine Higher bodyweight-based dosing is required
(partial) in neonates to achieve comparable anti-Xa
levels; graduated syringes are available for
only certain vial sizes, necessitating
judicious prescribing for paediatric
patients; a multiple-dose vial is available
Warfarin Oral Once daily 40 h No Vitamin K, Higher bodyweight-based dosing is required
factor in infants and young children to achieve a
repletion comparable INR; no liquid formulation is
available
Bivalirudin Intravenous Bolus followed 25 min No None The mean half-life is 15–18 min in infants
by continuous and children; bodyweight-based clearance
infusion decreases with age
Lepirudin Intravenous Continuous 70 min No None No prospective studies have been published
infusion
Argatroban Intravenous Continuous 40 min No None This is the only anticoagulant with specific
infusion paediatric dosing information on the drug
label
Fondaparinux Subcutaneous Once daily 17 h Yes None The limited available information in children
suggests a similar pharmacologic profile to
that of adults
INR international normalized ratio
Pharmacokinetics and Pharmacodynamics of Anticoagulants in Paediatric Patients

3 Unfractionated Heparin of action and the ultimate physiologic and clinical effect of
UFH in paediatric patients. These lines of evidence have
Unfractionated heparin (UFH) comprises a heterogeneous included an increase in the antithrombotic effect, as measured
mixture of glycosaminoglycans derived from porcine intes- by anti-Xa [19] and anti-IIa activity [18], and a decrease in the
tine or bovine lung, and functions as an anticoagulant by anti-Xa/anti-IIa ratio with increasing age [18, 20] for a given
potentiating the inhibitory effects of antithrombin on UFH dose. The physiology underlying this age dependence is
thrombin (factor IIa) and factor Xa, as well as tissue factor not fully understood but may be related to age-related differ-
pathway inhibitor [11]. Its major advantages include its long ences in the protein structure and/or binding between UFH and
history of clinical use and its short half-life and easy the many other moieties with which it interacts [21]. More-
reversibility—the latter two are advantageous in critical care over, the clinical laboratory assays used to monitor UFH’s
and perioperative settings. Its significant limitations include pharmacodynamic effects appear to perform differently in
UFH’s high inter- and even intra-patient variability in the children compared with adults. For example, although treat-
dose response, leading to poor achievement and mainte- ment guidelines for thrombotic disease in adults imply that the
nance of the therapeutic effect, and potentially an increased UFH concentration as measured by protamine titration
risk of bleeding or worsening thrombosis. This variability between 0.2 and 0.4 U/mL should correspond to anti-Xa
necessitates frequent monitoring and exists in addition to the activity of 0.35–0.7 U/mL and aPTT values that are elevated
inherent technical limitations of the assays [the activated but usually measurable [22], recent studies have demonstrated
partial thromboplastin time (aPTT) and anti-factor Xa assay] that these target ranges do not in fact correspond with each
most commonly used to measure anticoagulant or anti- other in children [14]. Furthermore, aPTT values corre-
thrombotic effects [12–14], as discussed further below. sponding to these target ranges are often elevated signifi-
cantly, even to the point of being unmeasurable [14, 23].
3.1 Pharmacokinetics In a recent study of 100 young infants requiring UFH via
continuous infusion, only 15 % of patients achieved anti-
Clinical pharmacokinetic data on UFH use in adults has Xa activity in the target range within 24 h of therapy ini-
been summarized, including that UFH has a half-life of tiation, compared with 81 % who achieved a target aPTT
30 min to 2 h, which varies directly with the dose admin- within the same timeframe [24], a percentage comparable
istered, has a usual volume of distribution approximating the with those observed in other cohorts for which the aPTT
plasma volume, and does not largely depend on renal was utilized [25]. Perhaps not surprisingly, correlation of
elimination mechanisms [15]. In comparison, a recent pae- these two commonly used pharmacodynamic measures
diatric study characterized UFH’s pharmacokinetic behav- with each other [24, 26, 27], as well as with the pharma-
iour using a one-compartment model, describing its mean cokinetic measure of heparin concentration determined by
clearance, volume of distribution and half-life (0.6 mL/kg/min, protamine titration (not routinely used in clinical practice
37.3 mL/kg and 45.6 min, respectively) in a group of 64 due to the lack of an automated method) [14], are poor,
children who received a single bolus dose prior to cardiac raising additional questions about the optimal assay
angiography [16]. In distinction, a similar analysis in 25 method for monitoring antithrombotic therapy with UFH in
preterm newborns revealed increased clearance, an children [28]. The specific assay reagents used in a given
increased volume of distribution and a shorter half-life in laboratory also appear to be extremely important [13].
this even younger population (1.37–1.49 mL/kg/min, Despite these issues, current guidelines for antithrom-
58–81 mL/kg and 36–42 min, respectively) [17]. Further botic therapy in children [4] continue to utilize a body-
paediatric pharmacokinetic data are limited, but these results weight-based dosing nomogram that accounts for patient
in aggregate suggest significant age-dependent pharmaco- age dichotomously between infants (\1 year old) and all
kinetic differences between neonates, infants, children, other children, and extrapolate pharmacodynamic moni-
adolescents and adults—differences that may in part account toring parameters directly from evidence obtained from
for the age-related pharmacodynamic differences described adult patients [22]. Although dose adjustments beyond
below. Indeed, the UFH concentration itself showed sig- what is outlined in the current dosing nomogram [4] are not
nificant age dependence in another paediatric study, typically suggested for specific patient populations, a
increasing by 0.6 U/mL for every year of patient age in recent retrospective study has suggested that obese patients
samples collected soon after bolus injection of UFH [18]. may require a lower bodyweight-based dose in order to
achieve and maintain comparable anticoagulation; the
3.2 Pharmacodynamics study further confirmed discrepancies between the aPTT
and anti-Xa monitoring assays [29].
From the standpoint of pharmacodynamics, evidence has Recent modifications to the United States Pharmacopeia
accumulated that supports age dependence of the mechanism monograph for UFH may have resulted in a change in the
D. L. Yee et al.

potency and hence the pharmacodynamic effect of UFH reproducibly achieved an anti-Xa level of 0.5–1.0 IU/mL
produced and marketed in the US, at least as measured by with a dose of 1 mg/kg. Infants \2 months of age (n = 6)
the activated clotting time (ACT), the method most com- required higher doses (average dose 1.64 mg/kg twice
monly used to monitor UFH in the setting of cardiovascular daily). All subjects had measurable levels of anti-Xa at
surgery [30], although it is known that the ACT correlates hour 12, demonstrating that twice-daily dosing was feasible
poorly with the heparin concentration [31]. At this time, in paediatric patients.
however, no additional paediatric studies have corrobo- In 2000, Punzalan et al. [37] performed a study in six
rated this finding using alternative pharmacodynamic patients receiving twice-daily enoxaparin (1 mg/kg per
measures such as the aPTT or anti-Xa activity. dose). Enoxaparin pharmacodynamics were found to
As with other anticoagulants discussed below, appro- approximate the profile in adults. In 2005, Kuhle et al. [38]
priate therapy targets for UFH use in children may differ performed a single-centre, open-label, phase II study of
from those for adults due to underlying age-related dif- tinzaparin, administered once daily (175 IU/kg; 250 IU/kg
ferences attributable to developmental haemostasis, such as in infants \3 months) in 35 children with VTE. Population
in thrombin formation and regulation [32]. In accordance pharmacokinetic analysis using nonlinear mixed-effect
with a recent International Society on Thrombosis and modelling techniques was performed. Samples were
Haemostasis position paper [28], future research must first obtained after patients had achieved target anti-Xa levels.
identify the optimal pharmacodynamic assay method and Pharmacokinetic data for anti-Xa activity were reported by
then incorporate paediatric-specific clinical outcome data age group and included maximum observed anti-Xa
in formulating the most appropriate target therapeutic activity ranging from 0.63 to 0.78 IU/mL, a time of max-
range for optimal use and monitoring of UFH in children. imum activity (Tmax) ranging from 2.2 to 4.3 h, a half-life
ranging from 3.45 to 8.83 h, and extravascular plasma
clearance ranging from 0.023 to 0.048 L/h/kg. Younger
4 Low Molecular Weight Heparins children had higher dose requirements, more rapid clear-
ance, a shorter time to peak anti-Xa levels, and an
Low molecular weight heparins (LMWHs) are derived from increased volume of distribution, and were more likely to
UFH but are enriched for shorter-length polysaccharide be below the target anti-Xa range than older children, with
chains. This feature changes several of the LMWHs’ proper- the majority of patients \1 year of age being subthera-
ties [33]. First, LMWHs have a more profound inhibitory peutic. Based on their findings, the authors recommended
effect on factor Xa than on thrombin, with resulting ratios of that anti-Xa levels be drawn 2–3 h post-injection in patients
the anti-Xa to anti-IIa effect of 1.5 to[10, which may confer a \5 years of age, and that anti-Xa levels be monitored at
lower bleeding risk [34, 35]. In addition, LMWHs have stable least twice monthly in infants due to their rapid growth.
pharmacokinetics and thus a more predictable clinical While enoxaparin is typically administered twice daily,
response than UFH, resulting in a less frequent need for it has also been used successfully as a once-daily medi-
monitoring. LMWHs are also administered rather conve- cation in the treatment of adult VTE. In 2007, O’Brien
niently via the subcutaneous route and possess longer half- et al. [39] published a dose-finding study of once-daily
lives, making them particularly useful in the outpatient setting. enoxaparin in 14 paediatric patients (aged 3 months–
As a result, LMWH use in children has increased substantially 16 years). Target anti-Xa activity at hours 4–6 (1–2 IU/mL
over the past decade, replacing UFH as the agent of choice in in adults) was achieved in only one patient using the adult
the initial treatment of thrombosis and joining warfarin as a recommended dose of 1.5 mg/kg. Eight children required a
viable option for longer-term management [1]. Although final dose of [2 mg/kg to achieve target activity. Eight
LMWHs appear to be the most commonly used anticoagulants patients completed 24-h pharmacodynamics monitoring.
in children, pharmacokinetic–pharmacodynamic studies have The shape of the pharmacodynamics curve in this cohort
been sparse. Most were performed based on measurements of approximated the profile in healthy adults after adminis-
anti-Xa activity and so are technically pharmacodynamic in tration of 1.5 mg/kg [40]. However, total drug exposure
nature. A review published in 2008 identified five paediatric differed, with a mean area under the plasma concentration–
LMWH pharmacologic studies [34]. time curve (AUC) of 10.1 IUh/mL in children, compared
with 16.4 IUh/mL in adults (P \ 0.001). Children also
4.1 Pharmacology demonstrated faster clearance; seven of eight participants
demonstrated sub-therapeutic anti-Xa activity (\0.5 IU/mL)
The initial paediatric dose-finding study of an LMWH was by hour 12 post-dose, and four of the eight had undetectable
published in 1996 [36]. In this prospective study of 20 levels by hour 18 post-dose.
patients receiving enoxaparin, anti-Xa levels peaked 4 h These findings of sub-optimal pharmacodynamics from
after a subcutaneous injection of enoxaparin. Ten children once-daily dosing of enoxaparin contrast with those of a
Pharmacokinetics and Pharmacodynamics of Anticoagulants in Paediatric Patients

Fig. 1 Two-compartment
population pharmacokinetics
model describing enoxaparin
kinetics for a twice-daily
enoxaparin dosing; and b once-
daily enoxaparin dosing in 126
paediatric patients. The dashed
lines indicate the 90 %
confidence interval, the solid
line indicates the model-
predicted median and dots
indicate the observed anti-factor
Xa (anti-FXa) activity levels.
The starting dose was 1 mg/kg
(1.5 mg/kg for patients
\12 months of age), adjusted to
achieve a peak anti-FXa level of
0.5–0.8 IU/mL. Reproduced
with permission from Trame
et al. [41]. Copyright 2010, John
Wiley & Sons, Inc.

more recent and larger study performed by Trame et al. proportion of children with measureable levels at
[41], in which a population pharmacokinetic model was hour 24.
developed using anti-Xa activity data from 126 children Two studies have investigated the pharmacodynamics of
receiving enoxaparin. Patients received enoxaparin prophylactic (low-dose) LMWH in paediatric patients at
1 mg/kg twice daily during the acute treatment period risk of VTE. In a study of 154 children undergoing open
(7–14 days) and transitioned to either once-daily dosing heart surgery (with a peak anti-Xa target of 0.2–0.4
(starting dose 1.5 mg/kg/day) or twice-daily dosing IU/mL), it was demonstrated that age and bodyweight
(starting dose 1 mg/kg/day) to complete their course of influenced the pharmacokinetic parameters of nadroparin
anticoagulation [42]. In both treatment arms, the prede- calcium, with bodyweight being the covariate that pro-
fined target anti-Xa activity was 0.5–0.8 IU/mL at hour 4 duced the largest improvement in the model fit [43]. Age
and [0.1 IU/mL at hour 24 [41]. A two-compartment was redundant when bodyweight was included in the
model was found to be adequate for describing enoxap- model. Large inter-patient variability was also described
arin kinetics. Bodyweight (as opposed to age and body and was only partially explained by bodyweight. Massi-
surface area) proved to be the most predictive covariate cotte et al. [44] performed a pharmacologic study of pro-
for clearance and the central volume of distribution. phylactic doses of reviparin in 19 children with central
Parameter estimates were as follows: clearance 15 mL/kg/h, venous lines. Eighty percent of anti-Xa levels were within
central volume of distribution 169 mL/kg, intercompart- the target range, with infants aged \3 months (starting
mental clearance 58 mL/h, peripheral volume of distri- dose 50 IU/kg twice daily) and older children (starting
bution 10 L and absorption rate 0.414/h. Inter-individual dose 30 IU/kg twice daily) having similar average peak
variability was found to be 54 % for clearance and 42 % levels (0.26 IU/mL) and trough levels (0.13 IU/mL). Intra-
for the volume of distribution, underscoring the need for individual variability in pharmacologic profiles was higher
close monitoring of anti-Xa activity and dose individu- in patients \3 months of age.
alization in paediatric patients. The median 24 h trough Several other relevant prospective [45–49] and retro-
level was above 0.1 IU/mL in only 53.2 % of study spective studies [50–56] on the use of LMWH for treat-
samples. However, a substantial proportion of children on ment of paediatric VTE have been published. These studies
once-daily dosing had peak anti-Xa levels below the highlighted the need for higher doses of enoxaparin in
target range (Fig. 1). Increasing the enoxaparin dose infants \2 months of age to achieve target anti-Xa levels,
based on these peak levels may have led to a higher especially for neonatal dosing higher than the currently
D. L. Yee et al.

recommended 1.5 mg/kg every 12 h [57]. Possible mech- underlying disorders such as renal insufficiency or con-
anisms that may account for the altered pharmacodynamics genital heart disease may require lower doses and closer
of enoxaparin in neonates include decreased thrombin monitoring. What remains unclear is whether the anti-Xa
generation, decreased vitamin K-dependent coagulation targets (0.5–1.0 IU/mL) utilized in the management of
factors, and decreased natural anticoagulants such as anti- adult thrombosis are appropriate for younger patients. To
thrombin and protein C [57]. Another large retrospective date, no clinical studies have assessed the safety and
study demonstrated two- to threefold variation in individ- efficacy of this target range in children. Additional pae-
ual dose requirements for LMWH and a need for higher diatric pharmacologic studies in LMWH are needed to
enoxaparin dosing in children up to 5 years of age [53]. (i) compare once- versus twice-daily dosing of enoxapa-
The increased dosing needs of younger children appear to rin; (ii) determine the timing of peak target anti-Xa levels
be due not only to differences in absorption and drug in various age groups; and (iii) most importantly, deter-
clearance but also to differences in competitive binding to mine the relationship between anti-Xa levels and efficacy
the endothelium, as studies of plasma samples from healthy and safety for paediatric patients.
infants, children, and adults have demonstrated that the
in vitro response to enoxaparin, as measured by the anti-Xa
assay, is actually not age dependent [58]. In a retrospective 5 Vitamin K Antagonists
review of two cohorts, Bauman et al. [50] showed that
using a higher starting dose in neonates (1.7 mg/kg dose) Orally administered vitamin K antagonists (VKAs) are
and older children (1.2 mg/kg dose) results in faster currently the most commonly used anticoagulants in adults
attainment of therapeutic anti-Xa levels and significantly and are also frequently used in children to prevent and treat
fewer venipunctures as compared with standard dosing (1.5 thrombotic events. Most of the published data in children
and 1.0 mg/kg). Fluctuations in anti-Xa levels during pertain to warfarin [60, 61], which is the focus of this
maintenance enoxaparin therapy also appear to be a con- section, but limited information exists for acenocoumarol
cern, particularly in neonates [46, 51, 52, 54]. However, in [62–64], phenprocoumon [65–67] and fluindione [64, 68].
these retrospective studies, no correlation was found The major advantage of warfarin is the oral route of
between the quality of LMWH therapy (time in the target administration and long half-life, allowing for once-daily
range) and treatment outcomes or adverse events [51, 52]. dosing. However, warfarin’s significant disadvantages
While it is known that children with renal disease include its narrow therapeutic index, its numerous drug
require lower doses of LMWH due to the renal clearance of interactions, its dependence on an individual’s vitamin K
these medications, studies have also shown that children status and the lack of an adequate liquid formulation for
with congenital heart disease appear to be at risk of supra- facile dosing in younger patients. Finally, the required
therapeutic anti-Xa levels [49, 52]. Very few studies have frequent monitoring of the drug’s pharmacodynamic effect
investigated the effects of obesity, an increasing epidemic is inconvenient and can be difficult, especially in infants
in paediatrics, on the pharmacokinetics and dosing of and small children.
LMWH in children. A recent retrospective study has sug- Warfarin is a racemic mixture of R and S enantiomers,
gested that dosing adjustments to the usual bodyweight- the latter possessing about three to five times the antico-
based enoxaparin initiation regimen may not be required, agulant potency of the former [69]. The drug is rapidly and
but also that more frequent therapeutic drug monitoring nearly completely absorbed, achieving the peak plasma
may be advisable in obese patients [59]. Additionally, concentration (Cmax) between 2 and 6 h after dosing [70].
many children receive LMWH injections via a subcutane- It binds extensively to plasma proteins such as albumin and
ous catheter in order to reduce the number of needle is metabolized by the hepatic cytochrome P450 (CYP)
insertions, but it is not known how LMWH absorption may system for elimination; only trace amounts of warfarin
differ with use of such catheters. appear in the urine. The elimination half-life of S-warfarin
In summary, published studies of LMWH pharmacol- is between 35 and 40 h [70]. R-warfarin has an even longer
ogy in paediatrics have clearly established the age- half-life, due to its reduced clearance, but is not discussed
dependent dosing required to achieve the anti-Xa target further in this review, given the anticoagulant dominance
(0.5–1.0 IU/mL) recommended for adults. It is also clear of the S-congener. Warfarin exerts its anticoagulant effect
that bodyweight is the strongest predictor covariate of through competitive inhibition of vitamin K epoxide
LMWH pharmacodynamics. Neonates and younger reductase (VKOR), which regenerates vitamin K in its
children require higher doses of LMWH due to faster reduced form. This reduced form of vitamin K is required
clearance, an increased volume of distribution and as a cofactor for gamma carboxylation of vitamin
developmental changes in haemostasis. Children with K-dependent clotting factors and intrinsic anticoagulants.
Pharmacokinetics and Pharmacodynamics of Anticoagulants in Paediatric Patients

5.1 Pharmacokinetics [72]. It has also been recognized that other aspects of
developmental haemostasis, such as increased levels of
Specific pharmacokinetic data on warfarin in paediatric haemostatic regulatory plasma proteins like alpha-2 mac-
patients are limited, but a cross-sectional study from Japan roglobulin, could influence the response to warfarin in
of 38 prepubertal patients (1–11 years), 15 pubertal paediatric patients [73].
patients (12–18 years) and 81 adult patients on long-term Such challenges of warfarin management in paediatrics,
warfarin therapy is instructive [71]. In this study, mean especially in younger patients, were well documented in
plasma concentrations of S-warfarin did not differ by age. the largest prospective study to date, which included 319
However, bodyweight-normalized clearance of the drug consecutive patients seen over a 5-year period at a single
was greater among prepubertal patients compared with centre, treated with warfarin under a uniform dosing and
adults and showed a negative correlation with age [71]. monitoring regimen [61]. Compared with older paediatric
Interestingly, drug clearance normalized for liver weight patients, patients under 1 year of age required increased
was similar between the prepubertal and adult groups, bodyweight-based dosing, longer periods of time to
suggesting that liver size may be of primary importance in achieve the targeted INR range and more frequent INR
determining dosing requirements, potentially accounting monitoring and dose adjustments; they also had fewer INR
more completely for warfarin dosing needs than such values within the target range. Children between 1 and
currently ascribed parameters as bodyweight, age and 6 years of age also demonstrated similar findings when
height (see the next subsection). Pharmacokinetic param- compared with patients between 7 and 18 years of age.
eters for pubertal patients did not differ significantly from Clinical factors such as a history of a Fontan procedure; use
those for adults (Table 2) [71]. of corticosteroids, phenobarbital or carbamazepine; and
enteral nutrition were all found to influence the required
5.2 Pharmacodynamics and Clinical Experience warfarin dose and/or INR measurements [61]. In this
in Paediatrics clinical setting, 79 % of patients achieved an INR within
the target range in less than 7 days, but maintaining an INR
Warfarin’s pharmacodynamic effect is most commonly within the target range proved difficult, as only 49–61 % of
assessed in the clinical arena via the international nor- measurements from this cohort during the 5-year study
malized ratio (INR). A target INR range of 2.0–3.0 is most period were within the target range [61], consistent with
commonly applied, although other target ranges exist, findings from other studies of patients receiving warfarin.
depending on the specific clinical setting (e.g., 2.5–3.5 or A highly clinically relevant finding from the study by
1.5–1.9) [4]. Wide variation in warfarin dosing require- Streif et al. [61] was the clear and distinctive inverse rela-
ments between individuals (especially in infants and chil- tionship between age and the maintenance dose requirement
dren) to achieve a target INR has long been recognized in per kilogram, with mean doses of 0.33, 0.15, 0.13 and
clinical practice. Historically, a large proportion of such 0.09 mg/kg/day for the age groups B1 year, [1 and \6
variability seemed attributable to other unknown variables years, C6 and \13 years and 13–18 years, respectively.
(see the next subsection on pharmacogenetics), even with Others have confirmed these findings in children; Nowak-
adoption of a uniform bodyweight-based dosing regimen Gottl et al. [66] reported that of several clinical parameters

Table 2 Pharmacokinetic parameters for S-warfarin with normalization to various physiological parameters in prepubertal, pubertal and adult
patients (adapted with permission from Takahashi et al. [71])
Parameter Prepubertal patients Pubertal patients Adult patients
(n = 38) (n = 15) (n = 81)

Plasma unbound fraction (%) 0.83 ± 0.28 0.82 ± 0.26 0.88 ± 0.17
Plasma unbound concentration (ng/mL) 1.80 ± 1.01 1.95 ± 1.02 1.93 ± 0.80
Unbound oral clearance (mL/min) 346 ± 217* 533 ± 285 637 ± 298
Unbound oral clearance normalized to bodyweight (mL/min/kg) 18.1 ± 9.2* 12.6 ± 8.1 11.6 ± 5.4
Unbound oral clearance normalized to body surface area (mL/min/m2) 575 ± 282** 387 ± 270 438 ± 200
Unbound oral clearance normalized to liver weight (mL/min/kg liver weight) 481 ± 244 419 ± 231 458 ± 209
Statistical comparisons were made among the groups for the respective parameters. The values are expressed as means ± standard deviations
* P \ 0.01 for the comparison between the prepubertal and adult groups
** P \ 0.05 for the comparison between the prepubertal and adult groups
P \ 0.05 for the comparison between the prepubertal and pubertal groups
D. L. Yee et al.

that were analyzed, age was the single most important Over the last decade, pharmacogenetic knowledge rel-
variable affecting the VKA dose requirement, accounting evant to these polymorphisms and their impact on warfarin
for 28 % of dose variation. Subsequent studies extended dosing has expanded significantly, leading to widely
these results by identifying patient height as an even better available dosing algorithms that incorporate an individual’s
variable to explain warfarin dose requirements, accounting genotypic profile along with other clinical variables, such
for 30–48 % of dose variability [68, 74]. Physiologically, as age, gender, anthropomorphic measurements and con-
these somewhat disparate findings can perhaps be best comitant medications. Utilizing these measures, recent
explained by the positive correlation between liver size, comprehensive models have accounted for approximately
patient age and height; the fact that clearance of warfarin 50–60 % of warfarin dose variability in adults and have
normalized for liver weight does not appear to be dependent proven to accurately predict individual warfarin dose
on age (Table 2) [71, 75]; the apparent direct relationship requirements [80]. However, such models and the under-
between liver size and warfarin clearance [76]; and the lying studies investigating the impact of the relevant gene
liver’s disproportionately large size compared with overall polymorphisms on warfarin dosing have generally not
patient size during early life [77]. Such notions may also included paediatric patients until very recently [66, 68, 74,
explain similar age-dependent variability in bodyweight- 81–85]—these paediatric studies are summarized in
based dosing requirements for other VKAs such as aceno- Table 3.
coumarol [62]. Additional patient-related factors such as Biss et al. [74] examined the applicability of one of
dietary vitamin K status and concurrent illnesses, which these widely utilized and validated adult-based dosing
impact vitamin K absorption, may contribute significantly algorithms [86] to a well-characterized cohort of children
to the variable pharmacodynamic response to warfarin receiving a stable dose of warfarin. They found that
within and between individuals, especially for children, although the adult-derived algorithm predicted mainte-
whose dietary intake may be variable and in whom acute nance doses for the children that were highly correlated
gastrointestinal illness is not uncommon [78]. However, with the actual doses (r = 0.76, P \ 0.001), it also con-
recommended dosing strategies for warfarin in children sistently overestimated warfarin doses by, on average,
remain entirely bodyweight-based for the time being [4], as 1.5 ± 1.4 mg/day [74], highlighting the need for dosing
they have over the last 20 years [60, 61]. strategies derived from paediatric cohorts. In fact, Biss
et al. [81] utilized the regression equation derived from this
5.3 Impact of Pharmacogenetics: Towards a More study to perform an independent validation study involving
Rational Dosing Strategy a separate cohort of 49 children on warfarin. This equation
incorporates patient height along with the warfarin indi-
In recent years, two genes—CYP2C9 and VKORC1, which cation and genotype information on CYP2C9 and VKORC1
affect warfarin pharmacokinetics and pharmacodynamics, status, since these four variables were found to account for
respectively—have been found to account for an important 72.4 % of interindividual dose variability [74]. Impor-
portion of the sources of interindividual variability in dos- tantly, the paediatric-derived equation showed better
ing requirements. A third gene, CYP4F2, affects vitamin K agreement (r = 0.833, P \ 0.001), without systematic bias
status and has been shown to have a minor effect on war- [81]. Current US Food and Drug Administration (FDA)-
farin dosing in adults [79] but was not found to be a sig- approved labelling for warfarin highlights the availability
nificant independent contributor to dosing requirements in of pharmacogenetic testing and its utility in individualizing
the two largest studies of children to date [68, 74]. CYP2C9 the required dosing for patients, but it does not include
is a cytochrome P450 microsomal hepatic enzyme that specific paediatric recommendations or guidance.
oxidizes warfarin to inactive metabolites (primarily 7-hy- Findings from the single paediatric pharmacokinetic
droxywarfarin); single nucleotide polymorphisms at resi- study of warfarin [71] must be viewed cautiously in light of
dues 144 and 359 (CYP2C9*2 and CYP2C9*3, its ethnically limited study population. Moreover, such
respectively) can result in enzyme variants with reduced pharmacokinetic information may hold limited value for an
metabolic activity, leading to relatively elevated plasma individual patient, potentially being overridden by phar-
warfarin concentrations in affected individuals compared macogenetic and other individualized specific influences.
with individuals with the wild-type gene. VKORC1 is Instead, individualized dosing algorithms that take into
the enzyme inhibited by warfarin; a polymorphism account clinical and pharmacogenetic information appear
(-1639G[A) in the promoter region of VKORC1 leads to well suited to refine warfarin dosing in children and adults,
decreased enzyme transcription and hence a lower warfarin although clinical outcome and cost-effectiveness benefits
dose requirement in affected individuals. Further details remain to be clearly demonstrated and nearly a third of
regarding these genes and their associated enzymes have interindividual dose variability remains unaccounted for
been well described in recent reviews [77, 80]. [87]. Future pharmacogenetic research should also include
Pharmacokinetics and Pharmacodynamics of Anticoagulants in Paediatric Patients

Table 3 Published reports on


Reference Na Age range (y) Key findingsb
the impact of pharmacogenetics
on warfarin dosing in Kosaki et al. [84] 31 12–34 VKORC1 variant status conferred increased warfarin
paediatrics sensitivity
Ruud et al. [85] 29 1–14 There was faster achievement of the target INR and
more frequent INRs above the target in patients
with CYP2C9 variants
Nowak-Gottl et al. [66] 59 (25) 1–19 VKORC1 accounted for 4 %, CYP2C9 accounted for
INR international normalized 0.4 % and age accounted for 28 % of warfarin
ratio dose variation
a Kato et al. [83] 48 0.42–19.25 VKORC1 variant status conferred increased warfarin
The parentheses indicate
patients receiving alternative sensitivity; age was another major covariate
vitamin K antagonists such as Biss et al. [74] 120 1–18 VKORC1 accounted for 27 %, CYP2C9 accounted
phenprocoumon [66] or for 13 % and height accounted for 30 % of
fluindione [68] warfarin dose variation
b
VKORC1 encodes vitamin K Moreau et al. [68] 118 (35) 0.25–18 VKORC1 accounted for 18 %, CYP2C9 accounted
epoxide reductase, the enzyme for 2 % and height accounted for 48 % of warfarin
inhibited by warfarin. CYP2C9 dose variation
encodes the cytochrome P450
Biss et al. [82] 51 1–17 Higher peak INR values and more values above the
microsomal hepatic enzyme that
target range at warfarin initiation were observed in
oxidizes warfarin to inactive
patients with VKORC1 and CYP2C9 variants
metabolites

paediatric patients with more diverse ethnic backgrounds to adults undergoing percutaneous coronary interventions.
more robustly validate recent comprehensive dose predic- The first prospective study of a DTI in children was per-
tion models, given the predominance of white children in formed using bivalirudin in 16 infants less than 6 months
study groups to date (Table 3). Finally, as for other anti- of age with VTE in a single-arm, open-label, dose-finding,
coagulants discussed in this review, the relationship safety and efficacy study [92]. This age group was spe-
between warfarin pharmacodynamics and clinical efficacy cifically chosen due to the physiologically low levels of
and toxicity in paediatrics also lacks sufficiently clear antithrombin in infants. The study utilized only a phar-
definition. macodynamic parameter (the aPTT) to assess the degree of
anticoagulation in order to titrate dosing. The study defined
the dosing for children of this age, utilizing a bolus fol-
6 Direct Thrombin Inhibitors (Intravenous) lowed by a continuous infusion. With respect to the bolus,
12 of 13 subjects with evaluable post-bolus testing
Three direct thrombin inhibitors (DTIs) have been used in achieved the target aPTT range, while 15 of the 16
children: bivalirudin, argatroban and lepirudin. Several case achieved the target aPTT range at the first measurement
reports and series have described children treated with these following initiation of the continuous infusion. A second,
agents [88–91]. As their name implies, all DTIs directly similarly designed, prospective study of bivalirudin in
inhibit thrombin, in contrast to UFH and LMWH, which older children (aged 6 months–18 years) with VTE was
exert their effect indirectly via antithrombin. The advantages recently completed, which enrolled 18 patients and inclu-
of DTIs are that they do not require antithrombin for their ded evaluation of both pharmacokinetics (plasma biva-
function, they have more predictable pharmacokinetics and lirudin concentrations) and pharmacodynamics (the aPTT)
pharmacodynamics, and they inhibit both circulating and [93]. Of note, the appropriate bodyweight-based dosing
clot-bound thrombin. The main disadvantage is their intra- strategy appeared to be the same for older children in this
venous administration, mostly by continuous infusion; they study as that used for infants in the earlier study.
are used only in the inpatient setting. Another significant Bivalirudin was also studied prospectively in 110 chil-
disadvantage is the general lack of paediatric experience dren undergoing cardiac catheterization for the prevention
with these agents. While small clinical trials have been of thrombosis, about a third of whom were infants [94].
completed for argatroban and bivalirudin, no paediatric trials This single-arm, safety, efficacy and dose-finding study
have been performed for lepirudin. used a higher dosing regimen than the VTE studies
described above, and similar to that used for the licensed
6.1 Bivalirudin adult indication (a bolus of 0.75 mg/kg/h and a continuous
infusion of 1.75 mg/kg/h). The data from this study dem-
Bivalirudin, a synthetic, rationally designed analogue of onstrated that the pharmacokinetic–pharmacodynamic
hirudin, is indicated for the prevention of thrombosis in response of bivalirudin was similar in children and adults,
D. L. Yee et al.

although there was a trend towards increased Cmax, mean


AUC and mean average concentration (Cavg) values with
increasing age from neonates to older children [94]. Mean
half-life values ranged from 15 to 18 min across all of the
age groups that were evaluated. Bodyweight-normalized
clearance decreased with increasing age (11.2 versus
5.98 mL/min/kg for neonates versus children aged 6 to
\16 years, respectively). This study utilized a different
pharmacodynamic endpoint (the ACT); a clear correlation
between the pharmacokinetic and pharmacodynamic
parameters in this study was not established, which may
reflect limitations of the ACT assay.
These three clinical trials of bivalirudin have provided
the largest prospectively collected pharmacologic data set
for any single anticoagulant in paediatrics, yet the hetero-
geneity in patient populations and dosing schemes should
Fig. 2 This figure, derived from pharmacometric simulations per-
be noted. Although the studies of paediatric VTE totalled formed on raw data, was used to determine the most appropriate
only 34 patients, both studies ultimately confirmed the initial dose of argatroban in children. The dose in lg/kg/min is shown
same dosing regimen for infants and children for this on the x-axis. The left y-axis depicts the percentage of patients
expected to have an activated partial thromboplastin time (aPTT)
indication. Despite bivalirudin’s significant pharmacologic
within the target range of 1.5–3 times the baseline value (squares).
advantages over UFH, lack of general clinical experience The right y-axis depicts the percentage of patients expected to exceed
with this agent has prevented its more widespread adoption the target range [[3 times the baseline aPTT value (triangles)]. Since
as an anticoagulant in children. nearly 60 % of patients are expected to be within the target range
and only 5 % are expected to exceed this range at a dose of 0.75
lg/kg/min, 0.75 lg/kg/min was chosen as the appropriate initial dose.
6.2 Argatroban Adapted with permission from Madabushi et al. [96]. Copyright 2011,
John Wiley & Sons, Inc.
Argatroban is a small molecule analogue of the amino
acid arginine, is approved in adults for the treatment of 96]. Although lepirudin, bivalirudin and fondaparinux are
heparin-induced thrombocytopenia (HIT) and is the only all acceptable options, only argatroban has been prospec-
anticoagulant with information on paediatric use in the tively evaluated for this purpose. It should be noted that
prescribing information. The prospective study that pro- argatroban is excreted mainly by the liver, and while FDA-
vided this dosing information evaluated the safety, efficacy guided dosing recommendations for argatroban are avail-
and dosing of argatroban in 18 children with either docu- able for paediatric patients with liver impairment, an
mented or suspected HIT or who required an alternative alternative agent with elimination independent of liver
anticoagulant for other reasons [95]. A detailed pharma- function, such as bivalirudin, may be appropriate.
cometric analysis was undertaken [96], which supported
the dosing schema now approved by the FDA. In this study,
a two-compartment population pharmacokinetic model was 7 Fondaparinux Sodium
adopted, which examined argatroban concentrations and
aPTT results from blood samples obtained from the clinical Fondaparinux is a synthetic, antithrombin-dependent inhibi-
study. The key results demonstrated that the pharmacoki- tor of factor Xa, approved in adults for the initial manage-
netic–pharmacodynamic relationship for argatroban in ment of VTE. It is given subcutaneously and has (relative to
paediatric patients is similar to that in adult patients, with a LMWH) a long half-life, such that it is dosed once daily.
linear relationship between argatroban levels and the aPTT Other advantages include no risk of HIT or contaminants,
in the region of therapeutic efficacy. From these data, a and no adverse effect on bone mineralization (in contrast to
prediction model was developed from which the recom- UFH and LMWH). Several case reports and one prospective
mended initial infusion dose of 0.75 lg/kg/min was cho- study have described its use in children [97–99].
sen, which now appears in the paediatric section of the The only prospective clinical trial of fondaparinux in
argatroban prescribing information (Fig. 2). children was a single-arm, open-label, dose-finding and
Based on these important findings, including specific safety study of 24 patients between 1 and 18 years of age
paediatric dosing information derived from the single [99]. The pharmacologic analysis featured a pharmacody-
prospective study and subsequent pharmacometric analy- namic endpoint (fondaparinux-based anti-factor Xa levels)
sis, argatroban is the drug of choice in paediatric HIT [95, and demonstrated that nearly all patients were therapeutic
Pharmacokinetics and Pharmacodynamics of Anticoagulants in Paediatric Patients

once-daily dosing. As such, the adoption of fondaparinux


into clinical use in paediatrics may increase in the future.

8 Novel Oral Anticoagulants

In recent years, three novel oral anticoagulants (dabigatran,


rivaroxaban and apixaban) have been approved by major
regulatory agencies and are thus widely available for a
variety of adult indications worldwide. Although the
pharmacology of these agents is well characterized for
adults [101], no specific paediatric studies have been
published to date. However, each agent is being studied
systematically in children and adolescents, including spe-
cific pharmacokinetic and pharmacodynamic studies.
These results, as well as subsequent initiation of larger
clinical trials examining the safety and efficacy of these
Fig. 3 The observed fondaparinux concentration data from the agents in paediatrics, are expected in the near future. For-
paediatric study (open circles) were overlaid on a plot of a simulation tunately, the need for such underlying rigour has become
of 1,000 adult subjects receiving the recommended dosing regimen widely recognized (and is required by drug regulatory
for treatment. The solid line indicates the median and the dashed lines
authorities), leading to well-defined and adequately funded
indicate the 95% prediction interval. The adult doses are the approved
bodyweight-based dosing for venous thromboembolism (5 mg for paediatric drug development programs for these and other
\50 kg, 7.5 mg for 50–100 kg and 10 mg for [100 kg). The novel anticoagulants in the pipeline [9, 28].
paediatric dosing was 0.1 mg/kg up to a maximum of 7.5 mg. It
can be seen that the observed data from the paediatric study fall
within the 95 % prediction interval of the adult data. Thus, the
fondaparinux exposure achieved in this paediatric population was 9 Conclusions
similar to that observed in adults given treatment doses of fondapar-
inux. Adapted with permission from Young et al. [99]. Copyright Use of anticoagulants in paediatrics has suffered from a
2011, John Wiley & Sons, Inc.
lack of prospectively designed studies. This dearth of
high-quality information has applied as much to pharmaco-
kinetics and pharmacodynamics—the focus of this review—
after the first dose and that once-daily dosing led to ther-
as to other arenas, such as drug safety and efficacy, leading
apeutic anticoagulation for 24 h in each patient. Further-
to frequent dilemmas for the practising clinician. For each
more, a population pharmacokinetic model demonstrated
anticoagulant, the most pressing need is better under-
that a dose of 0.1 mg/kg once daily in children recapitu-
standing of the relationship between the drug’s pharma-
lated the pharmacology of fondaparinux in adults. A
cokinetics–pharmacodynamics and its efficacy and
two-compartment model with first-order absorption and
toxicity. Only then can the most age- and indication-
elimination adequately fitted the data, and the pharmaco-
appropriate therapeutic window be targeted, as opposed to
kinetic parameters were in good agreement with the adult
current practice, whereby anticoagulant monitoring and
data. Simulations were performed to assess the 0.1 mg/kg/day
dosing are merely extrapolated from experience in adults.
dose by comparing steady-state peak and trough concentra-
This approach is likely to be suboptimal, given develop-
tions achieved in paediatric patients with corresponding levels
mental differences between children and adults in haemo-
in adults. The observed paediatric data fell within the 95 %
stasis and pharmacology. In contrast, the advent of novel
confidence intervals of the expected adult levels (Fig. 3). A
anticoagulants promising increased convenience and safety
follow-up study of this cohort of patients (supplemented by
holds forth new hope for the treatment of paediatric
additional patients not in the original study) was recently
thrombosis. Inclusion of rigorous and systematic paediatric
completed, which assessed long-term pharmacodynamic peak
studies in the drug development process lays proper
measurements only [100].
groundwork for rationally based dosing strategies for
In summary, fondaparinux can be considered an alterna-
children in the years ahead.
tive to the anticoagulant most commonly prescribed in
children, LMWH. The mechanism of action and pharmaco- Acknowledgments The authors thank Linda DeMuro, MLS, and
logic properties are similar to those of LMWH. The main Susi Miller, MLIS, AHIP, from the Grant Morrow III, MD Medical
advantage of fondaparinux is its longer half-life, allowing for Library at Nationwide Children’s Hospital (Columbus, OH, USA) for
D. L. Yee et al.

their assistance in designing the search strategy and performing the concentration and effect in children requiring unfractionated
initial literature search. Drs Yee and Young have no financial conflicts heparin. Thromb Haemost. 2010;103(5):1085–90.
of interest to disclose. Dr O’Brien is a member of a Bristol-Myers 19. Guzzetta NA, Miller BE, Todd K, Szlam F, Moore RH, Tosone
Squibb advisory board for paediatric apixaban studies. SR. An evaluation of the effects of a standard heparin dose on
thrombin inhibition during cardiopulmonary bypass in neonates.
Anesth Analg. 2005;100(5):1276–82 (table).
20. Newall F, Ignjatovic V, Summerhayes R, Gan A, Butt W,
References Johnston L, et al. In vivo age dependency of unfractionated
heparin in infants and children. Thromb Res. 2009;123(5):
1. Raffini L, Huang YS, Witmer C, Feudtner C. Dramatic increase 710–4.
in venous thromboembolism in children’s hospitals in the United 21. Ignjatovic V, Straka E, Summerhayes R, Monagle P. Age-spe-
States from 2001 to 2007. Pediatrics. 2009;124(4):1001–8. cific differences in binding of heparin to plasma proteins.
2. Stein PD, Kayali F, Olson RE. Incidence of venous thrombo- J Thromb Haemost. 2010;8(6):1290–4.
embolism in infants and children: data from the National Hos- 22. Hirsh J, Raschke R. Heparin and low-molecular-weight heparin:
pital Discharge Survey. J Pediatr. 2004;145(4):563–5. the seventh ACCP conference on antithrombotic and thrombo-
3. Vu LT, Nobuhara KK, Lee H, Farmer DL. Determination of risk lytic therapy. Chest. 2004;126(3 Suppl):188S–203S.
factors for deep venous thrombosis in hospitalized children. 23. Ignjatovic V, Summerhayes R, Than J, Gan A, Monagle P.
J Pediatr Surg. 2008;43(6):1095–9. Therapeutic range for unfractionated heparin therapy: age-rela-
4. Monagle P, Chan AK, Goldenberg NA, Ichord RN, Journeycake ted differences in response in children. J Thromb Haemost.
JM, Nowak-Gottl U, et al. Antithrombotic therapy in neonates 2006;4(10):2280–2.
and children: antithrombotic therapy and prevention of throm- 24. Schechter T, Finkelstein Y, Ali M, Kahr WH, Williams S, Chan
bosis, 9th ed: American College of Chest Physicians Evidence- AK, et al. Unfractionated heparin dosing in young infants:
Based Clinical Practice Guidelines. Chest. 2012;141(2 Suppl): clinical outcomes in a cohort monitored with anti-factor Xa
e737S–801S. levels. J Thromb Haemost. 2012;10(3):368–74.
5. Andrew M. Developmental hemostasis: relevance to thrombo- 25. Andrew M, Marzinotto V, Massicotte P, Blanchette V, Ginsberg
embolic complications in pediatric patients. Thromb Haemost. J, Brill-Edwards P, et al. Heparin therapy in pediatric patients: a
1995;74(1):415–25. prospective cohort study. Pediatr Res. 1994;35(1):78–83.
6. Monagle P, Barnes C, Ignjatovic V, Furmedge J, Newall F, 26. Kuhle S, Eulmesekian P, Kavanagh B, Massicotte P, Vegh P,
Chan A, et al. Developmental haemostasis: impact for clinical Lau A, et al. Lack of correlation between heparin dose and
haemostasis laboratories. Thromb Haemost. 2006;95(2):362–72. standard clinical monitoring tests in treatment with unfraction-
7. Ko RH, Young G. Pharmacokinetic- and pharmacodynamic- ated heparin in critically ill children. Haematologica. 2007;
based antithrombotic dosing recommendations in children. 92(4):554–7.
Expert Rev Clin Pharmacol. 2012;5(4):389–96. 27. Chan AK, Black L, Ing C, Brandao LR, Williams S. Utility of
8. Young G. Old and new antithrombotic drugs in neonates and aPTT in monitoring unfractionated heparin in children. Thromb
infants. Semin Fetal Neonatal Med. 2011;16(6):349–54. Res. 2008;122(1):135–6.
9. Young G. New anticoagulants in children: a review of recent 28. Newall F, Chan AK, Ignjatovic V, Monagle P. Recommenda-
studies and a look to the future. Thromb Res. 2011;127(2):70–4. tions for developing uniform laboratory monitoring of hepari-
10. Kozul C, Newall F, Monagle P, Mertyn E, Ignjatovic V. A noid anticoagulants in children. J Thromb Haemost.
clinical audit of antithrombin concentrate use in a tertiary pae- 2012;10(1):145–7.
diatric centre. J Paediatr Child Health. 2012;48(8):681–4. 29. Taylor BN, Bork SJ, Kim S, Moffett BS, Yee DL. Evaluation of
11. Newall F, Johnston L, Ignjatovic V, Monagle P. Unfractionated weight-based dosing of unfractionated heparin in obese children.
heparin therapy in infants and children. Pediatrics. 2009;123(3): J Pediatr. 2013. In Press.
e510–8. 30. Guzzetta NA, Amin SJ, Tosone AK, Miller BE. Change in
12. Kitchen S, Theaker J, Preston FE. Monitoring unfractionated heparin potency and effects on the activated clotting time in
heparin therapy: relationship between eight anti-Xa assays and a children undergoing cardiopulmonary bypass. Anesth Analg.
protamine titration assay. Blood Coagul Fibrinolysis. 2000; 2012;115(4):921–4.
11(2):137–44. 31. Martindale SJ, Shayevitz JR, D’Errico C. The activated coagu-
13. Ignjatovic V, Summerhayes R, Gan A, Than J, Chan A, Coch- lation time: suitability for monitoring heparin effect and neu-
rane A, et al. Monitoring unfractionated heparin (UFH) therapy: tralization during pediatric cardiac surgery. J Cardiothorac Vasc
which anti-factor Xa assay is appropriate? Thromb Res. 2007; Anesth. 1996;10(4):458–63.
120(3):347–51. 32. Andrew M, Mitchell L, Vegh P, Ofosu F. Thrombin regulation
14. Newall F, Ignjatovic V, Johnston L, Summerhayes R, Lane G, in children differs from adults in the absence and presence of
Cranswick N, et al. Clinical use of unfractionated heparin heparin. Thromb Haemost. 1994;72(6):836–42.
therapy in children: time for change? Br J Haematol. 33. Hirsh J, Levine MN. Low molecular weight heparin. Blood.
2010;150(6):674–8. 1992;79(1):1–17.
15. Estes JW. Clinical pharmacokinetics of heparin. Clin Pharma- 34. Nowak-Gottl U, Bidlingmaier C, Krumpel A, Gottl L, Kenet G.
cokinet. 1980;5(3):204–20. Pharmacokinetics, efficacy, and safety of LMWHs in venous
16. Newall F, Ignjatovic V, Johnston L, Lane G, Summerhayes R, thrombosis and stroke in neonates, infants and children. Br J
Cranswick N, et al. Unfractionated heparin has an age-depen- Pharmacol. 2008;153(6):1120–7.
dent pharmacokinetic profile in children. J Thromb Haemost. 35. Samama MM, Gerotziafas GT. Comparative pharmacokinetics
2009;7(s2):774. of LMWHs. Semin Thromb Hemost. 2000;26(Suppl 1):31–8.
17. McDonald MM, Jacobson LJ, Hay WW Jr, Hathaway WE. Hep- 36. Massicotte P, Adams M, Marzinotto V, Brooker LA, Andrew M.
arin clearance in the newborn. Pediatr Res. 1981;15(7):1015–8. Low-molecular-weight heparin in pediatric patients with
18. Newall F, Ignjatovic V, Johnston L, Summerhayes R, Lane G, thrombotic disease: a dose finding study. J Pediatr. 1996;
Cranswick N, et al. Age is a determinant factor for measures of 128(3):313–8.
Pharmacokinetics and Pharmacodynamics of Anticoagulants in Paediatric Patients

37. Punzalan RC, Hillery CA, Montgomery RR, Scott CA, Gill JC. 54. Lulic-Botica M, Rajpurkar M, Sabo C, Tutag-Lehr V, Natarajan
Low-molecular-weight heparin in thrombotic disease in children G. Fluctuations of anti-Xa concentrations during maintenance
and adolescents. J Pediatr Hematol Oncol. 2000;22(2):137–42. enoxaparin therapy for neonatal thrombosis. Acta Paediatr.
38. Kuhle S, Massicotte P, Dinyari M, Vegh P, Mitchell D, Mar- 2012;101(4):e147–50.
zinotto V, et al. Dose-finding and pharmacokinetics of thera- 55. Malowany JI, Knoppert DC, Chan AK, Pepelassis D, Lee DS.
peutic doses of tinzaparin in pediatric patients with Enoxaparin use in the neonatal intensive care unit: experience
thromboembolic events. Thromb Haemost. 2005;94(6):1164–71. over 8 years. Pharmacotherapy. 2007;27(9):1263–71.
39. O’Brien SH, Lee H, Ritchey AK. Once-daily enoxaparin in 56. Michaels LA, Gurian M, Hegyi T, Drachtman RA. Low molecular
pediatric thromboembolism: a dose finding and pharmacody- weight heparin in the treatment of venous and arterial thromboses
namics/pharmacokinetics study. J Thromb Haemost. 2007;5(9): in the premature infant. Pediatrics. 2004;114(3):703–7.
1985–7. 57. Malowany JI, Monagle P, Knoppert DC, Lee DS, Wu J,
40. Sanderink GJ, Le LA, Jariwala N, Harding N, Ozoux ML, McCusker P, et al. Enoxaparin for neonatal thrombosis: a call
Shukla U, et al. The pharmacokinetics and pharmacodynamics for a higher dose for neonates. Thromb Res. 2008;122(6):
of enoxaparin in obese volunteers. Clin Pharmacol Ther. 826–30.
2002;72(3):308–18. 58. Ignjatovic V, Summerhayes R, Newall F, Monagle P. The
41. Trame MN, Mitchell L, Krumpel A, Male C, Hempel G, in vitro response to low-molecular-weight heparin is not age-
Nowak-Gottl U. Population pharmacokinetics of enoxaparin in dependent in children. Thromb Haemost. 2010;103(4):855–6.
infants, children and adolescents during secondary thrombo- 59. Richard AA, Kim S, Moffett BS, Bomgaars L, Mahoney D Jr,
embolic prophylaxis: a cohort study. J Thromb Haemost. Yee DL. Comparison of anti-Xa levels in obese and non-obese
2010;8(9):1950–8. pediatric patients receiving treatment doses of enoxaparin.
42. Schobess R, During C, Bidlingmaier C, Heinecke A, Merkel N, J Pediatr. 2013;162(2):293–6.
Nowak-Gottl U. Long-term safety and efficacy data on childhood 60. Andrew M, Marzinotto V, Brooker LA, Adams M, Ginsberg J,
venous thrombosis treated with a low molecular weight heparin: Freedom R, et al. Oral anticoagulation therapy in pediatric
an open-label pilot study of once-daily versus twice-daily en- patients: a prospective study. Thromb Haemost. 1994;71(3):
oxaparin administration. Haematologica. 2006;91(12):1701–4. 265–9.
43. Laporte S, Mismetti P, Piquet P, Doubine S, Touchot A, De- 61. Streif W, Andrew M, Marzinotto V, Massicotte P, Chan AK,
cousus H. Population pharmacokinetic of nadroparin calcium Julian JA, et al. Analysis of warfarin therapy in pediatric
(Fraxiparine) in children hospitalised for open heart surgery. Eur patients: a prospective cohort study of 319 patients. Blood.
J Pharm Sci. 1999;8(2):119–25. 1999;94(9):3007–14.
44. Massicotte P, Julian JA, Marzinotto V, Gent M, Shields K, Chan 62. Bonduel M, Sciuccati G, Hepner M, Torres AF, Pieroni G,
AK, et al. Dose-finding and pharmacokinetic profiles of pro- Frontroth JP, et al. Acenocoumarol therapy in pediatric patients.
phylactic doses of a low molecular weight heparin (reviparin- J Thromb Haemost. 2003;1(8):1740–3.
sodium) in pediatric patients. Thromb Res. 2003;109(2–3):93–9. 63. Woods A, Vargas J, Berri G, Kreutzer G, Meschengieser S,
45. Bontadelli J, Moeller A, Schmugge M, Schraner T, Kretschmar Lazzari MA. Antithrombotic therapy in children and adoles-
O, Bauersfeld U, et al. Enoxaparin therapy for arterial throm- cents. Thromb Res. 1986;42(3):289–301.
bosis in infants with congenital heart disease. Intensive Care 64. Piquet P, Losay J, Doubine S. Acenocoumarol (Sintrom) and
Med. 2007;33(11):1978–84. fluinidione (Previscan) in pediatrics after cardiac surgical pro-
46. Dix D, Andrew M, Marzinotto V, Charpentier K, Bridge S, cedures. Arch Pediatr. 2002;9(11):1137–44.
Monagle P, et al. The use of low molecular weight heparin in 65. Gunther T, Mazzitelli D, Schreiber C, Wottke M, Paek SU,
pediatric patients: a prospective cohort study. J Pediatr. Meisner H, et al. Mitral-valve replacement in children under
2000;136(4):439–45. 6 years of age. Eur J Cardiothorac Surg. 2000;17(4):426–30.
47. Merkel N, Gunther G, Schobess R. Long-term treatment of 66. Nowak-Gottl U, Dietrich K, Schaffranek D, Eldin NS, Yasui Y,
thrombosis with enoxaparin in pediatric and adolescent patients. Geisen C, et al. In pediatric patients, age has more impact on
Acta Haematol. 2006;115(3–4):230–6. dosing of vitamin K antagonists than VKORC1 or CYP2C9
48. Nohe N, Flemmer A, Rumler R, Praun M, Auberger K. The low genotypes. Blood. 2010;116(26):6101–5.
molecular weight heparin dalteparin for prophylaxis and therapy 67. Wermes C, Bergmann F, Reller B, Sykora KW. Severe protein C
of thrombosis in childhood: a report on 48 cases. Eur J Pediatr. deficiency and aseptic osteonecrosis of the hip joint: a case
1999;158(Suppl 3):S134–9. report. Eur J Pediatr. 1999;158(Suppl 3):S159–61.
49. Streif W, Goebel G, Chan AK, Massicotte MP. Use of low 68. Moreau C, Bajolle F, Siguret V, Lasne D, Golmard JL, Elie C,
molecular mass heparin (enoxaparin) in newborn infants: a et al. Vitamin K antagonists in children with heart disease:
prospective cohort study of 62 patients. Arch Dis Child Fetal height and VKORC1 genotype are the main determinants of the
Neonatal Ed. 2003;88(5):F365–70. warfarin dose requirement. Blood. 2012;119(3):861–7.
50. Bauman ME, Belletrutti MJ, Bajzar L, Black KL, Kuhle S, 69. O’Reilly RA. Studies on the optical enantiomorphs of warfarin
Bauman ML, et al. Evaluation of enoxaparin dosing require- in man. Clin Pharmacol Ther. 1974;16(2):348–54.
ments in infants and children: better dosing to achieve thera- 70. Holford NH. Clinical pharmacokinetics and pharmacodynamics
peutic levels. Thromb Haemost. 2009;101(1):86–92. of warfarin: understanding the dose-effect relationship. Clin
51. Estepp JH, Smeltzer M, Reiss UM. The impact of quality and Pharmacokinet. 1986;11(6):483–504.
duration of enoxaparin therapy on recurrent venous thrombosis 71. Takahashi H, Ishikawa S, Nomoto S, Nishigaki Y, Ando F,
in children. Pediatr Blood Cancer. 2012;59(1):105–9. Kashima T, et al. Developmental changes in pharmacokinetics
52. Ho SH, Wu JK, Hamilton DP, Dix DB, Wadsworth LD. An and pharmacodynamics of warfarin enantiomers in Japanese
assessment of published pediatric dosage guidelines for enox- children. Clin Pharmacol Ther. 2000;68(5):541–55.
aparin: a retrospective review. J Pediatr Hematol Oncol. 2004; 72. Doyle JJ, Koren G, Cheng MY, Blanchette VS. Anticoagulation
26(9):561–6. with sodium warfarin in children: effect of a loading regimen.
53. Ignjatovic V, Najid S, Newall F, Summerhayes R, Monagle P. J Pediatr. 1988;113(6):1095–7.
Dosing and monitoring of enoxaparin (low molecular weight 73. Massicotte P, Leaker M, Marzinotto V, Adams M, Freedom R,
heparin) therapy in children. Br J Haematol. 2010;149(5):734–8. Williams W, et al. Enhanced thrombin regulation during
D. L. Yee et al.

warfarin therapy in children compared to adults. Thromb Hae- 88. Cetta F, Graham LC, Wrona LL, Arruda MJ, Walenga JM.
most. 1998;80(4):570–4. Argatroban use during pediatric interventional cardiac cathe-
74. Biss TT, Avery PJ, Brandao LR, Chalmers EA, Williams MD, terization. Catheter Cardiovasc Interv. 2004;61(1):147–9.
Grainger JD, et al. VKORC1 and CYP2C9 genotype and patient 89. Deitcher SR, Topoulos AP, Bartholomew JR, Kichuk-Chrisant
characteristics explain a large proportion of the variability in MR. Lepirudin anticoagulation for heparin-induced thrombo-
warfarin dose requirement among children. Blood. 2012;119(3): cytopenia. J Pediatr. 2002;140(2):264–6.
868–73. 90. Kawada T, Kitagawa H, Hoson M, Okada Y, Shiomura J.
75. Koukouritaki SB, Manro JR, Marsh SA, Stevens JC, Rettie AE, Clinical application of argatroban as an alternative anticoagulant
McCarver DG, et al. Developmental expression of human for extracorporeal circulation. Hematol Oncol Clin North Am.
hepatic CYP2C9 and CYP2C19. J Pharmacol Exp Ther. 2000;14(2):445–57, x.
2004;308(3):965–74. 91. Severin T, Zieger B, Sutor AH. Anticoagulation with recombi-
76. Murry DJ, Crom WR, Reddick WE, Bhargava R, Evans WE. nant hirudin and danaparoid sodium in pediatric patients. Semin
Liver volume as a determinant of drug clearance in children and Thromb Hemost. 2002;28(5):447–54.
adolescents. Drug Metab Dispos. 1995;23(10):1110–6. 92. Young G, Tarantino MD, Wohrley J, Weber LC, Belvedere M,
77. Visscher H, Amstutz U, Sistonen J, Ross CJ, Hayden MR, Nugent DJ. Pilot dose-finding and safety study of bivalirudin in
Carleton BC. Pharmacogenomics of cardiovascular drugs and infants \6 months of age with thrombosis. J Thromb Haemost.
adverse effects in pediatrics. J Cardiovasc Pharmacol. 2011; 2007;5(8):1654–9.
58(3):228–39. 93. O’Brien SH, Yee DL, Lira J, Goldenberg NA, Young G. Pro-
78. Biss TT, Adamson AJ, Seal CJ, Kamali F. The potential impact spective, open-label clinical trial of bivalirudin in children with
of dietary vitamin K status on anticoagulation control in children venous thromboembolism. Blood. 2012;120(21).
receiving warfarin. Pediatr Hematol Oncol. 2011;28(5):425–7. 94. Forbes TJ, Hijazi ZM, Young G, Ringewald JM, Aquino PM,
79. Caldwell MD, Awad T, Johnson JA, Gage BF, Falkowski M, Vincent RN, et al. Pediatric catheterization laboratory antico-
Gardina P, et al. CYP4F2 genetic variant alters required war- agulation with bivalirudin. Catheter Cardiovasc Interv. 2011;
farin dose. Blood. 2008;111(8):4106–12. 77(5):671–9.
80. Carlquist JF, Anderson JL. Using pharmacogenetics in real time 95. Young G, Boshkov LK, Sullivan JE, Raffini LJ, Cox DS, Boyle DA,
to guide warfarin initiation: a clinician update. Circulation. et al. Argatroban therapy in pediatric patients requiring nonheparin
2011;124(23):2554–9. anticoagulation: an open-label, safety, efficacy, and pharmacoki-
81. Biss T, Hamberg AK, Avery P, Wadelius M, Kamali F. War- netic study. Pediatr Blood Cancer. 2011;56(7):1103–9.
farin dose prediction in children using pharmacogenetics infor- 96. Madabushi R, Cox DS, Hossain M, Boyle DA, Patel BR, Young
mation. Br J Haematol. 2012;159(1):106–9. G, et al. Pharmacokinetic and pharmacodynamic basis for
82. Biss TT, Avery PJ, Williams MD, Brandao LR, Grainger JD, effective argatroban dosing in pediatrics. J Clin Pharmacol.
Kamali F. VKORC1 and CYP2C9 genotype is associated with 2011;51(1):19–28.
over-anticoagulation during initiation of warfarin therapy in 97. Boshkov LK, Kirby A, Shen I, Ungerleider RM. Recognition
children. J Thromb Haemost. 2013;11(2):373–5. and management of heparin-induced thrombocytopenia in
83. Kato Y, Ichida F, Saito K, Watanabe K, Hirono K, Miyawaki T, pediatric cardiopulmonary bypass patients. Ann Thorac Surg.
et al. Effect of the VKORC1 genotype on warfarin dose 2006;81(6):S2355–9.
requirements in Japanese pediatric patients. Drug Metab Phar- 98. Sharathkumar AA, Crandall C, Lin JJ, Pipe S. Treatment of
macokinet. 2011;26(3):295–9. thrombosis with fondaparinux (Arixtra) in a patient with end-
84. Kosaki K, Yamaghishi C, Sato R, Semejima H, Fuijita H, Tamura stage renal disease receiving hemodialysis therapy. J Pediatr
K, et al. 1173C[T polymorphism in VKORC1 modulates the Hematol Oncol. 2007;29(8):581–4.
required warfarin dose. Pediatr Cardiol. 2006;27(6):685–8. 99. Young G, Yee DL, O’Brien SH, Khanna R, Barbour A, Nugent
85. Ruud E, Holmstrom H, Bergan S, Wesenberg F. Oral antico- DJ. FondaKIDS: a prospective pharmacokinetic and safety study
agulation with warfarin is significantly influenced by steroids of fondaparinux in children between 1 and 18 years of age.
and CYP2C9 polymorphisms in children with cancer. Pediatr Pediatr Blood Cancer. 2011;57(6):1049–54.
Blood Cancer. 2008;50(3):710–3. 100. Ko RH, Michieli C, Bernardini L, Young G. FondaKids II: long-
86. Klein TE, Altman RB, Eriksson N, Gage BF, Kimmel SE, Lee term follow-up data of children receiving fondaparinux for
MT, et al. Estimation of the warfarin dose with clinical and treatment of venous thrombotic events. Blood. 2012;120(21).
pharmacogenetic data. N Engl J Med. 2009;360(8):753–64. 101. Eriksson BI, Quinlan DJ, Eikelboom JW. Novel oral factor Xa
87. Biss TT, Kamali F. Warfarin anticoagulation in children: is there and thrombin inhibitors in the management of thromboembo-
a role for a personalized approach to dosing? Pharmacoge- lism. Annu Rev Med. 2011;62:41–57.
nomics. 2012;13(11):1211–4.

You might also like