Pediatric Cardiac Intensive Care Society 2014 Consensus Statement: Pharmacotherapies in Cardiac Critical Care Anticoagulation and Thrombolysis

Page created by Judith Myers
 
CONTINUE READING
Pediatric Cardiac Intensive Care Society 2014
Consensus Statement: Pharmacotherapies in Cardiac
Critical Care Anticoagulation and Thrombolysis
       Therese M. Giglia, MD, FACC, FAAP, SCCM1; Char Witmer, MD2;
       David E. Procaccini, PharmD, BCPS3; Jonathan W. Byrnes, MD4

Objective: Thrombotic complications are increasingly being rec-              have been approved for a limited number of indications in adults,
ognized as a significant cause of morbidity and mortality in pedi-           studies on the safety and efficacy of these agents in children are
atric and congenital heart disease. The objective of this article is         pending. (Pediatr Crit Care Med 2016; 17:S77–S88)
to review the medications currently available to prevent and treat           Key Words: anticoagulation; congenital heart disease; Coumadin;
such complications.                                                          heparin; pharmacokinetics; thrombolysis
Data Sources: Online searches were conducted using PubMed.
Study Selection: Studies were selected for inclusion based on their
scientific merit and applicability to the pediatric cardiac population.

                                                                             T
Data Extraction: Pertinent information from each selected study or                   hrombotic complications are increasingly being recog-
scientific review was extracted for inclusion.                                       nized as a significant cause of morbidity and mortal-
Data Synthesis: Four classes of medications were identified                          ity in pediatric and congenital heart disease (1–3). The
as potentially beneficial in this patient group: anticoagulants,             purpose of this article is to review the medications available to
antiplatelet agents, thrombolytic agents, and novel oral antico-             prevent and treat such complications. Four classes of drugs are
agulants. Data on each class of medication were synthesized                  potentially efficacious; three classes are currently approved for
into the follow sections: mechanism of action, pharmacokinet-                the use in children: 1) anticoagulants, 2) antiplatelet agents,
ics, dosing, monitoring, reversal, considerations for use, and               and 3) thrombolytic agents. Although the fourth class, novel
­evidence to support.                                                        anticoagulants, has been approved for the use in adults with
 Conclusions: Anticoagulants, antiplatelet agents, and thrombolytic          selected conditions, none of the drugs in this class have yet been
 agents are routinely used successfully in the pediatric patient with        approved in children because the pharmacokinetics, safety, and
 heart disease for the prevention and treatment of a wide range of           efficacy have not yet been established in this population.
 thrombotic complications. Although the novel oral anticoagulants                For each drug in this section, the mechanism of action
                                                                             (MOA), pharmacokinetics, dosing, therapeutic goal, monitoring,
1
 Division of Cardiology, Department of Pediatrics, Children’s Hospital of    reversal, considerations for use, and evidence to support use will
  Philadelphia, Perelman School of Medicine at the University of Pennsyl-    be reviewed. This article should be used in conjunction with the
  vania, Philadelphia, Pennsylvania.                                         recently published guidelines for anticoagulation and treatment
2
 Division of Hematology, Department of Pediatrics, Children’s Hospital of    of thrombosis in children with pediatric and congenital heart
  Philadelphia, Perelman School of Medicine at the University of Pennsyl-
  vania, Philadelphia, Pennsylvania.                                         disease (1, 4) where specific clinical indications are elaborated.
3
 Department of Pediatrics, Pediatric Pharmacy, Johns Hopkins Medical
  Center, Baltimore, MD.
4
 Department of Pediatrics, Cincinnati Children’s Hospital Medical Center,
                                                                             ANTICOAGULANTS
  University of Cincinnati College of Medicine, Cincinnati, OH.
                                                                             Unfractionated Heparin
Dr. Witmer disclosed off-label product use: All anticoagulants but one
are off label. Dr. Byrnes’ institution received support from Daiichi-Sanyo
                                                                             MOA. Unfractionated heparin (UFH) is composed of a het-
(Edoxaban) (no direct financial support, but site PI for a phase I study.    erogeneous mixture of highly sulfated glycosaminoglycans (5).
Dr. Byrnes’ portion of the article had no mention of this or related prod-   UFH acts as an anticoagulant protein by binding antithrombin
ucts). The remaining authors have disclosed that they do not have any
potential conflicts of interest.                                             and potentiating its anticoagulant activity over 1,000-fold inacti-
For information regarding this article, E-mail: gigliat@email.chop.edu       vating coagulant factors IIa (thrombin), Xa, XIa, and XIIa (6–9).
Copyright © 2016 by the Society of Critical Care Medicine and the World         Pharmacokinetics. UFH is available as a continuous IV
Federation of Pediatric Intensive and Critical Care Societies                infusion or a subcutaneous injection. The half-life is short at
DOI: 10.1097/PCC.0000000000000623                                            1.5 ± 0.5 hours (10). Unfortunately, UFH can interact with

Pediatric Critical Care Medicine                                                                            www.pccmjournal.org            S77

                                            Unauthorized reproduction of this article is prohibited
Giglia et al

other plasma proteins, endothelial cells, and macrophages,            UFH. There can also be poor correlation between the aPTT
which alter the pharmacokinetics and anticoagulant effect             and the UFH anti-Xa level.
resulting in a high inter- and intrapatient dose-response.                Heparin resistance has been reported in up to 22% of adult
Elimination is thought to be via binding to endothelial cell          patients undergoing surgery with cardiopulmonary bypass
receptors and macrophages (11). Pediatric patients represent          (14) and may be the result of low antithrombin levels. Heparin
a challenge for heparin therapy secondary to developmental            resistance should be suspected when increased dosing does not
hemostasis with age-dependent changes in the target proteins          result in therapeutic aPTT or UFH anti-Xa levels and is treated
for UFH (12). Specifically, neonates have low antithrombin            with infusion of antithrombin or fresh frozen plasma.
levels. In addition, the clearance of UFH in infants is faster sec-       Reversal. Full reversal of heparin can be obtained with the
ondary to a larger plasma volume of distribution (13).                use of protamine sulfate (9). Approximately 1 mg of protamine
    Dosing. To achieve steady state more quickly, IV UFH is           will neutralize 100 U of UFH. Calculations should be made
usually started as a bolus followed by a continuous infusion          based on the total amount of heparin received in the prior
(4). The bolus should be deferred if there is a concern for sig-      2–2.5 hours (9).
nificant bleeding.                                                        Consideration for Use. UFH therapy can be a challenge in
    Initial loading bolus (if indicated): 75 U/kg over 10 minutes     pediatric patients secondary to the need for frequent monitor-
followed by age younger than 1 year old: continuous rate 28 U/        ing, high inter- and intrapatient dosing variability, and the need
kg/hr and age more than 1 year old: continuous rate 20 U/kg/hr.       for a dedicated line for the continuous infusion. The benefits
Further adjustments to the continuous infusion are based on           of UFH include the short half-life and the ability to completely
monitoring (see below).                                               reverse the anticoagulation effect. In addition, UFH can be used
    Monitoring. No clinical trials have determined the thera-         in the setting of renal failure, whereas low molecular weight
peutic range for UFH in pediatric patients, thus it is extrapo-       heparins (LMWH) cannot. In clinically unstable patients at risk
lated from the adult literature. The two most common assays           for hemorrhage who require anticoagulation, UFH should be
used to monitor UFH include the activated partial thrombo-            strongly considered. Transition to an alternative anticoagulant
plastin time (aPTT) and the UFH anti-Xa level.                        should occur once the patient is clinically stable.
    Therapeutic Goal. aPTT: 1.5–2.5 times control (60–85 s) or            The predominant risk associated with UFH therapy is hem-
UFH anti-Xa level: 0.3–0.7 U/mL (Table 1).                            orrhage. To minimize this bleeding risk, concurrent antiplatelet
    At this time, it is unclear as to the best method for monitor-    therapy should be avoided if possible. It is important to treat con-
ing UFH therapy in pediatric patients (4). The aPTT is not a          comitant severe thrombocytopenia and coagulopathy as well.
direct measure of the UFH effect and can be affected by many          Nonhemorrhagic complications associated with UFH include
other parameters. An elevated factor VIII or fibrinogen (acute        heparin-induced thrombocytopenia (HIT) and osteopenia.
phase reactants) will shorten the aPTT making it appear that          Prolonged use of UFH is associated with osteoporosis in adults,
the patient is heparin resistant (aPTT not prolonging despite         and there are case reports in children (15, 16). UFH-associated
receiving UFH). Alternatively, a deficiency in a coagulation fac-     HIT is characterized by thrombocytopenia and resultant arterial
tor (i.e., from liver failure or consumption) or the presence of      or venous thrombosis that can be catastrophic. The prevalence
an antiphospholipid antibody will prolong the aPTT making it          of HIT in the pediatric population appears to be less than adults
appear that the patient is therapeutic. Although the UFH anti-        with a reported range of 0–2.3% (17–20). Higher risk pediat-
Xa level is a direct measure of the heparin effect on coagulation     ric groups include those patients undergoing cardiopulmonary
factor X, it does not reflect additional anticoagulant targets of     bypass (21). The treatment of HIT includes the removal of all

Table 1.       Unfractionated Heparin Continuous Infusion Dose Titration
  Activated Partial
  Thromboplastin                Unfractionated Heparin
  Time (s)                          Anti-Xa (U/mL)                          Dose Adjustment                      Laboratory Monitoring

  < 50                                   < 0.1                        Bolus: 50 IU/kg                          4-hr postrate change
                                                                      Increase infusion rate by 10%
  50–59                                0.1–0.29                       Increase infusion rate by 10%            4-hr postrate change
  60–85                                 0.3–0.7                       No change                                4-hr then q24 hr
  86–95                                0.71–0.9                       Decrease infusion rate by 10%            4-hr postrate change
  96–120                                0.91–1                        Hold infusion for 30 min                 4-hr postrate change
                                                                      Decrease infusion rate by 10%
  > 120                                   >1                          Hold infusion for 60 min                 4-hr postrate change
                                                                      Decrease infusion rate by 15%
Supplement

heparin from the patient including avoidance of LMWH (i.e.,               Monitoring. Routine monitoring is currently recommended
Enoxaparin). Anticoagulation should be initiated with a non-          for pediatric patients receiving LMWH therapy (4). The target
heparin anticoagulant, such as a direct thrombin inhibitor (DTI)      therapeutic range is extrapolated from adult studies.
(i.e., argatroban or bivalirudin). In this setting, warfarin should       LMWH Anti-Xa (Peak Level). A dose level of 0.5–1 U/mL
never be initiated by itself due to an increased risk of skin         (drawn 4–6 hr post administration), see Monagle et al (4) for
necrosis and further thrombotic events. Warfarin can be ini-          adjustment recommendations based on peak anti-Xa level.
tiated once the platelet count has normalized and should be               Reversal. LMWH can be partially reversed (≈70%) with
overlapped with the nonheparin anticoagulant until the inter-         protamine.
nationalized ratio (INR) is therapeutic.                                  Consideration for Use. In general, LMWH is easier to use in
                                                                      pediatric patients because it does not require a dedicated line,
Evidence to Support                                                   and frequent monitoring is not needed. Unfortunately, a sub-
Indications for the use of UFH include the treatment of venous,       cutaneous injection, bid, is not insignificant for many pediatric
arterial, or intracardiac thrombi or to prevent thrombosis.           patients. It should not be used in the setting of renal failure.
Extensive reviews have been completed regarding the effective-            The predominant risk associated with LMWH therapy is
ness of UFH, and a complete discussion is beyond the scope            hemorrhage. To minimize this bleeding risk, concurrent anti-
of this article, but the authors refer readers to review the 2012     platelet therapy should be avoided if possible and treating severe
Chest guidelines on Antithrombotic Therapy in Neonates (4)            thrombocytopenia and coagulopathy. HIT has been reported
and Children as well as the 2013 American Heart Association           with the use of LMWH in pediatric patients (27, 28). Overall,
guidelines for the Prevention and Treatment of Thrombosis in          LMWH-induced HIT is thought to be less frequent when com-
Pediatric Congenital Heart Disease (1).                               pared with UFH (29). The frequency of osteoporosis from
                                                                      LMWH in pediatrics has not been studied, but in adults, LMWH
LMWH                                                                  is associated with less bone loss when compared with UFH (9).
MOA. LMWH are derived from UFH by chemical or enzy-                       Evidence to Support. Indications for LMWH therapy are
matic depolymerization and contain shorter length polysac-            the same as UFH and include the treatment of venous or arte-
charide chains. Similar to UFH, LMWH exert an anticoagulant           rial thrombi or prevention of thrombosis. Readers are referred
effect through binding antithrombin and potentiating the              to recent reviews that have been completed regarding the effec-
antithrombin anticoagulant activity, but when compared with           tiveness of LMWH (1, 4) .
UFH, there is a reduced inhibitory activity against factor IIa            Enoxaparin has been demonstrated to be effective in the
(thrombin) relative to factor Xa.                                     treatment of catheter-associated arterial thrombosis in pediat-
   Pharmacokinetics. LMWH are administered as a subcu-                ric patients with congenital heart disease (30, 31). In addition,
taneous injection. The pharmacokinetics properties are more           Enoxaparin has been associated with a reduction in the risk
stable than UFH. The half-life is 3–6 hours. Unlike UFH,              of thrombotic complications when compared with no therapy
LMWH is predominantly cleared by the kidneys. These favor-            in one study of pediatric patients with single ventricle cardiac
able pharmacokinetic features, when compared with UFH,                lesions after initial palliation (stage I) and after superior cavo-
have resulted in an increasing use of LMWH in children, and           pulmonary connection (stage II) (32).
Enoxaparin is currently the most common anticoagulant used
and studied in pediatric patients (22, 23).                           Fondaparinux
   Dosing. Pediatric patients exhibit a significant variation in      MOA. Fondaparinux is a synthetic analog of the antithrom-
LMWH dosing based on age and weight. Younger children and             bin-binding pentasaccharide found in heparin and LMWH.
neonates require higher doses of LMWH secondary to increased          The structure was modified to enhance its affinity for anti-
volume of distribution, increased clearance, and developmental        thrombin increasing the specific activity and half-life. Unlike
hemostasis (4, 24–26). Baseline laboratories should be obtained       UFH or LMWH, fondaparinux has no inhibitory activity
to ensure whether patient has a normal baseline coagulation           against factor IIa (thrombin) and only inactivates factor Xa.
state: complete blood count (CBC) (Thrombocytopenia is a                 Pharmacokinetics. Fondaparinux is administered via a
relative contraindication to anticoagulant therapy and should         subcutaneous injection. The half-life is longer than LMWH at
be corrected to ≥ 75,000 per mL before use), prothrombin time         17 hours. The clearance is exclusively renal.
(PT), aPTT, and serum creatinine (Enoxaparin is not recom-               Dosing. Pediatric: 0.1 mg/kg subcutaneous daily (33).
mended in the presence of renal failure).                                Monitoring. Routine coagulation monitoring is recom-
   Initial Enoxaparin Dose. Less than 3 months old: 1.7 mg/kg         mended for children. Specific assays calibrated for fondaparinux
subcutaneously every 12 hours.                                        should be used (34). Therapeutic goal is a fondaparinux anti-
   Three months–2 years old: 1.2 mg/kg subcutaneously every           Xa (peak): 0.5–1 mg/L (drawn 4 hr post administration).
12 hours.                                                                Reversal. There are no reversal agents available for
   More than 2 years old: 1 mg/kg subcutaneously every                fondaparinux. Unlike other forms of heparin, protamine does
12 hours.                                                             not bind fondaparinux. There are reports of using various
   Obese patients approximately 0.8 mg/kg subcutaneously              hemostatic therapies (prothrombin complex concentrates,
every 12 hours to maximum dose of 170 mg.                             activated prothrombin complex concentrates, or recombinant
Giglia et al

factor VIIa) to treat life-threatening bleeding secondary to         complex subunit 1 loci to create a nomogram for Coumadin
fondaparinux (35).                                                   dosing showed promise in reducing time to a therapeutic level,
   Consideration for Use. Similar to other anticoagulants, the       but further research is needed (42).
highest risk associated with fondaparinux therapy is hemor-              Monitoring. Coumadin monitoring relies on the INR; ther-
rhage. Due to the long half-life and lack of a reversal agent,       apeutic ranges vary with the indication. Limited studies exam-
this medication should only be used in patients who are clini-       ining patient self-testing have improved quality of life and time
cally stable with a low risk of hemorrhage. It should not be         in the therapeutic range (43, 44).
used in patients with renal insufficiency. Off-label use has been        Reversal. Reversal of Coumadin and related compounds is
reported in both adults and pediatric patients in the treatment      related to the urgency in which reversal is desired, the throm-
of HIT although further studies are warranted (36, 37).              botic risk associated with treatment and the severity of bleed-
   Evidence to Support. There is limited evidence in pediatrics      ing (45–47). Choices for reversal include vitamin K, fresh
regarding the use of fondaparinux. The strongest data include one    frozen plasma, and discontinuing Coumadin. In general, vita-
prospective dose-finding, single arm, pharmacokinetics study in      min K is avoided when the risk of thromboembolism is high,
24 pediatric patients and a long-term follow-up study of 34 pedi-    such as left ventricular assist devices (VADs) and mechanical
atric patients who received fondaparinux (33, 36). These limited     prosthetic heart valves; however, recently, a population of adult
data suggest that fondaparinux may be safe and efficacious.          left VAD patients overall tolerated reversal well (48).
                                                                         Consideration for Use. Coumadin has traditionally been
Warfarin                                                             avoided in pregnancy because of risk of fetal loss and birth
MOA. Coagulation factors II, V, VII, X, and proteins C, S, and Z     defects although by targeting a lower therapeutic range, these
undergo the vitamin K–dependent process of γ-carboxylation.          risks may be mitigated (49). After the Fontan procedure, the
The group of 4-hydroxycoumarin–related molecules, where              use of aspirin versus warfarin for thromboembolic prophylaxis
Coumadin is included, exerts their action by inhibiting vita-        remains controversial. Indications and INR ranges for warfa-
min K epoxide reductase preventing vitamin K1 regeneration           rin that are encountered in the cardiac ICU are discussed in
after γ-carboxylation.                                               more detail in the recommendations of the American College
    Pharmacokinetics. Coumadin is a racemic mixture of the           of Chest Physicians (4, 50) as well as in the 2013 American
R- and S-enantiomers. The S-enantiomer is more potent, but           Heart Association guidelines for the Prevention and Treat-
is more rapidly cleared. The half-life of racemic warfarin is        ment of Thrombosis in Pediatric Congenital Heart Disease (1).
36–42 hours. Significant variability in dose-response is related     Recommendations for the prophylaxis of mechanical valves
to genetic variation in vitamin K epoxide reductase com-             (51), arrhythmias (1), and cardiac systolic dysfunction (1) are
plex subunit 1, which is responsible for reducing vitamin K          extrapolated from the adult literature. Recommendations for
2,3-epoxide to active vitamin K1 (38–40).                            prophylaxis of VADs are specific to each particular device and
    Dosing. The interaction of age, varying targeted ranges,         are provided by the manufacturers.
genetic polymorphism of enzymatic breakdown, and active
site of vitamin K epoxide reductase makes pediatric Coumadin         Parental DTIs
dosing difficult. Initial bolus dosing of 0.2 mg/kg (maximum         MOA. DTI are direct, specific, and reversible inhibitors of
initial dose 10 mg) with adjustments on subsequent days based        thrombin. The three commercially available DTIs are bivali-
on daily INR values has been applied in two trials where all sub-    rudin, argatroban, and lepirudin. Argatroban is a univalent
jects attained an INR greater than 2 and most reached a target       DTI, which reversibly inhibits thrombin’s catalytic site. Bivali-
INR in less than 7 days (4, 41). See the 2012 Chest guidelines for   rudin and lepirudin are known as bivalent DTIs, which bind
suggested subsequent day dosing based on daily INR (4).              the active catalytic site of thrombin, as well as the thrombin-
    Patients who have had a Fontan procedure or have liver           fibrinogen binding site.
disease are generally more sensitive to warfarin, and so a con-          Pharmacokinetics. Steady state levels of argatroban are
servative loading dose of 0.1 mg/kg is recommended. Patients         attained after 1–3 hours. The elimination half-life is 39–51
under the age of 1 year often require much higher doses of           minutes and is principally excreted by the biliary route
warfarin. Guidance of a hematologist may be helpful.                 (52, 53). After discontinuation of the drug, aPTT normalized
    Initiation of warfarin without a bolus dose: an alternative      in less than 4 hours, but may take up to 20 hours in the context
regime approximates the dose of warfarin based on age and            of severe liver dysfunction. Bivalirudin is an engineered syn-
weight without the use of a bolus: age 2–12 years old: 0.09 mg/kg    thetic hirudin with a half-life is 25 minutes (54). It is cleaved
day, age more than 12 years old: 0.08 mg/kg day.                     by thrombin and subsequently excreted by the liver with
    Continue heparin and warfarin for 5 days. Check INR on           20% excreted by the kidney (55). Lepirudin is a recombinant
day 5. If the INR is therapeutic, stop the heparin (UFH or           hirudin with a half-life of 60 minutes, which is cleared by the
LMWH) and check INR in 3–5 days. If day 5 INR is subthera-           kidneys. Steady state levels are attained after 1–3 hours. Anti-
peutic, increase warfarin based on the table above and con-          bodies to the drug can form in as many as 40% of patients (56)
tinue heparin (UFH or LMWH) until INR is therapeutic.                although anaphylaxis is rare (57).
    Recently, a pilot study using the genotype of the cytochrome         Dosing. The recommendations of DTI dosing are extrapo-
p450 enzyme (CYP2C9) and vitamin K epoxide reductase                 lated from adult experience/protocols. Recent survey results

S80            www.pccmjournal.org                                                           March 2016 • Volume 17 • Number 3 (Suppl.)
Supplement

provide insight to commonly used pediatric dosing. Argatro-         (76). The disadvantage is that methods to monitor these medi-
ban is not commonly bolused and the initial infusion rate is        cations are not readily available (76). It is unclear at this time
typically 0.25–1 μg/kg/min. In advanced liver disease, arg-         whether monitoring will be required for pediatric patients.
atroban dosing must be reduced (58). Bivalirudin is bolused             Reversal. There are no reversal agents for NOACs (77).
at 0.125 mg/kg IV and then an infusion of 0.125 mg/kg/hr is         Dabigitran can be partially removed with hemodialysis (78).
initiated. According to national pediatric surveys and reviews      There are reports of using various hemostatic therapies (pro-
of administrative databases, lepirudin has not been reported        thrombin complex concentrates, activated prothrombin
or used frequently enough to have guidelines that are different     complex concentrates, or recombinant factor VIIa) to treat
from adult guidelines (59, 60).                                     life-threatening bleeding secondary to NOACs (77, 79). Spe-
    Monitoring. Most institutions report titrating the DTIs         cific antidotes are currently being developed (79).
based on aPTTs every 2–4 hours to aim for an aPTT 1.5–2.5×              Consideration for Use. At this time, NOACs should only be
normal (60). Issues with aPTT in monitoring DTIs are as fol-        used in pediatric patients in the setting of a clinical trial.
lows: aPTT does not reliably detect supratherapeutic DTI lev-           Evidence to Support. In adult studies, NOACs have been
els as the dose response curves are not linear at higher ranges;    demonstrated to be efficacious and in the United States are
there can be variation among aPTT reagents; and there can           approved for the use in stroke prevention from atrial fibrilla-
be unreliability in the context of antiphospholipid antibodies      tion, the treatment of acute deep vein thrombosis (DVT), sec-
(61–63). The diluted thrombin time assays and ecarin clotting       ondary prevention of DVT, DVT prevention post elective knee
assays are the most accurate and reproducible tests for moni-       and hip surgery (80), and in the treatment of acute pulmonary
toring DTIs (62, 64–66).                                            embolus (81, 82).
    Reversal. Limited DTI clinical application has led to lim-          At this time, NOACs should not be used in the setting of
ited knowledge of optimal means of reversal. Isolated products      mechanical heart valves. A clinical trial comparing dabigi-
(fresh frozen plasma and cryoprecipitate) or factors (activated     tran with warfarin in patients with either an aortic or mitral
factor VII) have been shown to be of inconsistent benefit in        mechanical heart valve was halted early secondary to increased
DTI-associated bleeding (66–69). The definitive reversal ther-      thrombotic and bleeding events in those patients receiving
apy is renal replacement therapy (70–72). Adult protocols for       dabigitran when compared with warfarin (83). A phase 2 clini-
reversal and management of DTI-associated hemorrhage are            cal trial is currently underway using rivaroxaban in the set-
reported elsewhere (73).                                            ting of mechanical aortic valve replacement (ClincalTrials.gov
    Consideration for Use. The only labeled indication for DTIs     Identifier: NCT02128841).
is for HIT and for coronary intervention in adults. DTIs are
increasingly used for HIT, and trials are underway examining
                                                                    Antiplatelet Agents
their role in ischemic stroke and renal replacement therapy
(74, 75).                                                           Aspirin Acetylsalicylic Acid
    Evidence to Support. At present, there are no large trials of   MOA. It irreversibly inhibits cyclooxygenase (COX-1 and
use of DTIs in pediatrics and no Food and Drug Administra-          COX-2) activity in the arachidonic acid pathway, inhibiting the
tion–approved uses; however, the use in HIT is accepted, and        formation of thromboxane (TXA2), inhibiting platelet activa-
the pediatric experience of its use continues to grow.              tion and aggregation (84).
                                                                        Pharmacokinetics. Upon oral administration, aspirin ace-
Novel Oral Anticoagulants                                           tylsalicylic acid (ASA) is rapidly absorbed after administration.
Since 2010, novel oral anticoagulants (NOACs) received Food         It is hydrolyzed to salicylate (active) by esterases in the fastrin-
and Drug Administration approval for use in adult patients.         testinal mucosa, RBC, synovial fluid, and readily distributes
Pediatric trials are currently ongoing, and we await these          into most body fluids and tissues. ASA is hepatically metabo-
results to direct the use of these medications in children.         lized and renally excreted. Although dose dependent, the half-
   MOA. All of the NOACs bind directly to a key coagulant           life (t½) is 15–20 minutes (84).
protein to inhibit fibrin formation. There are two broad cat-           Dosing for Antiplatelet Effect. Antiplatelet: 1–5 mg/kg/d
egories for MOA including direct anti-Xa inhibitors (rivar-         (maximum: 91 mg) (4, 85).
oxaban, apixaban, and edoxaban) and direct thrombin (IIa)               Monitoring. There are no studies that link outcome to
inhibitors (dabigitran).                                            measuring aspirin effect; however, platelet aggregation assays
   Pharmacokinetics. The advantage to all of the NOACs is           (i.e., light transmission aggregometry) may be used to inter-
more predictable pharmacokinetics with a wide therapeutic           pret arachidonic acid (goal < 40% of normal activity for aspi-
window and limited drug-drug interactions when compared             rin) and adenosine diphosphate (ADP) activity to monitor
with warfarin. They also have a relative rapid onset of action      theoretical response. The thromboelastogram with platelet
and shorter half-lives than warfarin.                               mapping assay is a modification of the standard thromboelas-
   Dosing. Pediatric dosing is not available at this time.          togram and is used to assess the effectiveness of antiplatelet
   Monitoring. The key advantage to predictable pharmacoki-         medications. This assay uses whole blood that is heparinized to
netics and a wide therapeutic window for NOACs is that rou-         suppress thrombin generation, and then known platelet ago-
tine therapeutic monitoring is not necessary in adult patients      nists are added to the sample to assess platelet activation. The

Pediatric Critical Care Medicine                                                                    www.pccmjournal.org            S81
Giglia et al

thromboelastogram with platelet mapping is commonly used                Reversal.
in pediatric patients with a left VAD to monitor antiplatelet           ●●   No reversal agents available.
therapy (86).                                                           ●●   Platelet aggregation has been shown to fully recover 4 days
Reversal.                                                                    after discontinuation of clopidogrel; however, it is com-
●●   No reversal agents available.                                           mon to discontinue therapy 5–7 days prior to surgery.
●●   Platelet aggregation may recover within 4 days after dis-          ●●   Type and cross pheresis.
     continuation of aspirin, 11 however due to the irreversible        ●●   Platelets 4–20 U/kg.
     platelet inhibition it is commonly recommended to dis-
                                                                           Evidence to Support. Clopidogrel is used instead of ASA in
     continue ASA 7 days before the surgery to allow full plate-
                                                                        the presence of ASA allergy or intolerance. It has also been used
     let regeneration.
                                                                        in combination with ASA in selected patients when additional
●●   Type and cross pheresis.
                                                                        antiplatelet effect is desired, for example, children with Kawasaki
●●   Platelets (4–20 U/kg) may be given to counteract the plate-
                                                                        disease and moderate sized aneurysms (1, 4, 89). Adding clopi-
     let aggregation inhibition from ASA.
                                                                        dogrel to chronic low-dose, ASA did not reduce overall mortality
    Evidence to Support. Low-dose aspirin has become the main-          or shunt-related morbidity in the Clopridigrel to Lower Arterial
stay of thromboprophylaxis for Blalock-Taussig shunts (1, 4, 87,        Thrombotic Risk in Neonates and Infants Trial (101)
88) and in the long-term management of coronary aneurysms in
Kawasaki Disease (1, 4, 89). Aspirin may be recommended after           Dipyridamole
stroke in children with heart disease depending on the age of the       MOA. It inhibits the activity of adenosine deaminase and
child, the etiology of the stroke (ischemic vs embolic), and the        phosphodiesterase, which causes an accumulation of adenos-
time after the stroke and the risk of recurrence (1, 4, 90). Aspirin    ine, adenine nucleotides, and cyclic AMP which inhibit platelet
is often used as a part of the thromboprophylaxis regime with           aggregation. Vasodilation, including coronary artery vasodila-
VADs although specifics vary with device. Controversy exists in         tion, has been reported (1, 102–104).
the use of aspirin in single hypoplastic left heart disease palliated      Pharmacokinetics. Although dipyridamole is available in IV
with a stage I with Sano modification (1, 4, 91), single ventricle      form, the oral formulation is what is primarily used in clinical
physiology beyond stage I (1, 4, 32, 92–94), and with intravas-         practice. Oral dipyridamole is absorbed slowly and with much
cular stents (1, 4). Aspirin has been recommended as an adjunct         variability (26–66%) (96, 105). Peak serum concentrations are
to warfarin in the thromboprophylaxis of mechanical valves in           reached around 75 minutes, and the t½ is 10–12 hours on aver-
the mitral and aortic positions although the practice in children       age (106, 107). Dipyridamole is hepatically metabolized and
varies (51, 95).                                                        excreted in bile as glucuronide conjugates and unchanged drug
                                                                        and eliminated in feces (103, 106, 107).
Clopidogrel                                                                Dosing. 1–5 mg/kg/d (1, 4).
MOA. It undergoes in vivo biotransformation to an active thiol             Monitoring. Although there are no studies that link out-
metabolite, which irreversibly blocks the P2Y12 component of            come to measuring dipyridamole effect, platelet aggregation
ADP receptors on the platelet surface, which prevents platelet          panels that assess arachidonic acid and ADP activity (goal
activation and therefore aggregation (96).                              20–40% of normal activity), as well as platelet mapping tech-
    Pharmacokinetics. Clopidogrel is only available in oral             niques, such as thromboelastogram monitoring (MA, α), are
formulations and is rapidly absorbed after administration. It           often used (86).
undergoes extensive hepatic metabolism via esterase-hydro-              Reversal.
lysis to an inactive carboxylic acid derivative and CYP2C19-            ●●   No reversal agents available.
mediated oxidation to an active thiol metabolite. The t½ of the         ●●   Type and cross pheresis.
parent drug is around 6 hours, however, that of the active thiol        ●●   Platelets 10–20 U/kg.
metabolite is approximately 30 minutes (96).
                                                                            Evidence to Support. Currently, the main use of dipyri-
    Dosing. Infants and children less than or equal to 24 months:
                                                                        damole is as adjunct therapy to other anticoagulant/antiplate-
initial dose of 0.2 mg/kg/dose once daily (97–99)
                                                                        let medications in the maintenance of mechanical circulatory
    Children 2 years old or older: initial dose of 1 mg/kg once
                                                                        support (108–112). Dipyridamole, in a dose-related fashion
daily; titrate to response (97–99)
                                                                        when combined with aspirin, has been shown to decrease both
    Monitoring. Although there are no studies in pediatric
                                                                        platelet aggregation and platelet adhesiveness more than aspirin
patients that link outcome to measuring clopidogrel effect, the
                                                                        alone (113).
VerifyNow platelet aggregation assay that measures percentage
of P2Y12 receptor inhibition is often used in clinical practice
(100); a level of less than 236 platelet activity units is generally    THROMBOLYTIC THERAPY
accepted as a therapeutic response based on positive outcomes           Unlike the anticoagulants and antiplatelet agents that prevent
in adults patients undergoing elective coronary stent implan-           thrombus formation and/or thrombus extension, the throm-
tation (100). thromboelastogram monitoring (MA, α) is also              bolytic agents augment the body’s natural systems of dissolv-
commonly used to monitor response to the drug (86).                     ing blood clots. The only available thrombolytic agents in the

S82            www.pccmjournal.org                                                               March 2016 • Volume 17 • Number 3 (Suppl.)
Supplement

United States currently are the biosynthetic (recombinant DNA               The decreased level of plasminogen in neonates (≈50% of
origin) forms of the enzyme human tissue-type plasminogen               that in adults) slows the generation of plasmin and reduces
activator (tPA): alteplase (TPA), recteplase, and tenecteplase.         the effect of thrombolytics in an in vitro system (120).
Urokinase, obtained from human neonatal kidney cells grown              Supplementation with plasminogen increases the thrombo-
in tissue culture and streptokinase, obtained from bacterial            lytic effect and may be helpful (4).
cultures, are no longer available in the United States. Whenever
possible, a hematology consultation is recommended to assist            Dosing
in evaluation of eligibility, administration, and monitoring.           Numerous dosing strategies for alteplase (tPA) have been used
                                                                        in pediatrics, and there is currently no consensus as to which
Safety and Efficacy                                                     approach is optimal. Higher doses may restore flow more rap-
Before initiation of thrombolytic therapy, hematologic                  idly, but appear to have a higher risk of bleeding. A 2009 survey
derangements, such as thrombocytopenia or vitamin K defi-               of hematology-oncology specialists showed no consensus in
ciency, should be corrected. The risk of bleeding with throm-           indications for thrombolysis, dose, mode of delivery, or length
bolytic therapy in children is significant mandating serious            of therapy (121).
consideration of the risk and benefits in each situation prior             Alteplase may be delivered systemically or locally by cath-
to initiation of therapy and consultation with a bleeding and           eter-directed therapy with the following dosing regimens (1):
clotting expert whenever possible. Older studies reported a 40–
                                                                        ●●   Systemic thrombolysis:
60% prevalence of bleeding complications when higher doses
of alteplase were used (> 0.5 mg/kg/hr). A report of alteplase               ○○   Low-dose alteplase (tPA) regimen: this regimen was
for femoral artery thrombosis in children documented a 1.5%                       initially recommended for patients with non–life-
prevalence of intracranial hemorrhage with the highest preva-                     threatening venous thrombotic events although it has
lence in preterm infants when compared with older children                        been successfully used in patient with prosthetic valve
(114). A more recent review reported major and minor bleed-                       thrombosis and more critical arterial thrombi.
ing complications with TPA as 17% and 26%, respectively                           ◾◾ Initial dose: 0.03–0.06 mg/kg/hr. Maximum dose for
(115). In the event of minor bleeding, local pressure and sup-                       low-dose alteplase (tPA) 2 mg/hr.
portive care are usually sufficient. Major bleeding is treated by                 ◾◾ Duration: this dose may be continued for a relatively
stopping the thrombolytic and administering cryoprecipitate                          prolonged duration (24–86 hr) and may be increased
(usual dose of 1 U/5 kg or 5–10 mL/kg), an antifibrinolytic, or                      within the 0.03–0.06 range providing that hema-
both along with other blood products as needed (4).                                  tologic parameters are appropriate and there is no
    Complete resolution of thrombosis with TPA has been                              hemodynamically significant bleeding.
reported to range from 39% to 87% with no response to ther-                       ◾◾ Ongoing monitoring of hematologic parameters (see
apy in about 20% (4) and resolution in arterial thrombi supe-                        monitoring below) and thrombus assessment are
rior to venous (116).                                                                essential.
    MOA. It initiates local fibrinolysis by binding to fibrin in a           ○○   High-dose alteplase (tPA) regimen: This regimen may
thrombus and converting entrapped plasminogen to plasmin                          be considered for patients with arterial or more critical
that in turn degrades fibrinogen (Lexicomp Online, Pediatric &                    thrombotic events.
Neonatal Lexi-Drugs, Lexicomp, Hudson, OH; April 7, 2015).                        ◾◾ Initial dose: 0.1–0.6 mg/kg/hr.
There is an evidence that the tPAs, especially alteplase, have                    ◾◾ Duration: the duration will depend upon the chosen
antiplatelet properties as well by inhibiting platelet aggregation.                  dose. For patients receiving doses of 0.5–0.6 mg/kg/hr,
Alteplase-related inhibition of platelet aggregation appears to be                   hematologic parameters and thrombus assessment
independent of plasmin generation, fibrinogen degradation and                        should be performed (see monitoring below) at 6 hours.
glycoprotein IIb/IIIa receptor, and P-selectin expression (117).                     If the progress is not adequate, hematologic parameters
    Pharmacokinetics. Pharmacokinetics have not been estab-                          are within acceptable range (see contraindications for
lished in infants and children. Pharmacokinetics in adults has                       thrombolysis) and the patient is stable, a second 6-hour
been described as followed:                                                          infusion can be administered. Patients at the lower end
●●   Duration: more than 50% present in plasma cleared approx-                       of this dose range may tolerate longer infusions.
     imately 5 minutes after infusion terminated, approximately              ○○   Pulmonary embolism protocol: thrombolysis may
     80% cleared within 10 minutes; fibrinolytic activity per-                    be considered for patients with massive pulmonary
     sists for up to 1 hour after infusion terminated (Lexicomp                   embolus (PE) who are hemodynamically unstable.
     Online, Pediatric & Neonatal Lexi-Drugs) (118).                              Patients who are hemodynamically stable with PE are
●●   Excretion: rapidly cleared from circulating plasma                           treated with anticoagulation. The following protocol
     (572 ± 132 mL/min) (119), primarily hepatic; more than 50%                   has been used in adults and would need to be individu-
     present in plasma is cleared within 5 minutes after the infusion             alized for younger patients.
     is terminated, approximately 80% cleared within 10 minutes                   ◾◾ Adult dose/duration: 10-mg alteplase over 10 minutes,
     (Lexicomp Online, Pediatric & Neonatal Lexi-Drugs) (118).                       followed by 90 mg over 2 hours.

                                                                                                           www.pccmjournal.org           S83

     Pediatric Critical Care Medicine
Giglia et al

     ○○   Stroke protocol: thrombolysis for stroke has not been        mL before initiation of therapy. Patients are often admitted to
          studied in pediatric patients and should be performed        an ICU for close monitoring during tPA therapy and must be
          under the guidance of a Stroke Team whenever pos-            on strict bed rest. A blood drawing IV or arterial line is rec-
          sible. Thrombolysis is considered when treatment can         ommended for frequent laboratory draws prior to initiation
          be initiated less than 4.5 hours after onset of symptoms     of therapy. Heparin flushes and heparin-containing solution
          and where imaging demonstrates thrombotic occlusion.         should not be used for this line.
          Onset of symptoms is defined as the time the patient
                                                                       ●●   Check PT, PTT, CBC, fibrinogen, D-dimer prior to start-
          was last seen well, or at usual clinical baseline.
                                                                            ing, and then every 4–6 hours while the patient is receiving
          ◾◾ Dose/duration: 0.9 mg/kg alteplase (maximum of                 the thrombolytic. An elevated D-dimer and drop in the
             90 mg) infused over 60 minutes with 10% of the total           fibrinogen is indicative of a lytic state.
             dose administered as an initial IV bolus over             ●●   Monitor neurologic status q1 hour for possible ICH. In
             1 minute.                                                      infants or critically ill patients, whose neurologic status is
●●   Catheter-directed thrombolysis: usually administered via a             difficult to assess, consider a daily head ultrasound or CT
     pulse-spray catheter located in the thrombus                           during therapy.
                                                                       ●●   Dose adjustment: if the fibrinogen drops to less than
     ○○   Dose/duration: 0.5–2 mg/hr. The duration will vary
                                                                            125 mg/dL, decrease the dose of thrombolytic by 50%.
          depending on the dose and indication, but long infu-
                                                                            If the fibrinogen drops greater than 100 mg/dL, consider
          sions (96 hr) have been well tolerated. Ongoing moni-
                                                                            holding thrombolytic or give cryoprecipitate.
          toring of hematologic parameters (see monitoring
                                                                       ●●   Avoid invasive procedures (blood draws, placement of
          below) and thrombus assessment are essential.
                                                                            arterial or venous catheters, intramuscular injections,
●●   Pharmacomechanical thrombolysis: it refers to the use of               nasogastric tubes, rectal temperature probes, and place-
     alteplase in conjunction with one of several devices for               ment of urinary catheters) during therapy and for at least
     mechanical disruption of clot. Specific devices include:               24 hours after high-dose tPA therapy.
     ○○   Angiojet: saline is injected under high pressure into        ●●   Avoid drugs that affect platelet function (e.g., aspirin,
          the thrombus, which helps to break up the thrombus.               nonsteroidal anti-inflammatory, dipyridamole) as they
          Simultaneously, the saline and thrombus are suctioned             may potentiate the risk of hemorrhage.
          back into the catheter. In patients without contraindi-      ●●   If a patient has significant bleeding, stop the thrombolytic
          cations to thrombolysis, alteplase may be added to the            agent and heparin, administer cryoprecipitate, and con-
          saline infusion. For an adolescent patient (> 50 kg),\mg          sider reversing heparin with protamine in life-threatening
          of alteplase is a typical dose. Although some of the              situations. Antifibrinolytics (aminocaproic acid) can also
          alteplase will be suctioned back, much will be systemic.          be considered.
     ○○   Trellis device: this device is placed into the thrombus          Reversal. Alteplase (tPA) cannot be reversed, but has a rel-
          and balloons inflated above and below. Alteplase is          atively short half-life. Holding alteplase (tPA) and heparin a
          instilled between the balloons, while a small wire rotates   minimum of 6 hours prior to surgical procedure or lumbar
          to disrupt the clot. The standard dose is 10 mg of tPA       puncture is recommended.
          for patients more than 50 kg, administered at a rate of          Consideration for Use. Thrombolytics have a low margin
          1 mg/min. After 10 minutes, the liquefied thrombus and       of safely and variable efficacy in children and therefore should
          remaining alteplase are aspirated back through the cath-
                                                                       be used with caution. Although anticoagulation alone is often
          eter so that the alteplase is not systemic.
                                                                       effective at managing thromboembolism, there are times when
    Use of Concomitant Anticoagulation. The use of UFH dur-            more rapid clot resolution is necessary or desirable. Indications
ing systemic and catheter-directed thrombolytic therapy may            for thrombolysis in the treatment of pediatric thromboembo-
be helpful in preventing ongoing thrombus formation but will           lism are not well established, primarily due to the paucity of
increase the risk of bleeding. This decision should be made on an      well-designed clinical studies. The benefit of rapid clot resolu-
individual basis. Usually low-dose heparin is used at 10 U/kg/hr.      tion must be weighed against the risk of major bleeding, which is
In cases where alteplase is used to treat acute ischemic stroke,       greater than with anticoagulation alone. Thrombolytic therapy
other antithrombotic treatments, such as heparin, warfarin,            in children should be restricted to situations in which the benefit
aspirin, are held for at least 24 hours.                               of rapid thrombus resolution is thought to outweigh the risk of
    Monitoring. Because of the risk of bleeding with tPA the           major hemorrhage and is usually reserved for thrombosis which
following laboratories are recommended prior to initiation of          is hemodynamically significant and/or limb threatening.
therapy: CBC, PT, PTT, fibrinogen, D-dimer, and to exclude                 Evidence to Support. Thrombolytic therapy has been used
intracranial hemorrhage, a head ultrasound in neonates and             in infants and children since this early 1980s; initially uroki-
infants younger than 1 month old and head CT in older chil-            nase and streptokinase (122–125) and then subsequently tPA
dren whose neurologic status is difficult to assess. Thrombocy-        were used (126–129). Although there have been no random-
topenia is a relative contraindication to thrombolytic therapy         ized trials, a substantial recent experience has been reported in
and should be corrected to greater than or equal to 75,000 per         children with heart disease in the following areas:

S84             www.pccmjournal.org                                                              March 2016 • Volume 17 • Number 3 (Suppl.)
Supplement

●●   General (1, 4, 130).                                                      20. Spadone D, Clark F, James E, et al: Heparin-induced thrombocytope-
                                                                                   nia in the newborn. J Vasc Surg 1992; 15:306–311
●●   Postcatheterization (1, 4, 130).
                                                                               21. Risch L, Huber AR, Schmugge M: Diagnosis and treatment of hepa-
●●   DVT (1, 4).                                                                   rin-induced thrombocytopenia in neonates and children. Thromb Res
●●   Mechanical prosthetic valves (95, 131–133).                                   2006; 118:123–135
●●   Intracardiac (130, 134–138).                                              22. Raffini L, Huang YS, Witmer C, et al: Dramatic increase in venous
●●   In combination with AngioJet/angioplasty (139).                               thromboembolism in children’s hospitals in the United States from
                                                                                   2001 to 2007. Pediatrics 2009; 124:1001–1008
●●   Occluded Blalock-Taussig Shunt (95) in combination with                   23. Dix D, Andrew M, Marzinotto V, et al: The use of low molecular weight
     balloon angioplasty/stent (140).                                              heparin in pediatric patients: A prospective cohort study. J Pediatr
●●   Stroke (90, 141, 142).                                                        2000; 136:439–445
                                                                               24. Michaels LA, Gurian M, Hegyi T, et al: Low molecular weight heparin
                                                                                   in the treatment of venous and arterial thromboses in the premature
                                                                                   infant. Pediatrics 2004; 114:703–707
REFERENCES                                                                     25. Ho SH, Wu JK, Hamilton DP, et al: An assessment of published
 1. Giglia TM, Massicotte MP, Tweddell JS, et al; American Heart                   pediatric dosage guidelines for enoxaparin: A retrospective review.
    Association Congenital Heart Defects Committee of the Council on               J Pediatr Hematol Oncol 2004; 26:561–566
    Cardiovascular Disease in the Young, Council on Cardiovascular and
                                                                               26. Bauman ME, Belletrutti MJ, Bajzar L, et al: Evaluation of enoxaparin
    Stroke Nursing, Council on Epidemiology and Prevention, and Stroke
                                                                                   dosing requirements in infants and children. Better dosing to achieve
    Council: Prevention and treatment of thrombosis in pediatric and con-
                                                                                   therapeutic levels. Thromb Haemost 2009; 101:86–92
    genital heart disease: A scientific statement from the American Heart
    Association. Circulation 2013; 128:2622–2703                               27. Dager WE, White RH: Low-molecular-weight heparin-induced throm-
                                                                                   bocytopenia in a child. Ann Pharmacother 2004; 38:247–250
 2. McCrindle BW, Li JS, Manlhiot C, et al: Challenges and priorities for
    research: A report from the National Heart, Lung, and Blood Institute      28. Klenner AF, Lubenow N, Raschke R, et al: Heparin-induced throm-
    (NHLBI)/National Institutes of Health (NIH) Working Group on throm-            bocytopenia in children: 12 new cases and review of the literature.
    bosis in pediatric cardiology and congenital heart disease. Circulation        Thromb Haemost 2004; 91:719–724
    2014; 130:1192–1203                                                        29. Warkentin TE, Levine MN, Hirsh J, et al: Heparin-induced thrombo-
 3. Monagle P: Thrombosis: Congenital heart disease and thrombosis:                cytopenia in patients treated with low-molecular-weight heparin or
    What do we know? Nat Rev Cardiol 2014; 11:132–134                              unfractionated heparin. N Engl J Med 1995; 332:1330–1335
 4. Monagle P, Chan AK, Goldenberg NA, et al: Antithrombotic therapy in neo-   30. Bontadelli J, Moeller A, Schmugge M, et al: Enoxaparin therapy for
    nates and children: Antithrombotic Therapy and Prevention of Thrombosis,       arterial thrombosis in infants with congenital heart disease. Intensive
    9th ed: American College of Chest Physicians Evidence-Based Clinical           Care Med 2007; 33:1978–1984
    Practice Guidelines. Chest 2012; 141(Suppl 2):e737S–801S.                  31. Glatz AC, Keashen R, Chang J, et al: Outcomes using a clinical
 5. Linhardt RJ, Gunay NS: Production and chemical processing of low               practice pathway for the management of pulse loss following pedi-
    molecular weight heparins. Semin Thromb Hemost 1999; 25(Suppl                  atric cardiac catheterization. Catheter Cardiovasc Interv 2015;
    3):5–16                                                                        85:111–117
 6. Damus PS, Hicks M, Rosenberg RD: Anticoagulant action of heparin.          32. Manlhiot C, Brandão LR, Kwok J, et al: Thrombotic complications and
    Nature 1973; 246:355–357                                                       thromboprophylaxis across all three stages of single ventricle heart
                                                                                   palliation. J Pediatr 2012; 161:513–519.e3
 7. Rosenberg RD, Lam L: Correlation between structure and function of
    heparin. Proc Natl Acad Sci U S A 1979; 76:1218–1222                       33. Young G, Yee DL, O’Brien SH, et al: FondaKIDS: A prospec-
                                                                                   tive pharmacokinetic and safety study of fondaparinux in children
 8. Lindahl U, Bäckström G, Höök M, et al: Structure of the anti-
                                                                                   between 1 and 18 years of age. Pediatr Blood Cancer 2011;
    thrombin-binding site in heparin. Proc Natl Acad Sci U S A 1979;
                                                                                   57:1049–1054
    76:3198–3202
                                                                               34. Paolucci F, Frasa H, Van Aarle F, et al: Two sensitive and rapid chro-
 9. Garcia DA, Baglin TP, Weitz JI, et al; American College of Chest
                                                                                   mogenic assays of fondaparinux sodium (Arixtra) in human plasma
    Physicians: Parenteral anticoagulants: Antithrombotic Therapy
                                                                                   and other biological matrices. Clin Lab 2003; 49:451–460
    and Prevention of Thrombosis, 9th ed: American College of Chest
    Physicians Evidence-Based Clinical Practice Guidelines. Chest              35. Elmer J, Wittels KA: Emergency reversal of pentasaccharide antico-
    2012; 141(Suppl 2):e24S–e43S                                                   agulants: A systematic review of the literature. Transfus Med 2012;
                                                                                   22:108–115
10. Estes JW: The kinetics of heparin. Ann N Y Acad Sci 1971;
    179:187–204                                                                36. Schindewolf M, Steindl J, Beyer-Westendorf J, et al: Frequent off-label
                                                                                   use of fondaparinux in patients with suspected acute heparin-induced
11. Estes JW: Clinical pharmacokinetics of heparin. Clin Pharmacokinet
                                                                                   thrombocytopenia (HIT)–findings from the GerHIT multi-centre regis-
    1980; 5:204–220
                                                                                   try study. Thromb Res 2014; 134:29–35
12. Newall F, Johnston L, Ignjatovic V, et al: Unfractionated heparin ther-
                                                                               37. Tokgoz H, Caliskan U, Demir M: Successful use of fondaparinux
    apy in infants and children. Pediatrics 2009; 123:e510–e518
                                                                                   in a child with heparin-induced thrombocytopenia. Blood Coagul
13. McDonald MM, Jacobson LJ, Hay WW Jr, et al: Heparin clearance in               Fibrinolysis 2012; 23:769–771
    the newborn. Pediatr Res 1981; 15:1015–1018
                                                                               38. Nguyen N, Anley P, Yu MY, et al: Genetic and clinical determinants
14. Spiess BD: Treating heparin resistance with antithrombin or fresh fro-         influencing warfarin dosing in children with heart disease. Pediatr
    zen plasma. Ann Thorac Surg 2008; 85:2153–2160                                 Cardiol 2013; 34:984–990
15. Murphy MS, John PR, Mayer AD, et al: Heparin therapy and bone              39. Vear SI, Ayers GD, Van Driest SL, et al: The impact of age and
    fractures. Lancet 1992; 340:1098                                               CYP2C9 and VKORC1 variants on stable warfarin dose in the paedi-
16. Sackler JP, Liu L: Heparin-induced osteoporosis. Br J Radiol 1973;             atric population. Br J Haematol 2014; 165:832–835
    46:548–550                                                                 40. Shaw K, Amstutz U, Hildebrand C, et al: VKORC1 and CYP2C9
17. Klenner AF, Fusch C, Rakow A, et al: Benefit and risk of heparin for           genotypes are predictors of warfarin-related outcomes in children.
    maintaining peripheral venous catheters in neonates: A placebo-con-            Pediatr Blood Cancer 2014; 61:1055–1062
    trolled trial. J Pediatr 2003; 143:741–745                                 41. Andrew M, Marzinotto V, Brooker LA, et al: Oral anticoagulation
18. Newall F, Barnes C, Ignjatovic V, et al: Heparin-induced thrombocyto-          therapy in pediatric patients: A prospective study. Thromb Haemost
    penia in children. J Paediatr Child Health 2003; 39:289–292                    1994; 71:265–269
19. Schmugge M, Risch L, Huber AR, et al: Heparin-induced thrombo-             42. Lala M, Burckart GJ, Takao CM, et al: Genetics-based pediatric warfa-
    cytopenia-associated thrombosis in pediatric intensive care patients.          rin dosage regimen derived using pharmacometric bridging. J Pediatr
    Pediatrics 2002; 109:E10                                                       Pharmacol Ther 2013; 18:209–219

Pediatric Critical Care Medicine                                                                                  www.pccmjournal.org                S85
Giglia et al

43. Bajolle F, Lasne D, Elie C, et al: Home point-of-care international nor-   65. Curvers J, van de Kerkhof D, Stroobants AK, et al: Measuring direct
    malised ratio monitoring sustained by a non-selective educational pro-         thrombin inhibitors with routine and dedicated coagulation assays:
    gram in children. Thromb Haemost 2012; 108:710–718                             Which assay is helpful? Am J Clin Pathol 2012; 138:551–558
44. Bauman ME, Black K, Bauman ML, et al: EMPoWarMENT: Edmonton                66. Lind SE, Boyle ME, Fisher S, et al: Comparison of the aPTT with
    pediatric warfarin self-management pilot study in children with primar-        alternative tests for monitoring direct thrombin inhibitors in patient
    ily cardiac disease. Thromb Res 2010; 126:e110–e115                            samples. Am J Clin Pathol 2014; 141:665–674
45. Ansell J, Hirsh J, Hylek E, et al; American College of Chest Physicians:   67. Gasparovic H, Nathan NS, Fitzgerald D, et al: Severe argatroban-
    Pharmacology and management of the vitamin K antagonists:                      induced coagulopathy in a patient with a history of heparin-induced
    American College of Chest Physicians Evidence-Based Clinical                   thrombocytopenia. Ann Thorac Surg 2004; 78:e89–e91
    Practice Guidelines (8th Edition). Chest 2008; 133:160S–198S               68. Malherbe S, Tsui BC, Stobart K, et al: Argatroban as anticoagulant
46. Kalus JS: Pharmacologic interventions for reversing the effects of oral        in cardiopulmonary bypass in an infant and attempted reversal with
    anticoagulants. Am J Health Syst Pharm 2013; 70:S12–S21                        recombinant activated factor VII. Anesthesiology 2004; 100:443–445
47. Tran HA, Chunilal SD, Harper PL, et al; Australasian Society of            69. Oh JJ, Akers WS, Lewis D, et al: Recombinant factor VIIa for refrac-
    Thrombosis and Haemostasis (ASTH): An update of consensus                      tory bleeding after cardiac surgery secondary to anticoagulation
    guidelines for warfarin reversal. Med J Aust 2013; 198:198–199                 with the direct thrombin inhibitor lepirudin. Pharmacotherapy 2006;
48. Jennings DL, Jacob M, Chopra A, et al: Safety of anticoagulation               26:569–577
    reversal in patients supported with continuous-flow left-ventricular       70. Willey ML, de Denus S, Spinler SA: Removal of lepirudin, a recom-
    assist devices. ASAIO J 2014; 60:381–384.                                      binant hirudin, by hemodialysis, hemofiltration, or plasmapheresis.
49. Hassouna A, Allam H: Limited dose warfarin throughout pregnancy                Pharmacotherapy 2002; 22:492–499
    in patients with mechanical heart valve prosthesis: A meta-analysis.       71. Fischer KG, van de Loo A, Bohler J: Recombinant hirudin (lepirudin)
    Interact Cardiovasc Thorac Surg 2014; 18:797–806                               as anticoagulant in intensive care patients treated with continuous
50. Whitlock RP, Sun JC, Fremes SE, et al; American College of Chest               hemodialysis. Kidney Int Suppl 1999:S46–S50
    Physicians: Antithrombotic and thrombolytic therapy for valvular           72. Benz K, Nauck MA, Böhler J, et al: Hemofiltration of recombinant hiru-
    disease: Antithrombotic Therapy and Prevention of Thrombosis, 9th              din by different hemodialyzer membranes: Implications for clinical use.
    ed: American College of Chest Physicians Evidence-Based Clinical               Clin J Am Soc Nephrol 2007; 2:470–476
    Practice Guidelines. Chest 2012; 141:e576S–e600S                           73. Winkler AM, Tormey CA; Education Committee of the Academy of
51. Nishimura RA, Otto CM, Bonow RO, et al; ACC/AHA Task Force                     Clinical Laboratory Physicians and Scientists: Pathology consultation
    Members: 2014 AHA/ACC Guideline for the Management of                          on monitoring direct thrombin inhibitors and overcoming their effects
    Patients With Valvular Heart Disease: Executive summary: A                     in bleeding patients. Am J Clin Pathol 2013; 140:610–622
    report of the American College of Cardiology/American Heart                74. Sugg RM, Pary JK, Uchino K, et al: Argatroban tPA stroke study:
    Association Task Force on Practice Guidelines. Circulation 2014;               Study design and results in the first treated cohort. Arch Neurol
    129:2440–2492                                                                  2006; 63:1057–1062
52. Walenga JM: An overview of the direct thrombin inhibitor argatroban.       75. Murray PT, Reddy BV, Grossman EJ, et al: A prospective comparison
    Pathophysiol Haemost Thromb 2002; 32(Suppl 3):9–14                             of three argatroban treatment regimens during hemodialysis in end-
53. Di Nisio M, Middeldorp S, Büller HR: Direct thrombin inhibitors. N             stage renal disease. Kidney Int 2004; 66:2446–2453
    Engl J Med 2005; 353:1028–1040                                             76. Baruch L: Laboratory monitoring of anticoagulant medications: Focus
54. Steinmetzer T, Stürzebecher J: From fibrinogen and hirudin to syn-             on novel oral anticoagulants. Postgrad Med 2013; 125:135–145
    thetic anticoagulants. Rational design of thrombin inhibitors. Pharm       77. Yates S, Sarode R: Novel thrombin and factor Xa inhibitors:
    Unserer Zeit 2004; 33:196–205                                                  Challenges to reversal of their anticoagulation effects. Curr Opin
55. Lee CJ, Ansell JE: Direct thrombin inhibitors. Br J Clin Pharmacol             Hematol 2013; 20:552–557
    2011; 72:581–592                                                           78. Khadzhynov D, Wagner F, Formella S, et al: Effective elimination
56. Greinacher A, Warkentin TE: The direct thrombin inhibitor hirudin.             of dabigatran by haemodialysis. A phase I single-centre study in
    Thromb Haemost 2008; 99:819–829                                                patients with end-stage renal disease. Thromb Haemost 2013;
                                                                                   109:596–605
57. Veach SA, Franks AM, Allan MC: Severe anaphylactic reaction after
    repeated intermittent exposure to lepirudin. Pharmacotherapy 2007;         79. Siegal DM, Garcia DA, Crowther MA: How I treat target-specific oral
    27:760–765                                                                     anticoagulant-associated bleeding. Blood 2014; 123:1152–1158
58. Yarbrough PM, Varedi A, Walker A, et al: Argatroban dose reduc-            80. Cowell RP: Direct oral anticoagulants: Integration into clinical prac-
    tions for suspected heparin-induced thrombocytopenia complicated               tice. Postgrad Med J 2014; 90:529–539
    by child-pugh class C liver disease. Ann Pharmacother 2012;                81. Galanis T, Merli GJ: Contemporary treatment of venous thromboem-
    46:e30                                                                         bolic disease. Cardiol Clin 2015; 33:49–57
59. Moffett BS, Teruya J: Trends in parenteral direct thrombin inhibitor use   82. Walter RJ, Moores LK, Jiménez D: Pulmonary embolism: Current and
    in pediatric patients: Analysis of a large administrative database. Arch       new treatment options. Curr Med Res Opin 2014; 30:1975–1989
    Pathol Lab Med 2014; 138:1229–1232                                         83. Eikelboom JW, Connolly SJ, Brueckmann M, et al; RE-ALIGN
60. Oschman A: Survey results: Characterization of direct thrombin                 Investigators: Dabigatran versus warfarin in patients with mechanical
    inhibitor use in pediatric patients. J Pediatr Pharmacol Ther 2014;            heart valves. N Engl J Med 2013; 369:1206–1214
    19:10–15                                                                   84. Levy G: Clinical pharmacokinetics of aspirin. Pediatrics 1978;
61. Francis JL, Hursting MJ: Effect of argatroban on the activated partial         62:867–872
    thromboplastin time: A comparison of 21 commercial reagents. Blood         85. Israels SJ, Michelson AD: Antiplatelet therapy in children. Thromb Res
    Coagul Fibrinolysis 2005; 16:251–257                                           2006; 118:75–83
62. Ivandic B, Zorn M: Monitoring of the anticoagulants argatroban and         86. Carroll RC, Craft RM, Chavez JJ, et al: A Thrombelastograph whole
    lepirudin: A comparison of laboratory methods. Clin Appl Thromb                blood assay for clinical monitoring of NSAID-insensitive transcel-
    Hemost 2011; 17:549–555                                                        lular platelet activation by arachidonic acid. J Lab Clin Med 2005;
63. Pendleton R, Wheeler MM, Rodgers GM: Argatroban dosing of                      146:30–35
    patients with heparin-induced thrombocytopenia and an elevated             87. Wells WJ, Yu RJ, Batra AS, et al: Obstruction in modified Blalock
    aPTT due to antiphospholipid antibody syndrome. Ann Pharmacother               shunts: A quantitative analysis with clinical correlation. Ann Thorac
    2006; 40:972–976                                                               Surg 2005; 79:2072–2076
64. Gosselin R, Hawes E, Moll S, et al: Performance of various laboratory      88. Li JS, Yow E, Berezny KY, et al: Clinical outcomes of palliative surgery
    assays in the measurement of dabigatran in patients receiving thera-           including a systemic-to-pulmonary artery shunt in infants with cyanotic
    peutic doses: A prospective study based on peak and trough plasma              congenital heart disease: Does aspirin make a difference? Circulation
    levels. Am J Clin Pathol 2014; 141:262–267                                     2007; 116:293–297

S86            www.pccmjournal.org                                                                         March 2016 • Volume 17 • Number 3 (Suppl.)
You can also read