Sunteți pe pagina 1din 12

Pediatric Anesthesia ISSN 1155-5645

REVIEW ARTICLE

Coagulation considerations for infants and children undergoing cardiopulmonary bypass


Michael P. Eaton & Ellen M. Iannoli
Department of Anesthesiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA

Keywords cardiac surgery; heparin; protamine; antibrinolytic; congenital heart disease; coagulation Correspondence Michael P. Eaton, Department of Anesthesiology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Ave, Box 604, Rochester, NY 14642, USA Email: Michael_Eaton@urmc.rochester.edu Section Editor: Greg Hammer Accepted 6 October 2010 doi:10.1111/j.1460-9592.2010.03467.x

Summary Cardiac surgery involving cardiopulmonary bypass imposes a signicant pathophysiologic burden on patients. Pediatric patients are especially predisposed to the adverse effects of surgery and bypass on the coagulation system, with resultant bleeding, transfusion, and poor outcomes. These risks accrue to pediatric patients in inverse proportion to their weight and are attributable to hematologic immaturity, coagulation defects associated with congenital heart disease, bypass equipment, and the nature of congenital heart surgery. Standard anticoagulation does not completely inhibit thrombin generation, and continuous consumption of coagulation factor continues throughout bypass. Conventional measurements of anticoagulation during bypass poorly reect this incomplete anticoagulation, and alternate methods may improve anticoagulant therapy. Emerging therapies for blocking the effects of bypass on the coagulation system hold promise for decreasing bleeding and related complications, and improving outcomes in congenital heart surgery.

Introduction The development of cardiopulmonary bypass (CPB) has brought cardiac surgery from a very limited and hazardous endeavor to a routine and relatively safe practice that addresses an incredible variety of diseases and conditions in patients from a few hours to nine or more decades old. CPB is, however, a remarkably pathological state. The myriad adverse effects in patients on bypass are well described and resistant to prophylaxis and treatment (13). Children are particularly affected by CPB and cardiac surgery, and this risk is inversely proportional to the childs age or weight (4). Our goals for this review are to synthesize published data regarding coagulation in children undergoing cardiac surgery for a global understanding of the pathophysiology involved and to suggest evidence-based therapies both to minimize the hemostatic defects induced by CPB and to treat these defects when they result in excess postoperative bleeding. We review the literature describing the adverse effects of cardiopulmonary bypass on hemostasis and explore how the unique physiology of neonatal and other
Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

pediatric patients interacts with CPB pathophysiology. We will discuss current therapies to minimize the hemostatic problems associated with CPB, issues associated with the application of those therapies, and other therapeutic modalities supported in the literature but not yet commonly employed in clinical practice. Finally, we review evidence-based treatment of postCPB bleeding, including the use of recombinant activated factor VII. Cardiopulmonary bypass coagulopathy In the intact organism, blood circulates through a cardiovascular system entirely lined with endothelial cells. One of the primary functions of the endothelium is to maintain the uidity of blood by inhibiting platelets and preventing adhesion of brin. Although advances in circuit biopassivation have been made, the CPB circuit remains profoundly nonphysiologic. Administration of high-dose heparin prevents complete thrombosis of blood entering the CPB circuit, but low-level intravascular and intracircuit coagulation continues throughout bypass (5,6) (Figure 1). Initiation of CPB
31

Coagulation in children on bypass

M.P. Eaton and E.M. Iannoli

CPB
Contact activation Factor XII Kallikrein Inflammation

Plasminogen

Plasmin

Fibrinogen

Fibrin

Thrombin

Platelets

Factor VII Tissue factor Surgical trauma

of these platelets fragment under conditions of shear stress (11). Platelet glycoprotein Ib (von Willebrand) receptors are degraded in the circulation by plasmin, impairing adhesion (12). Once activated prematurely during CPB, platelets are not available for hemostasis after surgery. CPB is also a profoundly proinammatory state, with activation of multiple aspect of humoral (e.g., complement, interleukins) and cellular (monocytes, neutrophils) inammation. Although a complete discussion of the inammatory response to CPB is outside the scope of this review, it is important to understand that the inammatory and coagulation systems interact at multiple levels (13). More effective suppression of thrombin generation during CPB will decrease the inammatory response to CPB (14), and conversely, interventions aimed at decreasing inammation may improve hemostasis (15). Most of these details of the interaction of CPB and hemostasis have been described in adults (5). The same pathophysiology does develop in the infant and child (6) although it is altered by the unique physiology of the young patient with congenital heart disease. Coagulation issues unique to the pediatric patient Developmental hemostasis The human hemostatic system is immature at birth. Most pro- and anticoagulant protein levels are 30 70% lower relative to adults (1618), although some enzymes are in higher concentrations in neonates relative to adults (Figure 2). In addition, some neonatal coagulation proteins, such as plasminogen and brinogen, exist as fetal isoforms with adult amino acid sequences, but signicant differences in glycosylation (17,18). These differences may explain the altered sensitivities of neonatal coagulant proteins to both endogenous (19,20) and exogenous modulation (21). Differences in brinogen and plasminogen structure and function are likely to be particularly important in the effects of CPB, heparin, and medications administered to prevent or reduce bleeding. Thus the hemostatic system of the neonate is fundamentally different than that of the adult, yet balanced so as to allow clotting and clot lysis in response to injury. Maturation of hemostasis approximates the adult system for the most part by 612 months, but in some respects not until signicantly later in childhood. The normal neonate is capable of mounting a competent thrombotic response to vascular injury, with functional brinolysis and remodeling following. However, the response of the neonate and small child to
Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

Figure 1 Activation of the coagulation, inammatory, and brinolytic systems by cardiopulmonary bypass and surgery.

produces activation of the contact phase of coagulation with cleavage of factor XII to XIIa and prekallikrein to kallikrein (7). Kallikrein has a positive feedback effect on factor XII as well as producing activation of inammation and brinolysis. Factor XIIa initiates the intrinsic cascade of coagulation and, ultimately, thrombin activation. Surgical trauma causes release of tissue factor which further activates thrombin via the extrinsic pathway (8). Thrombin has many diverse effects (9). In addition to cleaving brinogen to brin and activating factor XIII to induce brin crosslinking, it produces platelet activation via the PAR-1 and PAR-4 receptors, has positive feedback effects on other coagulation proteins, activates thrombin-activatable brinolysis inhibitor, and stimulates the endothelium to release tissue factor and von Willebrand factor. It also has antithrombotic effects such as stimulation of endothelial cells to release tissue-type plasminogen activator, nitric oxide, and prostacyclin and binding with thrombomodulin to activate protein C. Upon initiation of CPB, brinogen adheres to the articial surfaces of the circuit. Platelets become activated and bind to the brinogen and degranulate (10). Some
32

M.P. Eaton and E.M. Iannoli

Coagulation in children on bypass

(a)

(b)

Cyanotic heart disease has been particularly associated with defects in coagulation. Chronic hypoxemia represents a profound stimulant for erythropoiesis. Excess production of red blood cells suppresses platelet production (25), and platelet counts have been reported to vary inversely with hematocrit in patients with cyanotic heart disease (26,27), with thrombocytopenia being relatively common. Waldman et al. (28) have also demonstrated a decrease in mean platelet survival in cyanotic patients. Deciencies in platelet adhesion and aggregation have also been reported (29,30). Increased brinolysis may occur in cyanotic patients (31,32), but this appears to be a relatively infrequent nding, and it is not clear whether this is attributable to low-grade disseminated intravascular thrombosis, primary brinolysis, or simply increased catabolic turnover of brinogen (27). Poor cardiac output, hypoxemia, and chronic passive congestion of the liver may also lead to underproduction of coagulation factors, especially the vitamin-K-dependent factors II, VII, IX, and X (27). Patients with hypoplastic left heart syndrome are particularly affected by factor deciencies, with decreased levels of brinogen, antithrombin (ATIII), factors II, VII, IX, X, V, and VIII, and proteins C and S (33,34). These deciencies are most pronounced prior to rst-stage palliation, but many persist through Fontan completion (34). Given the likelihood of coagulation abnormalities in these patients, heightened vigilance and proactive therapy should be routine practice for cyanotic children. Hemodilution A normal-sized adult with a blood volume of 5000 ml will undergo hemodilution of almost 25% with a typical prime volume of 1600 ml (measuring hemodilution as a percent of the whole system). This moderate dilution does not typically reduce clotting factors to the point of causing coagulopathy, but signicant thrombocytopenia is not uncommon (35). A 3- to 3.5-kg neonate with a blood volume of 270315 ml is likely to undergo 60% hemodilution. Because an infants blood volume is much smaller than that of the prime in the CPB pump, hemodilution alone produces impaired hemostasis related to thrombocytopenia and coagulation factor dilution (6,36,37). This dilution is likely even more signicant given the lower initial concentrations of many factors. Even in the polycythemic cyanotic patient, inclusion of allogeneic red blood cells in the priming volume is nearly universal with neonates and small infants (38). Thus, these high-risk patients begin CPB with an unavoidable donor exposure. Inclusion of donor red cells in the pump prime
33

Figure 2 Concentration of procoagulant (a) and anticoagulant (b) proteins over time from birth through adulthood. ATIII, antithrombin III; Alpha-2 MG, alpha-2 macroglobulin. Data from Andrew et al. (16).

CPB-induced hemodilution is qualitatively different to that of adults as hemodilution is more profound, and baseline levels of coagulation proteins are signicantly different. Congenital heart disease and coagulation Congenital heart disease itself has long been known to be associated with coagulation abnormalities (22). Goldschmidt et al. (23) showed that patients with congenital heart disease have abnormalities of platelet turnover, with increased peripheral destruction and a higher proportion of young sticky platelets. Gill et al. (24) reported a series of patients with congenital heart disease with prolonged bleeding times associated with signicantly reduced levels of large multimer von Willebrand factor. Four of ve of the patients studied after correction of their congenital defects had normalization of the von Willebrand factor.
Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

Coagulation in children on bypass

M.P. Eaton and E.M. Iannoli

has been shown to adversely affect cardiopulmonary function and contribute to inammation (39). Transfusion presents signicant risks in all patients. Although the risk of viral disease transmission has decreased with more thorough testing of the blood supply, other adverse effects of transfusion remain, such as transfusion reactions, immune modulation, impairment of microvascular ow, and even mortality (40). Surgical complexity Congenital heart disease is extremely varied and complex. In the 20022005 New York State Congenital Cardiac Surgery database of 5466 cases, (http://www. health.state.ny.us/statistics/diseases/cardiovascular/heart_ disease/docs/2002-2005_pediatric_congenital_cardiac_ surgery.pdf, last accessed June 14, 2010), over 100 different congenital cardiac diagnoses are listed as the primary surgical indication. Twenty-four different diagnoses were present in at least 100 patients. Thus, the congenital heart surgeon has far less experience and familiarity with the operations he/she performs than the adult cardiac surgeon. A trend toward performing complete repairs in infancy and the renement of surgical and CPB techniques have increased the incidence of high-complexity repairs in very small patients (41). Repair of complex congenital anomalies often requires multiple suture lines, long CPB runs, deep hypothermia, and circulatory arrest, increasing the risk of bleeding and transfusion (42). Current and future therapies Anticoagulation It is perhaps counterintuitive that more anticoagulation results in less bleeding, but this is the case for inhibition of the coagulation system during CPB (8,43). Adequate preservation of the coagulation system during CPB minimizes postbypass coagulopathy and postoperative bleeding. An ideal anticoagulation regimen would completely suppress all aspects of coagulation during CPB and be completely reversible at the end of the bypass. There is no single drug that accomplishes this. Heparin Unfractionated heparin is a mixture of polysulfated glycosaminoglycans of molecular weights from 3000 to 30 000 Daltons (44). It was originally discovered in 1926, making it one of the oldest drugs still in use. Heparin exerts its anticoagulant effect by binding to ATIII, multiplying its activity 1000-fold (45). Suf34

ciently large (18 saccharides) heparin molecules produce inhibition of both factor Xa and thrombin, thus interfering with coagulation at two successive steps. However, heparin does not produce complete inhibition of coagulation during CPB; thrombin continues to be generated despite high heparin concentrations (9). Because heparin requires an intrinsic cofactor to work, individuals decient in ATIII will have a diminished anticoagulant response to routinely used doses. Heparin resistance is a relatively commonly encountered issue in adult cardiac surgery (46), and given the baseline lower concentration of ATIII in infants, it is not surprising that the anticoagulant effect of heparin in very young patients is both more variable and less likely to be adequate (47). Other intrinsic molecules, such as heparin cofactor II and a-2 macroglobulin, are more important inhibitors of thrombin in children (48). However, Guzzetta et al. (48) were unable to demonstrate any relationship between preoperative levels of ATIII, heparin cofactor II, or a-2 macroglobulin and heparin effect as measured by the activated clotting time (ACT) in children. Heparins anticoagulant effect is commonly measured using the ACT. Typically, infants and children are managed using an ACT value established in adults as reecting adequate anticoagulation for CPB, usually 400480 s, depending on institutional preference. It has been demonstrated that the ACT is a poor indicator of anticoagulation in infant undergoing CPB, as factors other than heparin, such as hemodilution and hypothermia, prolong the ACT despite diminishing heparin concentrations and ongoing thrombin generation (49,50). Thus, heparin as commonly employed in congenital heart surgery is associated with inadequate anticoagulation and ongoing formation of thrombin and consumption of coagulant proteins. Despite this, heparin remains the preferred anticoagulant for CPB in children. Better use of heparin may be facilitated by alternate strategies for dosing and monitoring in children. In a retrospective study of 90 adults and children receiving heparin for CPB, DErrico et al. (47) found that infants were signicantly less responsive to heparin and had greater variability in that response. The authors suggested an initial heparin dose as high as 500 unitskg)1 may be needed in infants, while 300 unitskg)1 was likely adequate in older children and adults. Guzzetta (51) has shown that ACT values are poorly reective of heparin effect in infants and that the use of patient-specic heparin management using automated heparin doseresponse calculation and heparin level monitoring was more effective in suppressing thrombin generation during CPB than conventional heparin dosing and monitoring (52).
Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

M.P. Eaton and E.M. Iannoli

Coagulation in children on bypass

Although individualized heparin dosing may be the best strategy, point-of-care heparin management systems may not be available at every institution. At the very least, it is important to recognize that standard celite- or kaolin-activated tube-technology ACTs are likely to overestimate heparin anticoagulation. Empiric interval redosing may avoid the effects of inadequate anticoagulation. Based on data from Guzzetta et al. (51,52), heparin levels may be expected to fall 4060% in infants during 80 min of CPB. Redosing with 1/3 to of the original heparin dose after 60 min of CPB may be reasonable. Heparin at least anticoagulates blood to the point that extracorporeal circulation is possible. It is also readily reversible at the completion of CPB with protamine. Protamine reversal of heparin in vivo was originally described by Jaques et al. in 1938 (53). Protamine is associated with a number of adverse reactions including histamine-dependent vasodilation (54), anaphylaxis (55), and catastrophic pulmonary hypertension (56). Severe reactions to protamine may be less common in infants and children than in adults (57,58). Determining the dose of protamine is difcult without ongoing knowledge of circulating concentrations of heparin. Basing the protamine dose on automated heparin/protamine titration will produce a smaller dose than predicted bases on the initial heparin bolus and the last ACT (47). It is important to avoid administering excess protamine, as unbound protamine has anticoagulant properties (59,60). A further complicating factor in heparin reversal is ultraltration, which is used in 75% of centers performing congenital heart surgery (38). As heparin molecules are too large to pass through the lter, heparin levels increase during ultraltration (61) and patients who have undergone high-volume ultraltration may require correspondingly higher doses of protamine. Heparin-induced thrombocytopenia (HIT) is a clinical syndrome resulting from the formation of antibodies to heparinplatelet factor 4 complexes. These antibodies cause platelet aggregation and thrombocytopenia, often complicated by thrombosis. Conversion to positive antibody status is common after adult heart surgery (62), and in older children undergoing congenital heart surgery (63), but is relatively uncommon in infants (64). There is increasing recognition that HIT may be a more common contributor to thrombotic morbidity in congenital heart surgery than previously thought (65). Use of heparin alone for CPB in patients with acute HIT is inadvisable as catastrophic thrombosis has been reported in this setting (66). Ideally, cardiac surgery should be delayed until antibodies have been cleared from the circulation, but
Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

occasionally, surgery is more urgent. In this setting, an alternative anticoagulant such as a direct thrombin inhibitor should be employed (67). In institutions without experience using alternate agents for CPB, it may be advisable to use heparin in combination with a platelet antagonist such as epoprostenol or tiroban (67). Alternate agents There is some evidence that direct thrombin inhibitors are actually better inhibitors of CPB-induced activation of coagulation than heparin (68). Agents other than heparin have been used successfully to anticoagulate pediatric patients for CPB, but experience is very limited (69,70). The direct thrombin inhibitors r-hirudin (71), argatroban (72), and bivalirudin (73) have all been used for CPB and are likely the best alternatives currently available. Excessive bleeding has been reported with the rst two agents (71,74,75), which have a duration of action too long to be ideal in the CPB setting. Bivalirudin has a shorter elimination halflife and organ-independent elimination and is most likely the best of these three agents as an alternative to heparin for CPB. As bivalirudin undergoes metabolism in the blood, it must be recognized that stasis in both the patient and circuit must be avoided and a continuous infusion used to maintain anticoagulation and avoid thrombosis (73). Early research has demonstrated some advantage to combining heparin and bivalirudin for CPB anticoagulation (76). No clear guidelines exist for dosing or monitoring of direct thrombin inhibitors in children. Antibrinolytics Hyperbrinolysis is one of the primary mechanisms leading to post-CPB bleeding (77,78) although this is perhaps less well established in infants and children than in adults (79,80). The best evidence for brinolysis contributing to post-CPB bleeding in infants is the efcacy of antibrinolytic therapy. The antibrinolytic drugs aprotinin, epsilon-aminocaproic acid (EACA), and tranexamic acid (TA) have been shown to decrease bleeding and transfusion in multiple studies of pediatric cardiac surgery (81). Prior to its withdrawal from marketing, 96% of pediatric cardiac surgery centers used aprotinin (38). However, the evidence of benet in pediatric patients is actually better for the lysine analogs EACA and TA (81). Aprotinin was withdrawn from the market in May 2008 after multiple reports surfaced concerning adverse effects of the drug, most notably renal failure, in adult patients (8284). Controversy continues over the riskbenet balance of this
35

Coagulation in children on bypass

M.P. Eaton and E.M. Iannoli

drug, particularly for pediatric patients in whom adverse effects have not yet been shown. The lysine analogs EACA and TA inhibit brinolysis by interfering with the binding of plasminogen to brin, a necessary step in the formation of plasmin, the primary brinolytic enzyme (85). Both drugs have been shown in multiple studies to decrease blood loss and transfusion in pediatric patients undergoing surgery with CPB (8694). Dosing schemes for these agents have been quite variable and not based on sound pharmacologic principles. Ririe et al. (95) studied the pharmacokinetics of EACA in 8 infants and children up to 4 years of age. Using these data, they recommended a loading dose of 75 mgkg)1, an infusion of 75 mgkg)1 per hour, and a pump prime dose of 75 mgkg)1. The volume of the CPB pump is relatively xed; it varies in a stepwise fashion based on patient size. The purpose of adding EACA or TA to the pump prime is to maintain plasma levels; therefore, basing the prime dose on the volume of prime rather than on the weight of the patient is more logical. We have found that a modication of Riries dosing scheme, with a prime dose of 250 mcgml)1 of prime volume, establishes and maintains the target concentration (130 mcgml)1) throughout CPB in neonates (96). Neonates however require lower levels of EACA to fully inhibit brinolysis (97), so lower doses may be justied. The pharmacokinetics of TA in the pediatric population has not been established, and pharmacologically based dosing guidelines are unavailable. Chauhan et al. (87) compared four doses of TA in 150 cyanotic children undergoing cardiac surgery. They found the most effective dose to be a 10 mgkg)1 load, 10 mgkg)1 in the pump prime, and a 10 mgmg)1 bolus after separation from CPB. Platelet anesthesia In vivo, the predominant mechanism for blood coagulation is somewhat dependent on environment (98). In the venous system, ow is relatively sluggish, and shear forces are low. Humoral (soluble factor-related) coagulation is predominant in this environment, as is demonstrated by the utility of medications focusing on factor inhibition in prophylaxis and treatment of venous thrombosis. In the arterial system, shear stresses are considerably higher and platelet activity is signicantly more important, as evidenced by the utility of platelet antagonists such as aspirin, dipyridamole and clopidogrel in the prevention of arterial thrombotic events. The CPB circuit also has portions of low (reservoir) and high (arterial line) shear stress. Despite its noted inadequacies, heparin addresses
36

humoral anticoagulation with some efcacy. Traditional approaches to anticoagulation do not address platelet activation. Platelet anesthesia is a concept explored in the 1990s and early 2000s in which an agent is administered at the beginning of CPB to minimize platelet activation, with the intention of improving platelet function and hemostasis after CPB. For this to be effective, the agent employed must be of relatively brief duration and highly effective in blocking platelet activation. The glycoprotein IIb-IIIa antagonists tiroban and eptibatide have both been found to protect platelets from activation during CPB (99,100). Koster et al. found that the addition of tiroban to heparin anticoagulation in adult patients undergoing aorto-coronary bypass preserved platelet numbers and decreased measures of inammation after CPB. Blood loss was not signicantly different between tiroban-treated patients and controls at 12 h after surgery despite measurably decreased platelet aggregation after CPB in the tiroban group. Nitric oxide has been shown effective in attenuating platelet activation and preserving post-CPB platelet function in a baboon model (99). Nitric oxide has the advantages of being very short-acting and may be administered via the oxygenator, which has the largest surface area for contact activation of the CPB circuit. The ultrashort-acting glycoprotein IIb-IIIa antagonist FK633 was shown to be effective in preventing CPB-induced thrombocytopenia and preventing platelet adhesion to the circuit (101). Little attention has been given to platelet anesthesia in pediatric cardiac surgery, but attenuation of the platelet response seems an obvious therapy to complete anticoagulation for CPB. Further development and approval of FK633 or other short-acting antiplatelet agents such as cangrelor (102) may spur research in this area. Improving the CPB circuit Technical limitations have dictated a lower limit to the size of perfusion equipment and CPB circuits used in pediatric patients. This has traditionally resulted in much greater degrees of hemodilution in infants and small children relative to adults with functionally important decreases in coagulation factors, platelets, and hematocrit (6,36,37). The conventional response to this has been to include donor red cells in the prime volume with many centers including whole blood or plasma (103). In a 2005 survey, Groom et al. (38) found that 24% of reporting hospitals used whole blood in the prime. The literature is mixed on the efcacy of fresh whole blood in improving outcomes in congenital heart surgery. Manno et al. reported in
Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

M.P. Eaton and E.M. Iannoli

Coagulation in children on bypass

1991 (103) that children receiving very fresh ( 24 h < old) or fresh (2448 h old) whole blood for a variety of congenital heart operations had signicantly less postoperative bleeding than patients treated with reconstituted whole blood (mixed-component red cells, plasma, and platelets). Mou et al. (104) however reported no improvement in hemostasis in infants randomized to receive fresh whole blood ( 48 h old) over < a group receiving a combination of plasma and red blood cells and found an increase in edema and ICU length of stay in the fresh whole blood group. Most recently, Greunwald et al. (105) randomized 64 neonates to receive either reconstituted fresh whole blood (single donor, 2 days old) or standard component therapy, nding less bleeding in the fresh whole blood group, as well as a decreased need for inotropes, shorter time on mechanical ventilation, and shorter length of stay. It should be noted that the mean age of transfused units in the control group in this study was 14 days, as opposed to 2 days in the fresh whole blood group, so it is unclear that these results are simply a reection of the morbidity resulting from transfusion of old blood (106,107). There is a lack of convincing evidence that including any blood products in the prime volume other than a minimum volume of (preferable fresh) red blood cells improves outcomes. Continued technological developments with greatly reduced circuit volumes may obviate discussion of what blood products to add to the prime by eliminating the need for even red cells in all but the smallest patients. Circuit prime volumes as low as 140180 ml are now being used routinely and have enabled the performance of CPB on small infants with an asanguineous prime (108,109). Although avoiding routine exposure to allogeneic blood components by reducing circuit size seems an obviously benecial goal (110), removing certain components of the circuit such as the arterial line lter in an attempt to decrease volume does not yet have sufcient evidence of benet to offset a theoretical increased risk of harm (111). Recently developed circuits incorporate an arterial line lter into the oxygenator, maintaining the safety of ltration while maintaining low prime volumes (112). Although tissue factor released because of surgical trauma may be a major source of coagulation activation during CPB, contact with the articial surfaces of the CPB circuit initiates activation of the coagulation, inammatory, and brinolytic systems (7). Making the circuit components more biocompatible should theoretically decrease contact activation and downstream consequences. Several biopassive coatings have been developed to enhance the biocompatibility of CPB circuits and minimize hemostatic and inammatory actiPediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

vation by mimicking some of the properties of endothelium, such as negative ionic charge and hydrophilicity. Partially (oxygenator) or completely coated circuits are currently used by the majority of institutions performing pediatric heart surgery (38). Original coated circuits employed heparin, bound to the circuits surfaces either ionically (Durao II; Baxter Healthcare, Deereld, IL, USA) or covalently (Carmeda; Medtronic Cardiovascular, Minneapolis, MN, USA). Recently, polyanionic hydrophilic or amphiphilic polymers have been developed such as polymethoxyethylacrylate (PMEA) (X-coating; Terumo Cardiovascular, Ann Arbor, MI, USA), phosphoryl choline (ECCO/Synergy; Sorin Group, Mirandola, Italy), and polyethylene oxide (Trillium Biosurface; Medtronic Cardiovascular, Minneapolis, MN, USA). The Trillium coating employs covalently bound heparin and negatively charged sulfate and sulfonate groups in addition to the polymer. (http://www.medtr onic.com/for-healthcare-professionals/products-therapi es/cardiovascular/perfusion-products/biocompatible-sys tems/trillium-biosurface/index.htm, last accessed June 21, 2010). Studies specically investigating the pediatric use of coated circuits are few and generally include small numbers of patients of varying age and diagnosis. Olsson et al. (113) and Jensen et al. (114) each demonstrated a decrease in complement activation in small groups (total N = 60) of children randomized to heparin-coated versus uncoated circuits. Suzuki et al. (115) randomized 11 children to PMEA-coated or uncoated CPB for repair of ventricular septal defects. Decreased thrombin and platelet activation was demonstrated in the PMEA group. Andreasson et al. studied 40 infants <10 kg randomized to a biocompatible circuit (closed circuit, centrifugal pump) including heparin coating to a standard circuit (open reservoir, roller pump) without heparin coating. Activation of brinolysis was lower in the biocompatible group, but there was no signicant difference in thrombin activation. Eisses et al. performed an observational study of 26 patients undergoing repair of atrial or ventricular septal defects using an uncoated or PMEA-coated oxygenator. They showed no difference in thrombin generation and a paradoxical increase in brinolysis in the PMEA group, but higher platelet counts and less bleeding at 12 and 24 h after surgery. Deptula et al. (116) compared 16 patients <15 kg undergoing CPB with a PMEA-coated circuit with retrospective matched controls. They found the PMEA patients received fewer transfusions of red blood cells and plasma and had a lower lactate level for the rst 12 postoperative hours. In the largest pediatric study,
37

Coagulation in children on bypass

M.P. Eaton and E.M. Iannoli

Miyaji et al. (117) retrospectively compared 100 selected patients <10 kg who had undergone cardiac surgery using a heparin-coated circuit with a matched group of 50 operated on with noncoated circuits. No differences in bleeding outcomes of length of stay were found, but the heparin-coated group was mechanically ventilated for 2 fewer days than the uncoated group. Given moderate evidence of benet even with very small studies and the clear evidence of poor control of thrombin generation with conventional heparinization and CPB, it seems reasonable to include biopassive circuits in an extracorporeal perfusion plan for cardiac surgery. Postoperative bleeding Transfusion algorithms Despite our best efforts at minimizing the coagulopathic effects of CPB, appropriate prophylaxis, and optimal anticoagulation, excessive postoperative bleeding is a relatively common problem in pediatric cardiac surgery (118). Treatment of excessive bleeding is often empirical, but guided therapy is more likely to be effective and safer (119). Multiple algorithms have been described to guide hemostatic transfusion in adult cardiac surgery (120123), with guided therapy showing benets of deceased transfusion with equivalent or less bleeding. Currently, no single algorithm has been demonstrated to be superior, so that it is impossible to recommend a specic set of tests (122,123). Although there are less data validating algorithm-guided transfusion in pediatric cardiac surgery, both routine laboratory testing (119) and thromboelastography (124) after bypass have been shown to correlate well with abnormal bleeding. The best choice of coagulation tests to guide post-CPB hemostatic transfusion likely depends most on what test one can obtain in a rapid and reliable fashion at the individual institution. Thromboelastography can be performed during CPB using heparinase correction, thus providing information useful even before weaning and protamine administration (80). Fibrinogen levels and platelet counts can similarly be performed during CPB. Recombinant factor VIIa Recombinant activated factor VII (rFVIIa) (Novoseven; Novo Nordisk Healthcare, Princeton, NJ, USA) is FDA-approved for patients with hemophilia A or B with inhibitors or congenital factor VII deciency. It is able to directly activate factor X in the presence of activated platelets, obviating the need for factors IX and VIII. rFVIIa has been used off-label to treat
38

intractable bleeding associated with trauma (125) and surgical bleeding, particularly in cardiac surgery (126,127). Published data on factor rFVIIa use in pediatric heart surgery are limited, with the entire published experience being barely over 100 cases. Individual case reports and case series have mostly shown control of bleeding after administration of VIIa (128 137). In one of the larger series, 24 pediatric cardiac surgical patients, including 12 on ECMO, were compared with retrospective controls. A signicant decrease in bleeding was seen in 23 of the 24 treated patients after rFVIIa administration (128). Six patients (25%) developed thrombotic complications. Karsies et al. (138) reported a case series of 25 patients who had received factor rFVIIa within a week after surgery for congenital heart disease. These were compared with 50 matched retrospective controls. Transfusion volume markedly decreased after administration of rFVIIa, as did chest tube output. No signicant difference in mortality or length of stay between groups was found. Eight percent of treated patients and 4% of controls had thrombotic complications, but this was not statistically signicant. In the only prospective randomized trial of rFVIIa in pediatric heart surgery, Ekert et al. (139) randomized 82 patients under 1 year old undergoing surgery for congenital heart disease to receive a single dose of rFVIIa (40 mcgkg)1) or placebo after protamine administration. No difference was found in bleeding or transfusion outcomes, and the time to chest closure was longer in the rFVIIa group. OConnell et al. (140) reviewed the FDA Adverse Event Reporting System database for thromboembolic events associated with the off-label use of factor VIIa in 2006. Thrombotic complication occurred in 185 adults and children (1 month to 91 years old) and included 50 deaths. Limited clinical evidence suggests that administration of rFVIIa is often effective in reducing coagulopathic bleeding resistant to conventional therapy after congenital heart surgery. This improvement comes with signicant risk of thrombosis in a group of patients already at high risk of thrombotic complications. rFVIIa cannot produce hemostasis alone and should only be administered after transfusion of sufcient platelets, plasma, and brinogen to form the substrate for hemostasis. Conclusions Patients undergoing surgery for congenital heart disease are at high risk of bleeding and transfusion during and after surgery. That risk can be reduced by understanding the issues specic to coagulation and anticoaPediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

M.P. Eaton and E.M. Iannoli

Coagulation in children on bypass

gulation in children, especially neonates, and using that understanding to craft a multimodal approach to minimizing the physiologic trespass of CPB. The published data upon which one must base a plan of care suffer from small numbers, differing subject populations, and outcomes but nevertheless offers some guiReferences
1 Mammen EF, Koets MH, Washington BC et al. Hemostasis changes during cardiopulmonary bypass surgery. Semin Thromb Hemost 1985; 11: 281292. 2 Kirklin JK, Blackstone EH, Kirklin JW. Cardiopulmonary bypass: studies on its damaging effects. Blood Purif 1987; 5: 168 178. 3 Weiland AP, Walker WE. Physiologic principles and clinical sequelae of cardiopulmonary bypass. Heart Lung 1986; 15: 3439. 4 Williams GD, Bratton SL, Riley EC et al. Association between age and blood loss in children undergoing open heart operations. Ann Thorac Surg 1998; 66: 870875. Discussion 875876. 5 Bick RL. Alterations of hemostasis associated with cardiopulmonary bypass: pathophysiology, prevention, diagnosis, and management. Semin Thromb Hemost 1976; 3: 5982. 6 Kern FH, Morana NJ, Sears JJ et al. Coagulation defects in neonates during cardiopulmonary bypass. Ann Thorac Surg 1992; 54: 541546. 7 Campbell DJ, Dixon B, Kladis A et al. Activation of the kallikrein-kinin system by cardiopulmonary bypass in humans. Am J Physiol Regul Integr Comp Physiol 2001; 281: R1059R1070. 8 De Somer F, Van Belleghem Y, Caes F et al. Tissue factor as the main activator of the coagulation system during cardiopulmonary bypass. J Thorac Cardiovasc Surg 2002; 123: 951958. 9 Edmunds LH Jr, Colman RW. Thrombin during cardiopulmonary bypass. Ann Thorac Surg 2006; 82: 23152322. 10 Addonizio VP Jr, Smith JB, Strauss JF III et al. Thromboxane synthesis and platelet secretion during cardiopulmonary bypass with bubble oxygenator. J Thorac Cardiovasc Surg 1980; 79: 9196. 11 Slaughter TF, Sreeram G, Sharma AD et al. Reversible shear-mediated platelet dysfunction during cardiac surgery as assessed by the PFA-100 platelet function analyzer. Blood Coagul Fibrinolysis 2001; 12: 8593. 12 Reilly TM, Forsythe MS, Racanelli AL et al. Recombinant plasminogen activator inhibitor-1 protects platelets against the

dance. It is clear that larger studies must be performed to discern real outcome benet of many of these therapies. It is almost certain that these studies will need to be multicenter in nature both to provide sufcient power and to ensure that the value of each intervention applies broadly.

13

14

15

16

17

18

19

20

21

22

23

24

inhibitory effects of plasmin. Thromb Res 1993; 71: 6168. Esmon CT. The interactions between inammation and coagulation. Br J Haematol 2005; 131: 417430. Koster A, Chew D, Kuebler W et al. High antithrombin III levels attenuate hemostatic activation and leukocyte activation during cardiopulmonary bypass. J Thorac Cardiovasc Surg 2003; 126: 906907. Cappabianca G, Rotunno C, de Luca Tupputi Schinosa L et al. Protective Effects of Steroids in Cardiac Surgery: A Meta-Analysis of Randomized Double-Blind Trials. J Cardiothorac Vasc Anesth 2010; (in press). Andrew M, Paes B, Milner R et al. Development of the human coagulation system in the full-term infant. Blood 1987; 70: 165172. Kuhle S, Male C, Mitchell L. Developmental hemostasis: pro- and anticoagulant systems during childhood. Semin Thromb Hemost 2003; 29: 329338. Albisetti M. The brinolytic system in children. Semin Thromb Hemost 2003; 29: 339348. Ries M, Easton RL, Longstaff C et al. Differences between neonates and adults in tissue-type-plasminogen activator (t-PA)-catalyzed plasminogen activation with various effectors and in carbohydrate sequences of brinogen chains. Thromb Res 2001; 103: 173184. Ries M, Easton RL, Longstaff C et al. Differences between neonates and adults in carbohydrate sequences and reaction kinetics of plasmin and alpha(2)-antiplasmin. Thromb Res 2002; 105: 247256. Zenker M, Ries M. Differences between neonates and adults in plasmin inhibitory and antibrinolytic action of aprotinin. Thromb Res 2002; 107: 1721. Bahnson H, Ziegler R. A Consideration of the causes of death following operation for congenital heart disease of the cyanotic type. Surg Gynecol Obstet 1950; 90: 6076. Goldschmidt B, Sarkadi B, Gardos G et al. Platelet production and survival in cyanotic congenital heart disease. Scand J Haematol 1974; 13: 110115. Gill JC, Wilson AD, Endres-Brooks J et al. Loss of the largest von Willebrand fac-

25 26

27

28

29

30

31

32

33

34

35

36

tor multimers from the plasma of patients with congenital cardiac defects. Blood 1986; 67: 758761. Beguin Y. Erythropoietin and platelet production. Haematologica 1999; 84: 541547. Gross S, Keefer V, Liebman J. The platelets in cyanotic congenital heart disease. Pediatrics 1968; 42: 651658. Henriksson P, Varendh G, Lundstrom NR. Haemostatic defects in cyanotic congenital heart disease. Br Heart J 1979; 41: 2327. Waldman JD, Czapek EE, Paul MH et al. Shortened platelet survival in cyanotic heart disease. J Pediatr 1975; 87: 7779. Mauer HM, McCue CM, Caul J et al. Impairment in platelet aggregation in congenital heart disease. Blood 1972; 40: 207 216. Bhargava M, Sanyal SK, Thapar MK et al. Impairment of platelet adhesiveness and platelet factor 3 activity in cyanotic congenital heart disease. Acta Haematol 1976; 55: 216223. Brodsky I, Gill DN, Lusch CJ. Fibrinolysis in congenital heart disease. Preoperative treatment with epsilon-aminocaproic acid. Am J Clin Pathol 1969; 51: 5157. Dennis LH, Stewart JL, Conrad ME. Heparin treatment of haemorrhagic diathesis in cyanotic congenital heart-disease. Lancet 1967; 1: 10881089. Cholette JM, Rubenstein JS, Aleris GM et al. Elevated risk of thrombosis in neonates undergoing initial palliative cardiac surgery. Ann Thorac Surg 2007; 84: 1320 1325. Odegard KC, Zurakowski D, DiNardo JA et al. Prospective longitudinal study of coagulation proles in children with hypoplastic left heart syndrome from stage I through Fontan completion. J Thorac Cardiovasc Surg 2009; 137: 934941. Khuri SF, Wolfe JA, Josa M et al. Hematologic changes during and after cardiopulmonary bypass and their relationship to the bleeding time and nonsurgical blood loss. J Thorac Cardiovasc Surg 1992; 104: 94107. Paparella D, Brister SJ, Buchanan MR. Coagulation disorders of cardiopulmonary bypass: a review. Intensive Care Med 2004; 30: 18731881.

Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

39

Coagulation in children on bypass

M.P. Eaton and E.M. Iannoli

37 Mossinger H, Dietrich W, Braun SL et al. High-dose aprotinin reduces activation of hemostasis, allogeneic blood requirement, and duration of postoperative ventilation in pediatric cardiac surgery. Ann Thorac Surg 2003; 75: 430437. 38 Groom RC, Froebe S, Martin J et al. Update on pediatric perfusion practice in North America: 2005 survey. J Extra Corpor Technol 2005; 37: 343350. 39 Karamlou T, Schultz JM, Silliman C et al. Using a miniaturized circuit and an asanguineous prime to reduce neutrophilmediated organ dysfunction following infant cardiopulmonary bypass. Ann Thorac Surg 2005; 80: 613. Discussion 1314. 40 Spiess BD. Transfusion of blood products affects outcome in cardiac surgery. Semin Cardiothorac Vasc Anesth 2004; 8: 267 281. 41 Zannini L, Borini I. State of the art of cardiac surgery in patients with congenital heart disease. J Cardiovasc Med (Hagerstown) 2007; 8: 36. 42 Williams GD, Bratton SL, Ramamoorthy C. Factors associated with blood loss and blood product transfusions: a multivariate analysis in children after open-heart surgery. Anesth Analg 1999; 89: 5764. 43 Despotis GJ, Joist JH, Hogue CW et al. The impact of heparin concentration and activated clotting time monitoring on blood conservation. J Thorac Cardiovasc Surg 1994; 110: 4654. 44 Hirsh J, Anand SS, Halperin JL et al. Mechanism of action and pharmacology of unfractionated heparin. Arterioscler Thromb Vasc Biol 2001; 21: 10941096. 45 Dietrich W, Braun S, Spannagl M et al. Low preoperative antithrombin activity causes reduced response to heparin in adult but not in infant cardiac-surgical patients. Anesth Analg 2001; 92: 6671. 46 Koster A, Fischer T, Gruendel M et al. Management of heparin resistance during cardiopulmonary bypass: the effect of ve different anticoagulation strategies on hemostatic activation. J Cardiothorac Vasc Anesth 2003; 17: 171175. 47 DErrico C, Shayevitz JR, Martindale SJ. Age-related differences in heparin sensitivity and heparin-protamine interactions in cardiac surgery patients. J Cardiothorac Vasc Anesth 1996; 10: 451457. 48 Guzzetta NA, Miller BE, Todd K et al. Clinical measures of heparins effect and thrombin inhibitor levels in pediatric patients with congenital heart disease. Anesth Analg 2006; 103: 11311138. 49 Guzzetta NA, Miller BE, Todd K et al. An evaluation of the effects of a standard heparin dose on thrombin inhibition dur-

50

51

52

53 54

55

56

57

58

59

60

61

62

ing cardiopulmonary bypass in neonates. Anesth Analg 2005; 100: 12761282. Owings JT, Pollock ME, Gosselin RC et al. Anticoagulation of children undergoing cardiopulmonary bypass is overestimated by current monitoring techniques. Arch Surg 2000; 135: 10421047. Guzzetta NA, Monitz HG, Fernandez JD et al. Correlations Between Activated Clotting Time Values and Heparin Concentration Measurements in Young Infants Undergoing Cardiopulmonary Bypass. Anesth Analg 2010; 111: 173179. Guzzetta NA, Bajaj T, Fazlollah T et al. A comparison of heparin management strategies in infants undergoing cardiopulmonary bypass. Anesth Analg 2008; 106: 419 425. Jaques LB. The nitrogen partition in blood clotting. Biochem J 1938; 32: 11811184. Kambam J, Meszaros R, Merrill W et al. Prophylactic administration of histamine1 and histamine2 receptor blockers in the prevention of protamine-related haemodynamic effects. Can J Anaesth 1990; 37: 420422. Nybo M, Madsen JS. Serious anaphylactic reactions due to protamine sulfate: a systematic literature review. Basic Clin Pharmacol Toxicol 2008; 103: 192196. Lowenstein E, Johnston WE, Lappas DG et al. Catastrophic pulmonary vasoconstriction associated with protamine reversal of heparin. Anesthesiology 1983; 59: 470473. Seifert HA, Jobes DR, Ten Have T et al. Adverse events after protamine administration following cardiopulmonary bypass in infants and children. Anesth Analg 2003; 97: 383389. Kimmel SE, Sekeres M, Berlin JA et al. Mortality and adverse events after protamine administration in patients undergoing cardiopulmonary bypass. Anesth Analg 2002; 94: 14021408. Grifn MJ, Rinder HM, Smith BR et al. The effects of heparin, protamine, and heparin/protamine reversal on platelet function under conditions of arterial shear stress. Anesth Analg 2001; 93: 2027. Lindblad B, Wakeeld TW, Whitehouse WM Jr et al. The effect of protamine sulfate on platelet function. Scand J Thorac Cardiovasc Surg 1988; 22: 5559. Despotis GJ, Levine V, Filos KS et al. Hemoltration during cardiopulmonary bypass: the effect on anti-Xa and anti-IIa heparin activity. Anesth Analg 1997; 84: 479483. Trossaert M, Gaillard A, Commin PL et al. High incidence of anti-heparin/platelet factor 4 antibodies after cardiopulmon-

63

64

65

66

67

68

69

70

71

72

73

ary bypass surgery. Br J Haematol 1998; 101: 653655. Mullen MP, Wessel DL, Thomas KC et al. The incidence and implications of anti-heparin-platelet factor 4 antibody formation in a pediatric cardiac surgical population. Anesth Analg 2008; 107: 371 378. Boning A, Morschheuser T, Blase U et al. Incidence of heparin-induced thrombocytopenia and therapeutic strategies in pediatric cardiac surgery. Ann Thorac Surg 2005; 79: 6265. Alsou B, Boshkov LK, Kirby A et al. Heparin-induced thrombocytopenia (HIT) in pediatric cardiac surgery: an emerging cause of morbidity and mortality. Semin Thorac Cardiovasc Surg Pediatr Card Surg Annu 2004; 7: 155171. Koster A, Crystal GJ, Kuppe H et al. Acute heparin-induced thrombocytopenia type II during cardiopulmonary bypass. J Cardiothorac Vasc Anesth 2000; 14: 300 303. Warkentin TE, Greinacher A, Koster A et al. Treatment and prevention of heparin-induced thrombocytopenia: American College of Chest Physicians EvidenceBased Clinical Practice Guidelines (8th Edition). Chest 2008; 133: 340S380S. Riess F, Potzsch B, Behr I et al. Recombinant hirudin as an anticoagulant during cardiac operations: experiments in a pig model. Eur J Cardiothorac Surg 1997; 11: 739745. Gates R, Yost P, Parker B. The Use of Bivalirudin for Cardiopulmonary Bypass Anticoagulation in Pediatric HeparinInduced Thrombocytopenia Patients. Artif Organs 2010; 34: 667669. Ciccolo ML, Bernstein J, Collazos JC et al. Argatroban anticoagulation for cardiac surgery with cardiopulmonary bypass in an infant with double outlet right ventricle and a history of heparin-induced thrombocytopenia. Congenit Heart Dis 2008; 3: 299302. Riess FC, Poetzsch B, Madlener K et al. Recombinant hirudin for cardiopulmonary bypass anticoagulation: a randomized, prospective, and heparin-controlled pilot study. Thorac Cardiovasc Surg 2007; 55: 233238. Follis F, Filippone G, Montalbano G et al. Argatroban as a substitute of heparin during cardiopulmonary bypass: a safe alternative? Interact Cardiovasc Thorac Surg 2010; 10: 592596. Dyke CM, Smedira NG, Koster A et al. A comparison of bivalirudin to heparin with protamine reversal in patients undergoing cardiac surgery with cardiopulmonary bypass: the EVOLUTION-ON study. J

40

Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

M.P. Eaton and E.M. Iannoli

Coagulation in children on bypass

74

75

76

77

78

79

80

81

82

83

84

85

86

Thorac Cardiovasc Surg 2006; 131: 533 539. Kurup V, Transue S, Wu Y et al. Cardiac surgery in a patient with heparininduced thrombocytopeniacautions with use of the direct thrombin inhibitor, argatroban. Conn Med 2006; 70: 245250. Gasparovic H, Nathan NS, Fitzgerald D et al. Severe argatroban-induced coagulopathy in a patient with a history of heparin-induced thrombocytopenia. Ann Thorac Surg 2004; 78: e89e91. Welsby IJ, Jones WL, Arepally G et al. Effect of combined anticoagulation using heparin and bivalirudin on the hemostatic and inammatory responses to cardiopulmonary bypass in the rat. Anesthesiology 2007; 106: 295301. Paramo JA, Rifon J, Llorens R et al. Intra- and postoperative brinolysis in patients undergoing cardiopulmonary bypass surgery. Haemostasis 1991; 21: 58 64. Ray MJ, Marsh NA, Hawson GA. Relationship of brinolysis and platelet function to bleeding after cardiopulmonary bypass. Blood Coagul Fibrinolysis 1994; 5: 679685. Jaggers JJ, Neal MC, Smith PK et al. Infant cardiopulmonary bypass: a procoagulant state. Ann Thorac Surg 1999; 68: 513520. Miller BE, Guzzetta NA, Tosone SR et al. Rapid evaluation of coagulopathies after cardiopulmonary bypass in children using modied thromboelastography. Anesth Analg 2000; 90: 13241330. Eaton MP. Antibrinolytic therapy in surgery for congenital heart disease. Anesth Analg 2008; 106: 10871100. Karkouti K, Beattie WS, Dattilo KM et al. A propensity score case-control comparison of aprotinin and tranexamic acid in high-transfusion-risk cardiac surgery. Transfusion 2006; 46: 327338. Mangano DT, Tudor IC, Dietzel C. The risk associated with aprotinin in cardiac surgery. N Engl J Med 2006; 354: 353365. Fergusson DA, Hebert PC, Mazer CD et al. A comparison of aprotinin and lysine analogues in high-risk cardiac surgery. N Engl J Med 2008; 358: 23192331. Van der Staak FH, de Haan AF, Geven WB et al. Surgical repair of congenital diaphragmatic hernia during extracorporeal membrane oxygenation: hemorrhagic complications and the effect of tranexamic acid. J Pediatr Surg 1997; 32: 594599. Bulutcu FS, Ozbek U, Polat B et al. Which may be effective to reduce blood loss after cardiac operations in cyanotic children: tranexamic acid, aprotinin or a combination? Paediatr Anaesth 2005; 15: 4146.

87 Chauhan S, Bisoi A, Kumar N et al. Dose comparison of tranexamic acid in pediatric cardiac surgery. Asian Cardiovasc Thorac Ann 2004; 12: 121124. 88 Chauhan S, Bisoi A, Modi R et al. Tranexamic acid in paediatric cardiac surgery. Indian J Med Res 2003; 118: 8689. 89 Chauhan S, Das SN, Bisoi A et al. Comparison of epsilon aminocaproic acid and tranexamic acid in pediatric cardiac surgery. J Cardiothorac Vasc Anesth 2004; 18: 141143. 90 Chauhan S, Kumar BA, Rao BH et al. Efcacy of aprotinin, epsilon aminocaproic acid, or combination in cyanotic heart disease. Ann Thorac Surg 2000; 70: 1308 1312. 91 Rao BH, Saxena N, Chauhan S et al. Epsilon aminocaproic acid in paediatric cardiac surgery to reduce postoperative blood loss. Indian J Med Res 2000; 111: 5761. 92 Zonis Z, Seear M, Reichert C et al. The effect of preoperative tranexamic acid on blood loss after cardiac operations in children. J Thorac Cardiovasc Surg 1996; 111: 982987. 93 McClure PD, Izsak J. The use of epsilonaminocaproic acid to reduce bleeding during cardiac bypass in children with congenital heart disease. Anesthesiology 1974; 40: 604608. 94 Williams GD, Bratton SL, Riley EC et al. Efcacy of epsilon-aminocaproic acid in children undergoing cardiac surgery. J Cardiothorac Vasc Anesth 1999; 13: 304 308. 95 Ririe DG, James RL, OBrien JJ et al. The pharmacokinetics of epsilon-aminocaproic acid in children undergoing surgical repair of congenital heart defects. Anesth Analg 2002; 94: 4449. 96 Eaton MPKK, Cable R, Haas CE et al. The pharmacokinetics of epsilon-aminocaproic acid in neonates undergoing cardiac surgery. SCA 32nd annual meeting. New Orleans, 2010. 97 Yurka HG, Wissler RN, Zanghi CN et al. The Effective Concentration of EpsilonAminocaproic Acid for Inhibition of Fibrinolysis in Neonatal Plasma in Vitro. Anesth Analg 2010; 111: 180184. 98 Rinder CS. Hematologic effects and coagulopathy. In: Gravlee GP, Davis RF, Stammers AH et al., eds. Cardiopulmonary Bypass, Principles and Practice. Philadelphia: Wolters Kluwer, 2008: 439458. 99 Suzuki Y, Malekan R, Hanson CW III et al. Platelet anesthesia with nitric oxide with or without eptibatide during cardiopulmonary bypass in baboons. J Thorac Cardiovasc Surg 1999; 117: 987993.

100 Koster A, Chew DP, Kuebler W et al. Effects of tiroban on hemostatic activation and inammatory response during cardiopulmonary bypass. Am J Cardiol 2003; 91: 346347. 101 Tabata S, Yamaguchi S, Nagamine H et al. Efcacy of FK633, an ultra-short acting glycoprotein IIb/IIIa antagonist on platelet preservation during and after cardiopulmonary bypass. Eur J Cardiothorac Surg 2004; 26: 289293. 102 Kastrati A, Ndrepepa G. Cangrelor - a champion lost in translation? N Engl J Med 2009; 361: 23822384. 103 Manno CS, Hedberg KW, Kim HC et al. Comparison of the hemostatic effects of fresh whole blood, stored whole blood, and components after open heart surgery in children. Blood 1991; 77: 930936. 104 Mou SS, Giroir BP, Molitor-Kirsch EA et al. Fresh whole blood versus reconstituted blood for pump priming in heart surgery in infants. N Engl J Med 2004; 351: 16351644. 105 Gruenwald CE, McCrindle BW, CrawfordLean L et al. Reconstituted fresh whole blood improves clinical outcomes compared with stored component blood therapy for neonates undergoing cardiopulmonary bypass for cardiac surgery: a randomized controlled trial. J Thorac Cardiovasc Surg 2008; 136: 1442 1449. 106 Koch CG, Li L, Sessler DI et al. Duration of red-cell storage and complications after cardiac surgery. N Engl J Med 2008; 358: 12291239. 107 Ranucci M, Carlucci C, Isgro G et al. Duration of red blood cell storage and outcomes in pediatric cardiac surgery: an association found for pump prime blood. Crit Care 2009; 13: R207. 108 Miyaji K, Kohira S, Miyamoto T et al. Pediatric cardiac surgery without homologous blood transfusion, using a miniaturized bypass system in infants with lower body weight. J Thorac Cardiovasc Surg 2007; 134: 284289. 109 Charette K, Hirata Y, Bograd A et al. 180 ml and less: cardiopulmonary bypass techniques to minimize hemodilution for neonates and small infants. Perfusion 2007; 22: 327331. 110 Hickey E, Karamlou T, You J et al. Effects of circuit miniaturization in reducing inammatory response to infant cardiopulmonary bypass by elimination of allogeneic blood products. Ann Thorac Surg 2006; 81: S2367S2372. 111 Gomez D, Olshove VF, Weinstein S et al. Use of a low-prime circuit for bloodless heart transplantation in xenotransplant of

Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

41

Coagulation in children on bypass

M.P. Eaton and E.M. Iannoli

112

113

114

115

116

117

118

119

120

121

5-7 kilogram primates. J Extra Corpor Technol 2000; 32: 138141. Gomez D, Preston TJ, Olshove VF et al. Evaluation of air handling in a new generation neonatal oxygenator with integral arterial lter. Perfusion 2009; 24: 107112. Olsson C, Siegbahn A, Henze A et al. Heparin-coated cardiopulmonary bypass circuits reduce circulating complement factors and interleukin-6 in paediatric heart surgery. Scand Cardiovasc J 2000; 34: 33 40. Jensen E, Andreasson S, Bengtsson A et al. Inuence of two different perfusion systems on inammatory response in pediatric heart surgery. Ann Thorac Surg 2003; 75: 919925. Suzuki Y, Daitoku K, Minakawa M et al. Poly-2-methoxyethylacrylate-coated bypass circuits reduce activation of coagulation system and inammatory response in congenital cardiac surgery. J Artif Organs 2008; 11: 111116. Deptula J, Glogowski K, Merrigan K et al. Evaluation of biocompatible cardiopulmonary bypass circuit use during pediatric open heart surgery. J Extra Corpor Technol 2006; 38: 2226. Miyaji K, Hannan RL, Ojito J et al. Heparin-coated cardiopulmonary bypass circuit: clinical effects in pediatric cardiac surgery. J Card Surg 2000; 15: 194198. Petaja J, Lundstrom U, Leijala M et al. Bleeding and use of blood products after heart operations in infants. J Thorac Cardiovasc Surg 1995; 109: 524529. Miller BE, Mochizuki T, Levy JH et al. Predicting and treating coagulopathies after cardiopulmonary bypass in children. Anesth Analg 1997; 85: 11961202. Nuttall GA, Oliver WC, Santrach PJ et al. Efcacy of a simple intraoperative transfusion algorithm for nonerythrocyte component utilization after cardiopulmonary bypass. Anesthesiology 2001; 94: 773 781. Discussion 775A776A. Shore-Lesserson L, Manspeizer HE, DePerio M et al. Thromboelastographyguided transfusion algorithm reduces transfusions in complex cardiac surgery. Anesth Analg 1999; 88: 312319.

122 Avidan MS, Alcock EL, Da Fonseca J et al. Comparison of structured use of routine laboratory tests or near-patient assessment with clinical judgement in the management of bleeding after cardiac surgery. Br J Anaesth 2004; 92: 178186. 123 Enriquez LJ, Shore-Lesserson L. Point-ofcare coagulation testing and transfusion algorithms. Br J Anaesth 2009; 103(Suppl 1): i14i22. 124 Moganasundram S, Hunt BJ, Sykes K et al. The relationship among thromboelastography, hemostatic variables, and bleeding after cardiopulmonary bypass surgery in children. Anesth Analg 2010; 110: 995 1002. 125 Patanwala AE. Factor VIIa (recombinant) for acute traumatic hemorrhage. Am J Health Syst Pharm 2008; 65: 16161623. 126 Despotis G, Avidan M, Lublin DM. Offlabel use of recombinant factor VIIA concentrates after cardiac surgery. Ann Thorac Surg 2005; 80: 35. 127 Warren O, Mandal K, Hadjianastassiou V et al. Recombinant activated factor VII in cardiac surgery: a systematic review. Ann Thorac Surg 2007; 83: 707714. 128 Agarwal HS, Bennett JE, Churchwell KB et al. Recombinant factor seven therapy for postoperative bleeding in neonatal and pediatric cardiac surgery. Ann Thorac Surg 2007; 84: 161168. 129 Egan JR, Lammi A, Schell DN et al. Recombinant activated factor VII in paediatric cardiac surgery. Intensive Care Med 2004; 30: 682685. 130 Pychynska-Pokorska M, Moll JJ, Krajewski W et al. The use of recombinant coagulation factor VIIa in uncontrolled postoperative bleeding in children undergoing cardiac surgery with cardiopulmonary bypass. Pediatr Crit Care Med 2004; 5: 246250. 131 Tobias JD, Berkenbosch JW, Russo P. Recombinant factor VIIa to treat bleeding after cardiac surgery in an infant. Pediatr Crit Care Med 2003; 4: 4951. 132 Leibovitch L, Kenet G, Mazor K et al. Recombinant activated factor VII for lifethreatening pulmonary hemorrhage after

133

134

135

136

137

138

139

140

pediatric cardiac surgery. Pediatr Crit Care Med 2003; 4: 444446. Niles SD, Burkhart HM, Duffey DA et al. Use of recombinant factor VIIa (NovoSeven) in pediatric cardiac surgery. J Extra Corpor Technol 2008; 40: 241248. Razon Y, Erez E, Vidne B et al. Recombinant factor VIIa (NovoSeven) as a hemostatic agent after surgery for congenital heart disease. Paediatr Anaesth 2005; 15: 235240. Veldman A, Neuhaeuser C, Akintuerk H et al. rFVIIa in the treatment of persistent hemorrhage in pediatric patients on ECMO following surgery for congenital heart disease. Paediatr Anaesth 2007; 17: 11761181. Wittenstein B, Ng C, Ravn H et al. Recombinant factor VII for severe bleeding during extracorporeal membrane oxygenation following open heart surgery. Pediatr Crit Care Med 2005; 6: 473476. Guzzetta NA, Huch S, Fernandez JD et al. Use of recombinant factor VIIa for uncontrolled bleeding in neonates after cardiopulmonary bypass. Paediatr Anaesth 2009; 19: 364370. Karsies TJ, Nicol KK, Galantowicz ME et al. Thrombotic risk of recombinant factor seven in pediatric cardiac surgery: a single institution experience. Ann Thorac Surg 2010; 89: 570576. Ekert H, Brizard C, Eyers R et al. Elective administration in infants of low-dose recombinant activated factor VII (rFVIIa) in cardiopulmonary bypass surgery for congenital heart disease does not shorten time to chest closure or reduce blood loss and need for transfusions: a randomized, double-blind, parallel group, placebo-controlled study of rFVIIa and standard haemostatic replacement therapy versus standard haemostatic replacement therapy. Blood Coagul Fibrinolysis 2006; 17: 389 395. OConnell KA, Wood JJ, Wise RP et al. Thromboembolic adverse events after use of recombinant human coagulation factor VIIa. JAMA 2006; 295: 293298.

42

Pediatric Anesthesia 21 (2011) 3142 2010 Blackwell Publishing Ltd

S-ar putea să vă placă și