GET THE APP

Monitoring of Haemostasis and Anticoagulation in Cardiopulmonary
Journal of Hematology & Thromboembolic Diseases

Journal of Hematology & Thromboembolic Diseases
Open Access

ISSN: 2329-8790

+44 1478 350008

Review Article - (2014) Volume 2, Issue 6

Monitoring of Haemostasis and Anticoagulation in Cardiopulmonary Bypass Patients

Yvonne P.J. Bosch1,2, Patrick W. Weerwind1,2, Baheramsjah Mochtar1,2 and Raed Al Dieri2*
1Department of Cardiothoracic Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
2Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre+, Maastricht, The Netherlands
*Corresponding Author: Raed Al Dieri, P. Debeyelaan 25, PO Box 5800, 6202 AZ Maastricht, The Netherlands, Tel: +31(0)43-3877069, Fax: +31(0)43-3875075 Email:

Abstract

Patients undergoing cardiac surgery with cardiopulmonary bypass are at risk of increased blood loss and transfusion requirements. Contact of blood with the surgical wound and the artificial surfaces of the extra-corporeal circuit results in a coagulopathy. Haemodilution during cardiopulmonary bypass also aggravates the reduction of haemostatic factors even further. On the other hand, systemic anticoagulation using high dose unfractionated heparin challenges the adequate anticoagulant monitoring. The most frequently used test to monitor systemic anticoagulation is the whole blood activated clotting time. This has, however, proven not be a reliable test. Other haemostatic assays have their own advantages and disadvantages, but tools to monitor anticoagulation and haemostasis adequately during and after cardiopulmonary bypass are to date lacking and/or show only a weak association with clinical bleeding. Viscoelastic whole blood techniques (TEG/ROTEM) have emerged as alternatives to the routine coagulation tests, and could be more suitable for providing information about the different phases of the blood clotting. Platelet function tests give more information about the presence of platelet dysfunction one, which is a major cause of bleeding after cardiac surgery. Likewise, thrombin generation tests reflect much, if not all, of the overall function of the blood clotting system. Further research of the applicability and added values of the viscoelastic whole blood tests and the thrombin generation tests in the cardiac surgical patient undergoing cardiopulmonary bypass is warranted. Simplicity in performing the test, preferably point-of-care, and rapid availability of the results, should thereby be the primary focus for targeted haemostatic interventions.

Keywords: Cardiopulmonary Bypass; Anticoagulation Monitoring; Blood Loss; Transfusion Requirements; Activated Clotting Time; Thromboelastometry; Platelet Function; Calibrated Automated Thrombography

Abbreviations

ACT: Activated Clotting Time; ETP: Endogenous Thrombin Potential; aPTT: Activated Partial Thromboplastin Time; MCF: Maximum Clot Firmness; AT: Antithrombin; PT: Prothrombin Time; CAT: Calibrated Automated Thrombography; ROTEM: Rotational Thromboelastometry; CFT: Clot Formation Time; TEG: Thromboelastography; CPB: Cardiopulmonary Bypass; TFPI: Tissue Factor Pathway Inhibitor; CT: Clotting Time; TG: Thrombin Generation

Cardiac Surgery, Cardiopulmonary Bypass, and Coagulopathy

During cardiopulmonary bypass (CPB) for cardiac surgery, contact of blood with the surgical wound and the artificial surfaces of the extra-corporeal circuit leads to haemostatic disturbances and triggers an angry defense reaction [1,2]. The resulting coagulopathy is caused by several factors: platelet dysfunction, low preoperative fibrinogen levels, hyperfibrinolysis, and continually produced thrombin [3-5]. In addition, haemodilution plays an important role. Patients undergoing CPB procedures are usually infused with crystalloids or colloids, used as pump prime and intra-venous fluids. These infusions lead to a state of dilution coagulopathy while there is a significant fall in clotting factors [3]. Secondly, excessive dilution could lead to an overestimation of the heparin effect, and a resulting overestimation of the required protamine neutralizing dose [6]. All these factors are predictive for an increased risk of excessive perioperative blood loss and subsequent transfusion requirements [7,8]. Other clinical consequences of coagulopathy are: mediastinal re-exploration, postoperative stroke due to hypercoagulability and increased mortality [2,9]. Furthermore, intensive care stay is prolonged and hospital costs are increased [7]. Haemostatic activation seems to be unavoidable in patients undergoing CPB, thereby upsetting their postoperative recovery.

Heparin Anticoagulation

Cardiac surgical patients undergoing CPB are typically anticoagulated using a very high dose of unfractionated heparin. This is the only anticoagulant that brings about an immediate effect and, that can be monitored bedside by a point-of-care test. Moreover, unfractionated heparin is easily neutralized by protamine at the end of the procedure [10]. The primary anticoagulant action of heparin appears to result from its binding with antithrombin (AT). The predominant action of AT is inactivation of factor IIa (thrombin) and factor Xa [11]. Following activation of AT, heparin is released and becomes available for interaction with other AT molecules. In contrast, AT is consumed and AT levels are progressively depleted [11]. Therefore, decreased heparin responsiveness is often attributed to AT deficiency. Besides binding with AT, heparin may bind thrombin directly or act through heparin-cofactor II. Heparin also exerts an anticoagulant effect by augmenting the activity of tissue factor pathway inhibitor (TFPI) [11]. Measuring the real anticoagulant effect of heparin is complicated and thus depends on many variables which determine the patients sensitivity to heparin, such as: 1) the availability of AT; 2) the binding of heparin to proteins (such as albumin and glycosamineglycans) in the bloodstream [12]; 3) the binding of heparin to endothelium [11]; and 4) platelets: both the release of platelet factor 4 and platelets themselves are heparin neutralizing [13].

Monitoring of Anticoagulation in Cardiac Surgery

Several laboratory methods are currently available for monitoring haemostasis and anticoagulation during cardiac surgery. Table 1 shows an overview of available haemostatic tests. Advantages and disadvantages are also listed.

Test Clinical indication Outcome P/WB Pros Cons
Clotting times  
ACT (activated clotting time) To guide heparinization of CPB patients; to guide heparin neutralization with protamine sulphate as patients come off CPB Time between activation of FXII by kaolin on the one hand, and the first bit of thrombin and/or fibrin at the other hand. WB Point-of-care in operating room; simple, safe, and cost-effective ACT-based protocols do not avoid thrombin generation during CPB; ACT values during CPB do not correlate with heparin concentrations as a result of hypothermia and haemodilution; return of the ACT to baseline is not an absolute validation of complete heparin neutralization; ACT is technique-dependent
aPTT
(activated partial thrombo-plastin time)
To screen overall
haemostasis, mainly the intrinsic pathway, to a lesser extent the com-mon pathway; to guide heparin dosing; also used to monitor hirudin, or argatroban
Time between activation of FXII by kaolin on the one hand, and the first bit of thrombin and/or fibrin at the other hand (intrinsic). P Available almost 24 h/day in almost all hospital laboratories; cost-effective High blood concentrations (over 1 IU/mL) cannot be accurately evaluated by aPTT; does not detect small variations in coagulation factors; not suitable to detect hypercoagulability; Gives only information about a small part of haemostasis
PT / INR
(prothrombin time / international normalized ratio)
To screen the integrity of the extrinsic (factor VII) and common (fibrinogen and factors II, V, and X) pathways of coagulation; to monitor warfarin anticoagulation Time between activation of FVII by tissue factor on the one hand, and first bit of thrombin/fibrin at the other hand (extrinsic). P Available almost 24 h/day in almost all hospital laboratories; cost-effective Does not detect small variations in coagulation factors; not suitable to detect hypercoagulability; Gives only information about a small part of haemostasis
Heparin concentration  
Hepcon HMS To establish whole blood heparin concen-tration using heparin–protamine titration Whole blood heparin level WB Point-of-care in operating room; simple, safe, and cost-effective May over-estimate the heparin dose; inability to evaluate the anticoagulant properties of heparin, or to detect heparin resistance.
Anti-Xa assay To monitor heparin therapy; a supplemental means to confirm adequate anticoagulation Heparin concentration P The anti-Xa assay is not susceptible to interference from elevated concen- trations of factor VIII or fibrinogen that result from acute phase reactions;
The anti-Xa assay is not influenced by factor deficiencies, with the possible exception of AT deficiency
The anti-Factor Xa level may be a surrogate mea- surement of the overall anticoagulant activity of unfractionated heparin; An anti-Factor Xa level only assesses how much exogenous factor Xa is inhibited by the patient’s plasma; the presence of a specialized coagulation technologist is necessary
Blood factors and platelets        
Fibrinogen To measure a haemo-static level (interpreted in the light of other tests like PT/ aPTT) Fibrinogen level P Suggested high predictive value: low fibrinogen levels can be associated with bleeding Gives only information about a small part of haemostasis; Test is time consuming; High concen- trations of unfractionated heparin (>0.8 IU/ml) may lead to an underestimate of the true fibrinogen level
Platelet count To measure a haemostatic level Platelet count WB Simple, safe, and cost-effective Only suitable to detect crude abnormalities in haemostasis; not suitable to detect hypercoagulability; provides no information on platelet functionality and thus not highly reflective for bleeding
D-dimers To detect hyperfibrino-lysis / hypercoagulability D-dimer level P High sensitivity Limited specificity; reflects only break down of fibrin at a specific moment, but does not reflect on-going coagulation activity; data obtained by different D-dimer assays are not interchangeable
Platelet function tests  
Multiplate To determine platelet function Platelet function WB Studies platelet function within anticoagulated whole blood without any sample processing; is sensitive for the monitoring of platelet function inhibitors; is sensitive for von Willebrand Disease Highly dependent on platelet number and sample quality; does not simulate physiological primary haemostasis; abnormal results can easily be induced by antibiotics, some vitamins, spices and foods
PFA-100 To detect pharma- cologic platelet dysfunction; may be able to accurately detect hypercoagulability; to identify von Willebrand’s disease, and other acquired and congenital platelet defects Whole blood platelet function by measuring the closure time (CT) WB Simulates in vivo conditions by measuring platelet adhesion and aggregation under conditions of high shear); sensitive for von Willebrand disease and moderate to- severe platelet abnormalities; High negative predictive value; Relatively insensitive to clotting factor deficiencies Low positive predictive value and thus not very useful in transfusion algorithms to direct transfusion therapy; less sensitive to the effect of anti-platelet medications; Closure times increase progressively with decreases in haematocrit and falls in platelet count below 100 x 109/L
Platelet Mapping To monitor the effect of clopidogrel and aspirin on platelets Platelet function WB It has the potential to evaluate the
function of different platelet receptors in the presence of coagulation factors and fibrinogen mimicking conditions
It is not a true point-of-care instrument as it requires pipetting
Visco elastic tests  
TEG To assess the global haemostatic function from a single blood sample; allow for diagnosis of hyperfibrinolysis in bleeding patients Reflections of clotting time, clot strength, degree of fibrinolysis, influence of fibrinogen WB Is assessed in whole blood, allowing in vivo coagulation system interactions with platelets and red blood cells; reflects fibrinogen levels; clot development can be visually displayed in real-time; can identify if bleeding is due to residual heparin Although physiological clot development is better depicted by analysing whole blood, these techniques measure haemostasis under static conditions in vitro, and results must be carefully interpreted; mainly dependent on fibrinogen levels and no information about thrombin generation
ROTEM To assess the global haemostatic function from a single blood sample; allow for diagnosis of hyperfibrinolysis in bleeding patients Reflections of clotting time, clot strength, degree of fibrinolysis, influence of fibrinogen WB Is assessed in whole blood, allowing in vivo interactions with platelets and red blood cells; fibrin-specific clot formation (FIBTEM) can be demonstrated; clot development can be visually displayed in real-time; can identify if bleeding is due to residual heparin Inability to detect impairment in platelet function induced by anti-platelet agents; although physiological clot development is better depicted by analysing whole blood, these techniques measure haemostasis under static conditions in vitro, and results must be carefully interpreted; mainly dependent on fibrinogen levels and no information about thrombin generation
Thrombin generation  
CAT To detect bleeding tendency and hypercoagulability; to monitor anticoagulants and procoagulant factors Amount of thrombin formed by plasma after activation of coagulation in time, reflecting total haemostatic capacity P/WB Thrombin generation depends on almost all coagulation factors and anticoagulant proteins; it detects changes in platelet function by changing the sensitivity of the test –and adjust this for the target group-by varying the concentration of added tissue factor. Lack of clinical and animal studies; not well validated; standardization is necessary before application in laboratories or operation room can take place.

Table 1: Overview of the tests used for monitoring haemostasis and anticoagulation in cardiopulmonary bypass patients, P= Plasma, WB= whole blood.

Determination of Heparin Concentration

Heparinization is currently monitored by either measuring heparin concentration or by measuring its effect on clotting times. Chromogenic tests for measuring heparin concentration are less sensitive to interfering substances than the clotting tests are. Plasma heparin concentration is determined via its accelerating function in the inhibition of factor Xa or thrombin by AT. Although thrombin based tests are possible, the majority of the commercial heparin assays are performed using anti-factor Xa-activity (anti-Xa), in the haematology laboratory. There are two different designs of the anti-Xa assay. Some tests use a saturating concentration of AT in the reagent. This test determines the ‘total heparin’. The other design does not add AT and relies on the AT in the sample. This determines the AT/heparin complex (or the ‘active heparin’) [14]. Heparin concentration can also be measured bedside by a haemostasis management system (Hepcon HMS, Medtronic, Minneapolis MN). This is a multichannel clot-timing instrument that uses the principle of heparin-protamine titration to quantitatively determine the heparin concentration in whole blood. In each channel, a plastic rod is rapidly lifted and dropped by an oscillating metal bar. Whole blood is automatically placed into each channel by a 3 ml syringe inserted into the machine. The time for blood to clot is displayed for each channel [15]. Six levels of heparin can be determined by using discrete cut offs [16]. Excess protamine inhibits clot formation and insufficient protamine does not fully antagonize heparin, therefore the channel with the fastest clot formation will represent the protamine concentration that optimally neutralizes the existing heparin [17]. The imprecision of the Hepcon HMS could potentially be caused by the effects of release of TFPI by heparin in vivo but not ex vivo. In the absence of an endothelial source of TFPI, the contribution of TFPI to heparin responsiveness may be underestimated by the Hepcon HMS [16]. Heparin concentration monitoring has not uniformly been associated with less bleeding, as it may be associated with heparin rebound postoperatively [17]. Between the anti-Xa assay and the HMS assay there is a lack of agreement [18]. It can be postulated that the HMS assay is affected by factors like haemodilution, hypothermia and platelet dysfunction [18], while the anti-Xa assay only reflects the heparin concentration that inhibits factor Xa. The fraction of heparin that is responsible for anti-factor IIa activity is not measured, meaning that the anti-Xa assay will not give a real and complete reflection of heparin concentration [19]. Thus, monitoring of heparin concentration is only a surrogate for monitoring of anticoagulation, since there are important variations in the response of patients to heparin [20].

Clotting-Time Based Methods

Although routine preoperative coagulation testing contributes little to patient care, this is justified in case of cardiac operations with CPB since patients will undergo an operation with major effects on haemostasis and many will require blood transfusions [21]. Prothrombin time (PT) and activated partial thromboplastin time (aPTT), performed in plasma, are by far the most common screening tests for coagulation abnormalities. These tests correspond respectively to the extrinsic and the intrinsic pathways of coagulation. PT monitoring is widely used as a preoperative screening test for patients receiving oral anticoagulation. The sensitivity for prothrombin and fibrinogen is less pronounced than for other factors. The international normalized ratio (INR) is the ratio of the PT of the patient and the normal PT standardized with the international sensitivity index (ISI). This is to correct for specific laboratory methods. The aPTT is used to guide unfractionated heparin (< 1 IU/ml) clinically. After heparin neutralizing, the aPTT could be measured to control heparin rebound. The PT and aPTT do not reflect the complexity of haemostasis in vivo. One disadvantage of the PT and aPTT is the impossibility to provide information about the interaction with platelets since it is measured in platelet poor plasma. Consequently, these tests will not reflect the extent of bleeding, as platelet count and function may vary [22]. It was clear that even though deficiencies of each of the factors in the intrinsic pathway could have equally long aPTT values, they had dramatically different risks of hemorrhage [23]. Hence the PT and aPTT, providing information on the ability to clot, are poor predictors of bleeding despite their widespread use [24-27]. Another disadvantage is that the PT and aPTT remain normal under hyperfibrinolytic conditions, meaning that bleeding caused by increased fibrin breakdown will not be detected [22]. Although there are many available coagulation assays, it is rather complicated to find the ideal test reflecting the clinical situation of the CPB patient.

During cardiac surgery the prolongation of the clotting time as a reflection of the anticoagulant status is measured. This is performed by the point-of-care activated clotting time (ACT). The principle of the ACT is adding whole blood to a tube containing a contact activator, such as kaolin, celite or silica. Clotting activation and fibrin formation will be initiated and detected. The ACT was first described by Hattersley in 1966 [28], and introduced into cardiac surgery by Bull in the 70’s [29]. The safe minimal level of the ACT was first proposed at 300 s. However, this was stepwise increased to 480 s, after observing visible clots in the extracorporeal circuit and because of a three to six fold variation in heparin effect and a fourfold variation in heparin-life. The most important cofactor that heparin requires for its anticoagulant effect is AT. Therefore, it is surprising that there is no relation between the ACT and AT level [30-32]. In the early 90’s variability between the different systems was noted, i.e. ACT analyzers differ by activators, detection systems, and use different sample volumes [33-35]. Consequently, the ACT value differs for each analyzer, suggesting that extrapolation between different systems would not be appropriate [36]. The ACT is also influenced by factors like the balance of coagulation factors and inhibitors, fibrinogen, platelets, plasma proteins which bind heparin, TFPI release from vasculature, increased haemodilution, and hypothermia of the patient. Nowadays, growing evidence suggests that coagulation is increasingly activated with time during CPB, even with ACT-based protocols [37]. This might lead to a progressive consumptive coagulopathy resulting in a prolonged ACT although heparin levels are decreased, resulting in an incorrect estimation of the anticoagulation status [38]. As a result, thrombin might be only partially suppressed and microscopic emboli are produced [2]. Further, the majority of thrombin generation is triggered via factor VIIa and tissue factor, whereas the ACT reflects inhibition of contact activation, which is not indicative for the amount and time of thrombin generation [39]. Additionally, it is questionable whether adequate heparin neutralization will be achieved postoperatively, since ACT is poorly sensitive to low heparin concentrations. This could result in over dosage of protamine, or incomplete heparin reversal [33]. Measuring the anticoagulant effect of heparin with the ACT, may have little or no relationship to what is clinically happening [11]. Therefore, when monitoring anticoagulation exclusively with the ACT, there are a lot of pitfalls to overcome.

Monitoring of Haemostasis in Cardiac Surgery

The complex process of haemostasis can be broken down into four components: platelet activation, thrombin generation, clot formation/stabilization, and fibrinolysis [40]. To get a better understanding of these processes during and after CPB, it would be desirable to have a coagulation test that provides information about all these phases. However, such a tool to monitor haemostasis adequately is lacking [2,8,41,42]. Standard coagulation tests do not reflect the activity of both the coagulation factors and the natural anticoagulant proteins, and the interaction with platelets [8,22]. In addition, tests are often time consuming and the results not rapidly available, so they do not reflect the current state of the coagulation system and could lead to inappropriate treatment [43]. At present a combination of haemostatic assays may provide more effective diagnostic information than the use of any single test [44], because the current tests are on its own often limited in utility.

Standard Haemostatic Assays

Platelet count is only a quantitative measurement and, does not give any information about the platelet functionality, therefore it is less reflective for bleeding [45]. However, in studies by Karkouti et al. [46] and Coakley et al. [47] it was shown that loss of platelets reflected by both pre- and post- CPB platelet count was associated with increased blood loss. Fibrinogen on the other hand is only a single factor, excluding the influence of other blood cells. Large decreases in fibrinogen and subsequent low fibrinogen levels both pre- and post- CPB, are shown to be predictive for increasing blood loss [46,48]. Though, Coakley et al. did not find postoperative measured fibrinogen to be associated with blood loss. Similarly, low plasma concentrations of fibrinogen were not significantly associated with increased homologous blood usage [49].

D-dimer levels are split products of F-XIII crosslinked fibrin and are determined with monoclonal antibody assays. An increase of D-dimer over the baseline value indicates excessive fibrin formation, stabilization by F-XIIIa and subsequent fibrinolysis [50], that is measured at a specific moment, but which does not reflect on-going coagulation activity. D-dimer levels do increase during cardiac surgery, but there is no association with blood loss [51]. Kuepper et al. concluded that the D-dimer assay has a relatively high negative predictive value (measured 15 minutes after protamine administration), which might be useful to identify patients who are unlikely to have an excessive postoperative bleeding [50]. This could be helpful to avoid unnecessary transfusions after cardiac surgery. One should be critical to interpret the results of D-dimer assays, because there is a known variability among different assays due to heterogeneity of fibrin degradation products in patient samples, reactivity of antibodies, different calibrators and different assay formats [50].

Viscoelastic Whole Blood Techniques

Viscoelastic whole blood techniques seem to be more suitable for reflecting the complex coagulation system. Thromboelastography (TEG) was first described by Hartert in 1948 [52]. Using this assay, whole blood is added to a cup in which a pin is suspended connected to a detector system. The cup initiates the movement and the viscoelastic changes that occur during coagulation are measured and recorded in a graphical representation of the fibrin polymerization process. This technique was very sensitive to vibrations with the resultant unstable reproducibility. Over the years, the technique was refined, and nowadays there are two systems available: the TEG (Haemoscope Corporation, IL, USA) using the original principle, and the rotational thromboelastometry (ROTEM, TEM International GmbH, Munich, Germany), which uses a modified principle where the pin inside a standing cuvette is moving. The TEG device has two independent measuring channels, which includes a platelet mapping assay. The latter provides information about a patients’ response to anti-platelet therapy. ROTEM is also performed in whole blood, and it provides therefore information on the contribution of fibrinogen and platelets to clot formation. The rate of fibrin polymerization as well as the overall clot strength is assessed. During the clotting process (conversion of fibrinogen to fibrin), the clot increasingly restricts the rotation of the pin with rising clot firmness. This is detected and converted in a graph of clot formation and subsequent lysis. ROTEM is suitable for a more timely assessment of fibrinogen levels (<15-20 minutes). The following different measurements can be performed in four independent channels:

EXTEM: extrinsically activated coagulation, by tissue factor

INTEM: intrinsically activated coagulation, by ellagic acid

FIBTEM: extrinsically activated coagulation, by tissue factor; any contribution of platelets to clot formation is eliminated by cytochalasin D, allowing detection of fibrinogen deficiency or fibrin polymerization disorders.

APTEM: extrinsically activated coagulation, by tissue factor; aprotinin is added in order to block fibrinolysis; this reveals hyperfibrinolysis when compared with the EXTEM.

HEPTEM: intrinsically activated coagulation, by ellagic acid, contains heparinase to eliminate any heparin effect. Compared with the INTEM assay, it specifically confirms the presence of residual heparin.

The nomenclature used for the TEG and the ROTEM are presented in Table 2, and the graphical representation of the ROTEM is shown in Figure 1. The curve of the TEG is comparable with the curve of the ROTEM.

Definition TEG ROTEM
Time between addition of the activator and start of clot formation r value coagulation time (CT)
Time between start of clot formation and reaching an amplitude of 20 mm k value clot formation time (CFT)
Steepness of curve alpha angle alpha angle
Greatest amplitude maximum amplitude maximum clot firmness
Amplitude measured 60 minutes after onset of clotting, as a percentage of the greatest amplitude lysis index lysis index

Table 2: Nomenclature used for TEG and ROTEM.

haematology-thromboembolic-diseases-clotting-time

Figure 1: Schematic presentation of ROTEM tracing. The commonly reported variables are indicated: clotting time (CT), clot formation time (CFT), alpha angle (α), maximum clot firmness (MCF) and clot lysis (CL)

A disturbed activation of coagulation is indicated by a prolonged clotting time. An abnormal clot formation is indicated by a prolonged clot formation time and/or reduced clot firmness. Fibrinolysis is detected by lysis of the clot (> 15%) or, in case of ROTEM, by finding of a better clot formation (shorter CFT, greater MCF) in APTEM as compared to EXTEM. The clot formation time is used as a guide for fresh frozen plasma, the clot strength to judge platelet infusion. The degree of lysis is used to indicate the need for antifibrinolytic therapy.

Besides the primary difference of the movement of the cup and the pin, a second difference between the TEG and the ROTEM assay is the detector system, which is a torsion wire in the TEG, and an optical detector in the ROTEM [53]. Thirdly, the TEG system uses a kaolin/phospholipid mixture as activator, whereas in the ROTEM either ellagic acid or tissue factor is used. Lastly, the TEG uses a larger and less diluted blood volume in the cuvette [54]. Results in viscoelastic whole blood measurement using either TEG or ROTEM are therefore not completely interchangeable and clinical interpretation should be used with caution [54]. Wasowicz et al. [55] concluded in an observational study comprising 434 CPB patients that on-CPB thromboelastography could improve risk stratification for excessive blood loss postoperatively. In contrast, Coakley et al concluded that thromboelastometry was not predictive for postoperative bleeding in cardiac surgical patients undergoing CPB [47]. Consumption of allogenic blood products tends to decrease in case viscoelastic tests are used [56-58]. However, viscoelastic clot-based assays are mainly dependent on fibrinogen levels [47,59,60]. A great deal of haemostasis is not captured by the fibrin clotting endpoints used [61]. Only 5% of thrombin is formed when clotting-based assays terminated. This is not directly related to haemostasis and thrombosis, like thrombin concentrations are, but a covariable of one of the effects of thrombin formation, i.e. fibrin (-monomer) polymerization [62].

Platelet function tests

Although thrombocytopenia occurs during cardiac surgery, this alone infrequently leads to increased blood loss if the platelet function is normal [17]. Platelet dysfunction, is one of the most important causes of non-surgical bleeding after CPB, and is not assessed by standard haematology laboratory assays [15]. Nowadays, several platelet function tests are available. We will discuss a test based on aggregation, one based on shear stress, and the last based on platelet contribution to clot strength.

Classical light transmission aggregometry performed in platelet-rich plasma is the “gold standard” for platelet function testing [63,64], but data showing correlation with blood loss are lacking [64]. The Multiplate analyser is an instrument with five channels for parallel aggregometry measurements in whole blood. Blood and reagents were pipetted into test cells containing four electrodes that formed two independent sensor units. The ability of platelets to adhere to the metal sensors was measured by the change of electrical resistance between two sensor wires. This impedance change is plotted against time and the area under the aggregation curve was used to measure the aggregation response, quantified in arbitrary aggregation units (U). Whole blood platelet aggregometry is likely affected by a reduced haematocrit and become unreliable at platelet counts < 50*109/l [63,65]. The Multiplate device will produce a result in 10 min after sampling, except when adenosine diphosphate is used as an agonist [63]. Unfortunately, it does not reflect the complex interaction interaction between thrombin, fibrinogen and platelets, and the end result of the coagulation process, the formation of the clot.

The PFA-100 platelet function analyser is an in vitro system that provides a measure of platelet function in citrated whole blood. The PFA-100 assesses the time taken for a platelet plug to occlude a microscopic aperture within a membrane coated with platelet agonists, collagen and, depending on the cartridge, epinephrine or adenosine diphosphate (ADP). The time from the start of the test until the occlusion of the aperture by a platelet plug was reported as the PFA-100 closure time (s). The test can be performed in 5-8 minutes. The PFA-100 system provides automated assessment for inherited, acquired (CPB), or drug-induced platelet dysfunction.

The Platelet-Mapping assay is a modification of the TEG, measuring percentage platelet aggregation in the presence of ADP or arachidonic acid (AA). It was designed to monitor the effect of clopidogrel and aspirin on platelets and in decision-making for timing of cardiac surgery or other invasive procedures after discontinuation of antiplatelet therapy. This assay uses a viscoelastic test in whole blood to measure the clot strength (maximum amplitude) and detects the percentage of platelets inhibited by aspirin and clopidogrel. TEG platelet-mapping tracing shows the strength of coagulation formed by activated platelets [66]. In general, platelet function analyzers are sensitive enough to detect the effects of antiplatelet drugs on platelet function in cardiac surgical patients [15,63,64]. There is evidence that platelet function analyzers were able to predict patients at risk for increased postoperative blood loss [66-69] when performed in either the pre- or post-CPB setting. However, after correcting for haematocrit and platelet count there were no differences anymore between bleeders and non-bleeders measured by the PFA-100 [69], what led to the conclusion that PFA-100 is not useful for routine use after cardiac surgery [69]. Rahe-Meyer et al. found no correlation between decreased peri-operative platelet aggregometry measured by Multiplate and blood loss in the 24-h period after surgery [65]. Although both low pre- and post-operative platelet function tests defined groups of patients with significantly higher transfusion of platelet concentrates [65,66], there is no consensus to conclude that measuring platelet function is useful in transfusion algorithms [15,37].

Calibrated Automated Thrombography (CAT)

Thrombin is the most important enzyme in the thrombotic portion of the haemostatic reaction during CPB. Therefore, monitoring thrombin generation (TG) is of utmost importance. TG reflects much - if not all - of the overall function of the blood clotting system [62] and plays several roles such as modulating the cleavage of fibrinogen to fibrin, activating platelets and the process of fibrinolysis [61,70]. It extends from coagulation activator and inhibitor to cellular regulator. In 1953 the thrombin generation test was described by Macfarlane and Biggs [71], as to be a valid function test of the clotting system. Later, the calibrated automated thrombography (CAT) test, following triggering of plasma with tissue factor, was developed by Hemker and co-workers [62,72,73] for monitoring of the rate and extent of TG by the continuous measurement of the fluorescent split-product of the fluorogenic substrate. The course of TG is graphically represented in a thrombogram. TG is measured in a fluorometer equipped with 96 wells. In one well a calibrator has been added. In a second well TG is triggered with tissue factor and phospholipids in platelet poor plasma (PPP), and only with tissue factor in PRP. A fresh mixture of fluorogenic substrate (Z-Gly-Gly-Arg-AMC) and CaCl2 (called FluCa) is dispensed to all the wells to be measured. The readings from the TG are compared to the readings of the calibrator well, thrombin concentration is calculated and thrombin concentration in time is displayed. CAT parameters are: 1) lag time (min): the initiation phase of clotting, that equals the clotting time; 2) peak height (nM): the maximal amount of thrombin formed; 3) endogenous thrombin potential (ETP) (nM*min): the area under the TG curve, representing thrombin generation and decay in time; and 4) time to peak (min): the time needed to achieve the peak height (Figure 2). The total time course of CAT to provide the required clinical relevant information could be less than 30 minutes (depending on the haemostatic condition of the patient). The first parameters are already known within 5 minutes.

haematology-thromboembolic-diseases-endogenous-thrombin

Figure 2: Schematic presentation of CAT tracing. The commonly reported variables are indicated: lag time, peak height, time to peak and endogenous thrombin potential (ETP).

Recently, it became possible to measure TG in whole blood instead of plasma. This is an important development as it is approaching the in vivo conditions more closely. No centrifugation step is required, which could lead to uncontrolled variations in cell counts and activation of blood cells. The whole blood CAT assay is described elsewhere [74].

CAT can be used for detection and quantification of bleeding tendency, for detecting hyper coagulability, and assessing substitution and antithrombotic treatments. There is already evidence that thrombin generation measured by CAT is predictive for increased bleeding risk after cardiac surgery [47,75-77], however, further research involving the clinical benefits is warranted.

Optimal Approach for Monitoring of Haemostasis in Cardiac Surgical Patients Undergoing CPB

The ideal way to monitor haemostasis and anticoagulation during CPB, should be a test that includes all factors that interact to create haemostasis at a specific moment: blood cells, plasma proteins, contributions of the vessel wall, flow conditions, and the influence of anticoagulants. Including all these aspects into one assay will reflect closer the in vivo circumstances. Measuring of thrombin generation in whole blood could be such a test. Besides the most ideal test conditions, the test should also be clinically useful. The clinical utility of coagulation tests is the likelihood that a test result will lead to an improved health outcome. In cardiac surgery this means to get more control over individual blood loss and to facilitate optimal, targeted administration of pharmacological agents and blood components in case of excessive bleeding [78]. The clinical consequences of a disturbed haemostatic condition could be reduced with more accurate control of heparin anticoagulation and its reversal. Novel pharmacological approaches have resulted in the introduction of new oral anticoagulants (NOACs) into the adult clinical setting. These, direct thrombin and factor Xa, inhibitors have been developed to reduce the well-established drawbacks of currently available anticoagulants (vitamin K antagonists and heparins). They are currently administered at fixed doses without control of their effect on the clotting system. However, measurement of their anticoagulant effect may be desirable in situations like bleeding and emergent surgery particularly when patients did not quit intake of NOACs. Whether NOACs require routine monitoring cannot be answered at present, as relevant data are lacking and monitoring is inadequate. Another practical aspect of running the ideal test is the simplicity, and results should be rapidly available. Point-of-care, preferably at the patient’s bedside, would be the goal of haemostatic monitoring, where interventions could be done immediately. Obviously, development of coagulation monitors in cardiac surgery starts with understanding the process of coagulation, the disturbances of coagulation, but also the snags and pitfalls. The clinical utility, especially prediction of bleeding postoperatively, is the next step in investigating available tests. The most ideal goal is to create an optimal test from bench to bedside in the near future.

Conclusion

Earlier diagnosis of haemostatic abnormalities and risk of bleeding in cardiac surgical patients undergoing cardiopulmonary bypass is warranted. Viscoelastic whole blood tests could improve timely identification of haemostatic disturbances. However, CAT gives a more complete reflection of the overall blood coagulation system. Further research of the clinical applicability of viscoelastic tests and CAT, preferable in whole blood, is required. Finally, several improvements such as simplicity in running the test, and a short turnaround time would be highly desirable.

References

  1. Despotis GJ, Gravlee G, Filos K, Levy J (1999) Anticoagulation monitoring during cardiac surgery: a review of current and emerging techniques.Anesthesiology 91: 1122-1151.
  2. Edmunds LH Jr, Colman RW (2006) Thrombin during cardiopulmonary bypass.Ann Thorac Surg 82: 2315-2322.
  3. Chandler WL (2005) Effects of hemodilution, blood loss, and consumption on hemostatic factor levels during cardiopulmonary bypass.J Cardiothorac Vasc Anesth 19: 459-467.
  4. Waldén K, Jeppsson A, Nasic S, Backlund E, Karlsson M (2014) Low preoperative fibrinogen plasma concentration is associated with excessive bleeding after cardiac operations.Ann Thorac Surg 97: 1199-1206.
  5. Woodman RC, Harker LA (1990) Bleeding complications associated with cardiopulmonary bypass.Blood 76: 1680-1697.
  6. Bull BS, Hay KL, Herrmann PC (2009) Postoperative bypass bleeding: a bypass-associated dilutional (BAD) coagulopathy?Blood Cells Mol Dis 43: 256-259.
  7. Görlinger K, Shore-Lesserson L, Dirkmann D, Hanke AA, Rahe-Meyer N, et al. (2013) Management of hemorrhage in cardiothoracic surgery.J Cardiothorac Vasc Anesth 27: S20-34.
  8. Innerhofer P, Kienast J (2010) Principles of perioperative coagulopathy.Best Pract Res Clin Anaesthesiol 24: 1-14.
  9. Bevan DH (1999) Cardiac bypass haemostasis: putting blood through the mill.Br J Haematol 104: 208-219.
  10. Koster A, Fischer T, Gruendel M, Mappes A, Kuebler WM, et al. (2003) Management of heparin resistance during cardiopulmonary bypass: the effect of five different anticoagulation strategies on hemostatic activation. J Cardiothorac Vasc Anesth 17: 171-175.
  11. Blaisdell FW (1996) Matas Lecture. Heparin--controversies and misconceptions.Cardiovasc Surg 4: 691-700.
  12. Hirsh J, Raschke R, Warkentin TE, Dalen JE, Deykin D, et al. (1995) Heparin: mechanism of action, pharmacokinetics, dosing considerations, monitoring, efficacy, and safety. Chest 108: 258S-275S.
  13. Horne MK 3rd, Chao ES (1989) Heparin binding to resting and activated platelets.Blood 74: 238-243.
  14. Walenga JM, Hoppensteadt DA (2004) Monitoring the new antithrombotic drugs.Semin Thromb Hemost 30: 683-695.
  15. Avidan MS, Alcock EL, Da Fonseca J, Ponte J, Desai JB, et al. (2004) 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 92: 178-186.
  16. Garvin S, FitzGerald DC, Despotis G, Shekar P, Body SC (2010) Heparin concentration-based anticoagulation for cardiac surgery fails to reliably predict heparin bolus dose requirements.Anesth Analg 111: 849-855.
  17. Shore-Lesserson L (2005) Evidence based coagulation monitors: heparin monitoring, thromboelastography, and platelet function. Semin Cardiothorac Vasc Anesth 9: 41-52.
  18. Hardy JF, Belisle S, Robitaille D, Perrault J, Roy M, et al. (1996) Measurement of heparin concentration in whole blood with the Hepcon/HMS device does not agree with laboratory determination of plasma heparin concentration using a chromogenic substrate for activated factor X. J Thorac Cardiovasc Surg 112: 154-161.
  19. Hemker HC, Béguin S (1993) Standard and method independent units for heparin anticoagulant activities.Thromb Haemost 70: 724-728.
  20. al Dieri R, Alban S, Béguin S, Hemker HC (2004) Thrombin generation for the control of heparin treatment, comparison with the activated partial thromboplastin time.J Thromb Haemost 2: 1395-1401.
  21. de Moerloose P (1996) Laboratory evaluation of hemostasis before cardiac operations.Ann Thorac Surg 62: 1921-1925.
  22. Tanaka KA, Key NS, Levy JH (2009) Blood coagulation: hemostasis and thrombin regulation.Anesth Analg 108: 1433-1446.
  23. Hoffman M, Monroe DM (2007) Coagulation 2006: a modern view of hemostasis.Hematol Oncol Clin North Am 21: 1-11.
  24. Levy JH, Dutton RP, Hemphill JC 3rd, Shander A, Cooper D, et al. (2010) Multidisciplinary approach to the challenge of hemostasis.Anesth Analg 110: 354-364.
  25. Chee YL, Crawford JC, Watson HG, Greaves M (2008) Guidelines on the assessment of bleeding risk prior to surgery or invasive procedures. British Committee for Standards in Haematology.Br J Haematol 140: 496-504.
  26. Gelb AB, Roth RI, Levin J, London MJ, Noall RA, et al. (1996) Changes in blood coagulation during and following cardiopulmonary bypass: lack of correlation with clinical bleeding.Am J Clin Pathol 106: 87-99.
  27. Gravlee GP, Arora S, Lavender SW, Mills SA, Hudspeth AS, et al. (1994) Predictive value of blood clotting tests in cardiac surgical patients.Ann Thorac Surg 58: 216-221.
  28. Hattersley PG (1966) Activated coagulation time of whole blood.JAMA 196: 436-440.
  29. Bull BS, Korpman RA, Huse WM, Briggs BD (1975) Heparin therapy during extracorporeal circulation. I. Problems inherent in existing heparin protocols.J Thorac Cardiovasc Surg 69: 674-684.
  30. Garvin S, Fitzgerald D, Muehlschlegel JD, Perry TE, Fox AA, et al. (2010) Heparin dose response is independent of preoperative antithrombin activity in patients undergoing coronary artery bypass graft surgery using low heparin concentrations. Anesth Analg 111: 856-861.
  31. Lemmer JH Jr, Despotis GJ (2002) Antithrombin III concentrate to treat heparin resistance in patients undergoing cardiac surgery.J Thorac Cardiovasc Surg 123: 213-217.
  32. Nicholson SC, Keeling DM, Sinclair ME, Evans RD (2001) Heparin pretreatment does not alter heparin requirements during cardiopulmonary bypass.Br J Anaesth 87: 844-847.
  33. Flom-Halvorsen HI, Ovrum E, Abdelnoor M, Bjørnsen S, Brosstad F (1999) Assessment of heparin anticoagulation: comparison of two commercially available methods.Ann Thorac Surg 67: 1012-1016.
  34. Prisco D, Paniccia R (2003) Point-of-Care Testing of Hemostasis in Cardiac Surgery.Thromb J 1: 1.
  35. Svenmarker S, Appelblad M, Jansson E, Häggmark S (2004) Measurement of the activated clotting time during cardiopulmonary bypass: differences between Hemotec ACT and Hemochron Jr apparatus.Perfusion 19: 289-294.
  36. Bowers J, Ferguson JJ 3rd (1994) The use of activated clotting times to monitor heparin therapy during and after interventional procedures.Clin Cardiol 17: 357-361.
  37. Slaughter TF, LeBleu TH, Douglas JM Jr, Leslie JB, Parker JK, et al. (1994) Characterization of prothrombin activation during cardiac surgery by hemostatic molecular markers.Anesthesiology 80: 520-526.
  38. Koster A, Despotis G, Gruendel M, Fischer T, Praus M, et al. (2002) The plasma supplemented modified activated clotting time for monitoring of heparinization during cardiopulmonary bypass: a pilot investigation. Anesth Analg 95: 26-30.
  39. Hemker HC, Béguin S (2000) Phenotyping the clotting system.Thromb Haemost 84: 747-751.
  40. Weber CF, Zacharowski K (2012) Perioperative point of care coagulation testing.Dtsch Arztebl Int 109: 369-375.
  41. Enriquez LJ, Shore-Lesserson L (2009) Point-of-care coagulation testing and transfusion algorithms.Br J Anaesth 103 Suppl 1: i14-22.
  42. Shore-Lesserson L, Manspeizer HE, DePerio M, Francis S, Vela-Cantos F, et al. (1999) Thromboelastography-guided transfusion algorithm reduces transfusions in complex cardiac surgery.Anesth Analg 88: 312-319.
  43. Toulon P, Ozier Y, Ankri A, Fléron MH, Leroux G, et al. (2009) Point-of-care versus central laboratory coagulation testing during haemorrhagic surgery. A multicenter study.Thromb Haemost 101: 394-401.
  44. Faraday N, Guallar E, Sera VA, Bolton ED, Scharpf RB, et al. (2002) Utility of whole blood hemostatometry using the clot signature analyzer for assessment of hemostasis in cardiac surgery.Anesthesiology 96: 1115-1122.
  45. Despotis G, Avidan M, Eby C (2009) Prediction and management of bleeding in cardiac surgery.J Thromb Haemost 7 Suppl 1: 111-117.
  46. Karkouti K, McCluskey SA, Syed S, Pazaratz C, Poonawala H, et al. (2010) The influence of perioperative coagulation status on postoperative blood loss in complex cardiac surgery: a prospective observational study.Anesth Analg 110: 1533-1540.
  47. Coakley M, Hall JE, Evans C, Duff E, Billing V, et al. (2011) Assessment of thrombin generation measured before and after cardiopulmonary bypass surgery and its association with postoperative bleeding.J Thromb Haemost 9: 282-292.
  48. Pillai RC, Fraser JF, Ziegenfuss M, Bhaskar B (2014) The influence of circulating levels of fibrinogen and perioperative coagulation parameters on predicting postoperative blood loss in cardiac surgery: a prospective observational study. J Card Surg 29: 189-195.
  49. Josefy S, Briones R, Bryant BJ (2011) Preoperative coagulation studies to predict blood component usage in coronary artery bypass graft surgery.Immunohematology 27: 151-153.
  50. Kuepper F, Dangas G, Mueller-Chorus A, Kulka PM, Zenz M, et al. (2003) Fibrinolytic activity and bleeding after cardiac surgery with cardiopulmonary bypass and low-dose aprotinin therapy.Blood Coagul Fibrinolysis 14: 147-153.
  51. Snircova J, Jares M, Maly M, Straka Z, Spegar J, et al. (2008) Postoperative blood loss in coronary surgery. No real impact of fibrinolysis detected by thromboelastography and D-dimers. A prospective, randomized study.Int Heart J 49: 25-38.
  52. HARTERT H (1951) [Thrombelastography, a method for physical analysis of blood coagulation].Z Gesamte Exp Med 117: 189-203.
  53. Luddington RJ (2005) Thrombelastography/thromboelastometry.Clin Lab Haematol 27: 81-90.
  54. Venema LF, Post WJ, Hendriks HG, Huet RC, de Wolf JT, et al. (2010) An assessment of clinical interchangeability of TEG and RoTEM thromboelastographic variables in cardiac surgical patients.Anesth Analg 111: 339-344.
  55. Wasowicz M, McCluskey SA, Wijeysundera DN, Yau TM, Meinri M, et al. (2010) The incremental value of thrombelastography for prediction of excessive blood loss after cardiac surgery: an observational study.Anesth Analg 111: 331-338.
  56. Ak K, Isbir CS, Tetik S, Atalan N, Tekeli A, et al. (2009) Thromboelastography-based transfusion algorithm reduces blood product use after elective CABG: a prospective randomized study.J Card Surg 24: 404-410.
  57. Aoki K, Sugimoto A, Nagasawa A, Saito M, Ohzeki H (2012) Optimization of thromboelastography-guided platelet transfusion in cardiovascular surgery.Gen Thorac Cardiovasc Surg 60: 411-416.
  58. Weber CF, Görlinger K, Meininger D, Herrmann E, Bingold T, et al. (2012) Point-of-care testing: a prospective, randomized clinical trial of efficacy in coagulopathic cardiac surgery patients.Anesthesiology 117: 531-547.
  59. Lee GC, Kicza AM, Liu KY, Nyman CB, Kaufman RM, et al. (2012) Does rotational thromboelastometry (ROTEM) improve prediction of bleeding after cardiac surgery?Anesth Analg 115: 499-506.
  60. Theusinger OM, Schroder CM, Eismon J, Emmert MY, Seifert B, et al. (2013) The influence of laboratory coagulation tests and clotting factor levels on Rotation Thromboelastometry (ROTEM(R)) during major surgery with hemorrhage. Anesth Analg 117: 314-321.
  61. Mann KG, Brummel K, Butenas S (2003) What is all that thrombin for?J Thromb Haemost 1: 1504-1514.
  62. Hemker HC, Giesen P, AlDieri R, Regnault V, de Smed E, et al. (2002) The calibrated automated thrombogram (CAT): a universal routine test for hyper- and hypocoagulability.Pathophysiol Haemost Thromb 32: 249-253.
  63. Reece MJ, Klein AA, Salviz EA, Hastings A, Ashworth A, et al. (2011) Near-patient platelet function testing in patients undergoing coronary artery surgery: a pilot study.Anaesthesia 66: 97-103.
  64. Velik-Salchner C, Maier S, Innerhofer P, Kolbitsch C, Streif W, et al. (2009) An assessment of cardiopulmonary bypass-induced changes in platelet function using whole blood and classical light transmission aggregometry: the results of a pilot study. Anesth Analg 108: 1747-1754.
  65. Rahe-Meyer N, Winterhalter M, Boden A, Froemke C, Piepenbrock S, et al. (2009) Platelet concentrates transfusion in cardiac surgery and platelet function assessment by multiple electrode aggregometry.Acta Anaesthesiol Scand 53: 168-175.
  66. Chowdhury M, Shore-Lesserson L, Mais AM, Leyvi G (2014) Thromboelastograph with Platelet Mapping(TM) predicts postoperative chest tube drainage in patients undergoing coronary artery bypass grafting. J Cardiothorac Vasc Anesth 28: 217-223.
  67. Preisman S, Kogan A, Itzkovsky K, Leikin G, Raanani E (2010) Modified thromboelastography evaluation of platelet dysfunction in patients undergoing coronary artery surgery.Eur J Cardiothorac Surg 37: 1367-1374.
  68. Weitzel NS, Weitzel LB, Epperson LE, Karimpour-Ford A, Tran ZV, et al. (2012) Platelet mapping as part of modified thromboelastography (TEG®) in patients undergoing cardiac surgery and cardiopulmonary bypass.Anaesthesia 67: 1158-1165.
  69. Forestier F, Coiffic A, Mouton C, Ekouevi D, Chêne G, et al. (2002) Platelet function point-of-care tests in post-bypass cardiac surgery: are they relevant?Br J Anaesth 89: 715-721.
  70. Koestenberger M, Cvirn G, Nagel B, Rosenkranz A, Leschnik B, et al. (2008) Thrombin generation determined by calibrated automated thrombography (CAT) in pediatric patients with congenital heart disease.Thromb Res 122: 13-19.
  71. MACFARLANE RG, BIGGS R (1953) A thrombin generation test; the application in haemophilia and thrombocytopenia.J Clin Pathol 6: 3-8.
  72. Hemker HC, Giesen P, Al Dieri R, Regnault V, de Smedt E, et al. (2003) Calibrated automated thrombin generation measurement in clotting plasma.Pathophysiol Haemost Thromb 33: 4-15.
  73. Hemker HC, Giesen PL, Ramjee M, Wagenvoord R, Béguin S (2000) The thrombogram: monitoring thrombin generation in platelet-rich plasma.Thromb Haemost 83: 589-591.
  74. Ninivaggi M, Apitz-Castro R, Dargaud Y, de Laat B, Hemker HC, et al. (2012) Whole-blood thrombin generation monitored with a calibrated automated thrombogram-based assay.Clin Chem 58: 1252-1259.
  75. Bosch Y, Al Dieri R, ten Cate H, Nelemans P, Bloemen S, et al. (2013) Preoperative thrombin generation is predictive for the risk of blood loss after cardiac surgery: a research article.J Cardiothorac Surg 8: 154.
  76. Bosch YP, Al Dieri R, ten Cate H, Nelemans PJ, Bloemen S, et al. (2014) Measurement of thrombin generation intra-operatively and its association with bleeding tendency after cardiac surgery.Thromb Res 133: 488-494.
  77. Hayashi T, Sakurai Y, Fukuda K, Yada K, Ogiwara K, et al. (2011) Correlations between global clotting function tests, duration of operation, and postoperative chest tube drainage in pediatric cardiac surgery. Paediatr Anaesth 21: 865-871.
  78. Despotis GJ, Joist JH, Goodnough LT (1997) Monitoring of hemostasis in cardiac surgical patients: impact of point-of-care testing on blood loss and transfusion outcomes.Clin Chem 43: 1684-1696.
Citation: Bosch YPJ, Weerwind PW, Baheramsjah Mochtar B, Dieri RA (2015) Monitoring of Haemostasis and Anticoagulation in Cardiopulmonary Bypass Patients. J Hematol Thrombo Dis 3:179.

Copyright: © 2014 Bosch YPJ, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top