Your activity: 28588 p.v.
your limit has been reached. plz Donate us to allow your ip full access, Email: [email protected]

Reversal of anticoagulation and management of bleeding after cardiopulmonary bypass

Reversal of anticoagulation and management of bleeding after cardiopulmonary bypass
Authors:
Kamrouz Ghadimi, MD
Ian J Welsby, BSc, MBBS, FRCA
Section Editor:
Jonathan B Mark, MD
Deputy Editor:
Nancy A Nussmeier, MD, FAHA
Literature review current through: Feb 2022. | This topic last updated: Jul 02, 2021.

INTRODUCTION — Blood and coagulation management are key components of anesthetic management for cardiac surgical procedures that require cardiopulmonary bypass (CPB) because of the need for full systemic anticoagulation during CPB and its reversal after weaning from CPB. Other major factors influencing coagulation include hemodilution due to fluid priming of the extracorporeal circuit, fibrinolysis, platelet dysfunction, coagulation factor consumption, and systemic hypothermia that occurs during CPB, as well as blood loss during surgery involving the heart and great vessels [1,2].

This topic discusses reversal of anticoagulation and management of bleeding after cardiac surgery with CPB. Strategies to avoid or minimize blood loss and transfusion of blood products before and during CPB are discussed separately. (See "Blood management and anticoagulation for cardiopulmonary bypass".)

General principles for perioperative blood management are discussed in separate topics. (See "Perioperative blood management: Strategies to minimize transfusions" and "Intraoperative transfusion of blood products in adults".)

REVERSAL OF ANTICOAGULATION — Systemic heparin anticoagulation is reversed with administration of protamine after weaning from CPB [3].

Administration and dosing of protamine

Timing of initial administration – Neutralization of systemic heparin with protamine is typically initiated after weaning from CPB but before aortic decannulation. This timing allows rapid reinstitution of CPB in the event of a catastrophic protamine reaction [4]. An initial "test dose" of protamine 10 mg is administered to allow early detection of serious hemodynamic changes that may indicate a protamine reaction. If there is no adverse reaction, the remainder of the protamine dose is administered slowly, typically over a 5 to 15 minute period, preferably by infusion [5,6]. Slower infusion may avoid potential vasodilation. (See 'Management of protamine reactions' below.)

Suctioning of blood from the surgical field into the pump reservoir (ie, "cardiotomy suction" or "pump suckers") is discontinued either at the onset of protamine administration or when a small portion (typically less than one-third) of the total initial dose has been administered. It is hypothesized that neutralization of heparin in the blood remaining in the CPB reservoir may cause clot formation, which would prohibit emergency reinstitution of CPB.

Initial dosing – Calculation of protamine dosing for heparin reversal is ideally based on a point-of-care (POC) titration to existing heparin in the blood [7], or if a POC heparin-protamine titration assay is not available, by administering 0.7 mg protamine per 100 units of the initial pre-bypass heparin dose. After administration of the initial calculated protamine dose, further evaluation for residual heparin effect is accomplished by measuring the activated clotting time (ACT), rechecking the heparin-protamine titration assay, and/or by checking viscoelastic clot strength with POC tests of hemostatic function [8-11] (see "Intraoperative transfusion of blood products in adults", section on 'Overall hemostatic function'). Plasma concentrations of residual heparin can insidiously increase after administration of the initial protamine dose as heparinized blood from the CPB pump is reinfused, and as peripheral tissues gradually release endothelial bound heparin into the circulation as reperfusion occurs. Additional protamine is administered according to heparin-protamine titration assay measurements of residual heparin, or an additional 25 to 100 mg of protamine may be administered if evidence of residual heparin effect is noted on the ACT or with viscoelastic testing. However, protamine overdosing should be avoided [3]. In most cases, there is no reason to exceed 1 mg protamine per 100 units of total administered heparin; in practice, we rarely administer more than 250 mg of protamine.

There are numerous institutional variations in protamine dosing practices to achieve heparin neutralization. Traditional methods relied on fixed protamine dosing based on body weight and/or the amount of heparin administered. A 2013 meta-analysis of these traditional methods versus use of a heparin-protamine titration assay found significantly less mean postoperative blood loss with protamine titration compared with standard (fixed) dosing [12]. Other studies have noted that excess protamine is associated with prolonged ACT, international normalized ratio (INR), and activated partial thromboplastin time (aPTT) values, as well as inhibited platelet function, factor V activation, and excessive bleeding after CPB, particularly if the ratio of protamine to heparin exceeds 2.6 mg protamine per 100 units of heparin [7,8,13-17]. We agree with practice guidelines stating that it is reasonable to limit this ratio to ≤2.6 [7,18].

Postbypass and postoperative protamine administration – We administer a protamine infusion of 25 to 50 mg/hour over two to four postoperative hours to potentially reduce blood loss by avoiding "heparin rebound" [7,18-20]. Release of heparin into the intravascular space may occur after the initial protamine dose has been metabolized. This "heparin rebound" is particularly likely in patients who received a large dose of heparin due to altered heparin responsiveness [7,18,21] (see "Blood management and anticoagulation for cardiopulmonary bypass", section on 'Heparin resistance'). Also, heparin may accumulate in adipose tissue due to its lipophilicity, with slow eventual release [19,22]. Furthermore, active cooling during CPB causes vasoconstriction in peripheral tissues, especially adipose tissue, leading to a potential depot of heparin for later release after reperfusion.

Administration of a postbypass and postoperative protamine infusion allows optimal neutralization of heparin that may be slowly released from such "depots." Heparin levels measured during these phases usually range from 0.1 to 0.3 international units/mL, which is at the low end of the therapeutic range. Notably, the ACT and even activated partial thromboplastin time (aPTT) are poorly sensitive measurements of residual heparin in this low range; plasma heparin concentrations may be more accurate [23,24].

Management of protamine reactions — Mechanisms responsible for protamine reactions include:

Vasodilation due to direct and indirect effects of protamine and potential complement activation by heparin-protamine complexes [4,18,25-27]. Such reactions are common, associated with a faster rate of protamine infusion, and are treated with administration of vasopressors as needed (table 1).

A more severe anaphylactic reaction with vasoplegia is less common, but may occur in individuals who have antibodies to protamine due to previous exposure (eg, protamine-containing insulin) or due to allergy [4,7,18,25,26,28-30]. In such cases, cardiovascular collapse may be unrelated to the rate of protamine administration. These reactions are treated in the same manner as other perioperative anaphylactic reactions (table 2). (See "Perioperative anaphylaxis: Clinical manifestations, etiology, and management", section on 'Initial management'.)

Acute pulmonary vasoconstriction leading to right ventricular failure is more rare, and may be accompanied by bronchospasm or noncardiogenic pulmonary edema due to thromboxane release from platelets [4,7,18,25-27,29,31-33]. Such severe reactions are likely caused by either IgG antibodies or heparin-protamine complexes that trigger thromboxane release. The differential diagnosis includes blood product administration causing transfusion-related acute lung injury (TRALI), which can produce a similar reaction [34]. Treatment consists of stopping protamine and implementing resuscitative measures. Systemic reheparinization may be necessary to allow immediate reinstitution of CPB [7,31].

When resuscitation from any severe protamine reaction requires heparin readministration for reinstitution of CPB for temporary hemodynamic support, we administer anti-inflammatory and vasoactive agents. Specifically, we administer a systemic steroid dose (eg, methylprednisolone 125 mg), together with the H1 antihistamine diphenhydramine 50 mg and a histamine-2 receptor antagonist (eg, famotidine, cimetidine), as noted in the table (table 2). An infusion of epinephrine should be administered, and norepinephrine or vasopressin should be initiated if necessary to treat refractory hypotension (table 1). In patients with persistent refractory vasodilatory shock, it is reasonable to administer a dose of methylene blue 1 to 2 mg/kg before attempting to wean from CPB [35-37].

For patients with suspected allergic reaction, a blood sample should be collected at or shortly after the event, which may reveal elevations in tryptase. After postoperative recovery patients are referred to an allergy specialist. (See "Perioperative anaphylaxis: Clinical manifestations, etiology, and management", section on 'Laboratory tests at the time of the reaction' and "Perioperative anaphylaxis: Clinical manifestations, etiology, and management", section on 'Referral for allergy evaluation'.)

After stabilization on CPB we consider next steps in consultation with members of the anesthesiology, surgical, and perfusionist teams. There are no clear guidelines regarding the safety of readministering protamine to neutralize the additional systemic dose of heparin after a catastrophic protamine reaction [18,27,30]. In some cases, clinicians have safely readministered protamine without incident [30]. In theory, the initial anaphylactic reaction leads to a post-anaphylaxis refractory period due to temporary depletion of inflammatory mediators [30,34]. However, some clinicians have avoided readministration of protamine, electing instead to reverse anticoagulation with blood products. With this option, excessive bleeding may require transfusion of large volumes of fresh frozen plasma (FFP), platelets, and fibrinogen, as well as red blood cells (RBCs), with the associated adverse effects of massive transfusion [30,38]. (See "Massive blood transfusion".)

In rare cases when noncardiogenic pulmonary edema or respiratory distress syndrome is severe after a protamine reaction, temporary extracorporeal membrane oxygenation may be necessary after weaning from CPB. (See "Intraoperative problems after cardiopulmonary bypass", section on 'Extracorporeal membrane oxygenation'.)

Alternatives to protamine — There are no approved alternatives to protamine for heparin reversal [18]. With a half-life of approximately 60 to 90 minutes, the anticoagulant effect of heparin may persist for four to six hours if not reversed, although blood loss replacement with blood products and/or volume expanders can accelerate the decrease in heparin concentration.

Bivalirudin is an alternative anticoagulant agent used to avoid heparin and protamine in patients with known severe allergy to either agent, or those with heparin-induced thrombocytopenia (HIT) with HIT antibodies present (table 3) (see "Management of heparin-induced thrombocytopenia (HIT) during cardiac or vascular surgery"). Bivalirudin has a half-life of approximately 45 minutes in normothermic patients with normal renal function; thus, all anticoagulant effects typically resolve within approximately two hours of administration of the last dose. If postbypass bleeding persists after administration of bivalirudin, reversal of anticoagulant effects may be achieved using combinations of therapies such as blood product replacement, hemodialysis, or plasmapheresis with plasma exchange [18].

MANAGEMENT OF BLEEDING — Bleeding necessitating transfusion occurs commonly after cardiac surgery with CPB, with transfusion rates widely varying between institutions (10 to 90 percent) [39,40]. Risk factors for perioperative bleeding and blood transfusion during cardiac surgery include advanced age, decreased preoperative red blood cell (RBC) volume (eg, small body size, preoperative anemia), and complex or redo operations [41-44]. Patients who are transfused have worse outcomes than those without transfusions [45].

Causes of excessive bleeding during and after weaning from CPB include inadequate surgical hemostasis, loss of platelets and coagulation factors due to persistent surgical bleeding, effects of hemodilution, hypothermic coagulopathy, presence of residual heparin (see 'Reversal of anticoagulation' above), and coagulopathy due to platelet activation (and consumption) and hyperfibrinolysis induced by the extracorporeal circuit. (See "Blood management and anticoagulation for cardiopulmonary bypass", section on 'Effects of cardiopulmonary bypass on hemostasis'.)

Fastidious surgical hemostasis — Meticulous surgical technique and systematic intraoperative checking of potential surgical sites of bleeding are critically important after CPB. In a meta-analysis of patients requiring mediastinal re-exploration for bleeding or cardiac tamponade after cardiac surgery, a surgical site of bleeding was identified in two-thirds of cases [46]. Excessive bleeding requiring mediastinal re-exploration has been associated with adverse outcomes that include mortality, need for mechanical circulatory support, stroke, acute renal failure, sternal wound infection, and prolonged mechanical ventilation [47].

Maintenance of normothermia — Mild (32 to 35°C) or moderate (28 to 32°C) hypothermia may be induced to provide neurologic and cardiac protection for many patients undergoing CPB, while deep hypothermia to temperatures as low as 16 to 18°C may be employed for selected patients undergoing elective circulatory arrest. (See "Management of cardiopulmonary bypass", section on 'Management during cooling and hypothermia' and "Anesthesia for aortic surgery requiring deep hypothermia", section on 'Effects of deep hypothermia'.)

Hypothermia is associated with coagulopathy due to impairment of platelet aggregation and reduced activity of clotting enzymes [48-50]. This combination of platelet and enzyme impairment typically reduces clot formation and increases perioperative blood loss and the need for transfusion [51-53].

Active warming techniques must be employed to achieve normothermia during the rewarming phase of CPB, then to maintain normothermia during the postbypass and postoperative periods. Further details regarding warming strategies are available in other topics:

(See "Management of cardiopulmonary bypass", section on 'Management during rewarming and weaning'.)

(See "Anesthesia for aortic surgery requiring deep hypothermia", section on 'Rewarming strategies'.)

(See "Perioperative temperature management", section on 'Active warming devices'.)

Use of transfusion algorithms — Similar to practice guidelines of several professional societies, we use a goal-directed protocol or algorithm to guide transfusion decisions, based on measurement of hemoglobin (Hgb) or hematocrit (Hct), as well as assessment of specific abnormalities of hemostasis using standard laboratory tests (algorithm 1), and/or point-of-care (POC) tests of hemostasis [40,41,54-57]. Use of such transfusion protocols and algorithms to guide decision-making in cardiac surgery can avoid or reduce unnecessary transfusions of blood products including RBCs, fresh frozen plasma (FFP), platelets, and cryoprecipitate [40,57-68]. (See "Intraoperative transfusion of blood products in adults", section on 'Use of a transfusion algorithm or guideline'.)

Hemoglobin or hematocrit levels – We check Hgb or Hct levels following weaning from CPB, and if the patient is actively bleeding, approximately every 30 minutes or more frequently in the profusely bleeding patient. We transfuse RBCs if Hgb level is <7 to 8 g/dL (or Hct <21 to 24 percent) [41,42] but may target a higher Hgb transfusion threshold in patients with uncontrolled hemorrhage. (See 'Red blood cells' below.)

Standard laboratory coagulation tests – Standard laboratory tests include prothrombin time (PT), activated partial thromboplastin time (aPTT), international normalized ratio (INR), fibrinogen level, and platelet count. Transfusion algorithms generally recommend administration of platelets for a platelet count <50,000 to 100,000, FFP when persistent bleeding is accompanied by a PT/INR or aPTT >1.5 times normal value, or cryoprecipitate or fibrinogen concentrate when fibrinogen levels are <150 to 200 mg/dL. (See 'Fresh frozen plasma' below and 'Platelets' below and 'Cryoprecipitate' below.)

When available, tests of platelet function are performed (table 4). However, testing for platelet dysfunction is unreliable in the actively bleeding patient since most tests need a relatively normal platelet count. Furthermore, most platelet function tests are not accurate following dilutional changes and platelet activation after CPB [69]. (See "Clinical use of coagulation tests" and "Platelet function testing".)

Point-of-care tests of hemostatic function – Similar to recommendations made in the 2019 Society of Cardiovascular Anesthesiologists Clinical Practice Improvement Advisory for Management of Perioperative Bleeding and Hemostasis in Cardiac Surgical Patients, we employ POC viscoelastic coagulation tests to guide transfusion therapy [40]. Such viscoelastic coagulation tests (eg, thromboelastography [TEG] or an adaptation of TEG known as rotational thromboelastometry [ROTEM] (table 5)) supplement standard laboratory tests of hemostatic function, and may be superior to standard tests and/or clinical judgment by allowing more rapid assessments of coagulopathy and responses to interventions (eg, transfusion of blood products, administration of hemostatic agents). Meta-analyses of randomized trials in cardiac surgical patients have noted that use of transfusion algorithms guided by viscoelastic testing reduces RBC and platelet transfusions compared with standard care (eg, standard laboratory coagulation tests and/or clinical judgment) [70-73]. Observational studies have reported similar results [62,64-66,68,74,75]. However, POC viscoelastic tests are not available in every institution, and conventional laboratory coagulation tests are also recommended as additional or alternative assessment tools [40].

Notably, platelet function defects due to aspirin, dipyridamole, P2Y12 receptor antagonists, or lower levels of reversible GP IIbIIIa antagonists (eg, eptifibatide) may not be detected by these standard viscoelastic tests because thrombin is generated in the TEG or ROTEM sample cups and produce a "false" normal test result despite the presence of clinical coagulopathy in the patient. This is consistent with the responses to thrombin agonists during platelet aggregometry testing, and is important to recognize when interpreting TEG/ROTEM results.

Clinically significant bleeding at any time during the postbypass period should trigger repeat evaluation of standard laboratory tests (algorithm 1), and/or POC tests to determine which algorithm-based therapies are most likely to reverse existing and developing abnormalities.

Further information regarding POC tests of coagulation is available in separate topics:

(See "Intraoperative transfusion of blood products in adults", section on 'Overall hemostatic function'.)

(See "Clinical use of coagulation tests", section on 'Point-of-care testing'.)

(See "Coagulopathy in trauma patients", section on 'Thromboelastography-based transfusion'.)

Transfusion of blood products

Red blood cells — We transfuse RBCs for Hgb <7 to 8 g/dL, similar to professional society practice guidelines for blood conservation during cardiac surgery [3,41,42]. However, transfusion decisions are individualized, taking into account patient-related factors (eg, age, severity of illness, cardiac function, risk for critical end-organ ischemia), laboratory parameters (eg, low mixed venous oxygen saturation [SVO2]), evidence of myocardial ischemia on electrocardiogram (ECG) or transesophageal echocardiography (TEE), and the clinical setting (massive or active blood loss) [41-43,76]. When blood loss is rapid, immediate transfusion may be life-saving and necessary before quantitative laboratory assessment of Hgb can be obtained, based on the rate of bleeding, expected volume of ongoing bleeding, and the preoperative red cell mass [41,56]. When RBC transfusion is necessary, leukocyte-reduced blood is preferred. (See "Intraoperative transfusion of blood products in adults", section on 'Red blood cells' and "Indications and hemoglobin thresholds for red blood cell transfusion in the adult", section on 'Cardiac surgery' and "Leukoreduction to prevent complications of blood transfusion".)

When transfusion is necessary, available salvaged blood is returned first, followed by reinfusion of blood units harvested via normovolemic hemodilution, then allogeneic RBCs [40]. (See "Surgical blood conservation: Blood salvage" and "Blood management and anticoagulation for cardiopulmonary bypass", section on 'Acute normovolemic hemodilution'.)

The optimal Hgb level during and after cardiac surgery is not known. In a multicenter randomized trial conducted in more than 5000 cardiac surgical patients, the primary composite outcome that included death, myocardial infarction, renal failure, and stroke was similar with use of a restrictive transfusion strategy (Hgb trigger set at <7.5 g/dL) compared with use of a liberal transfusion strategy (Hgb trigger set at <9.5 g/dL) during the intraoperative and postoperative periods [77]. In the restrictive group, fewer patients received any RBC transfusion (52 versus 73 percent; odds ratio [OR] 0.41, 95% CI 0.37-0.47), and fewer total RBC units were transfused (two versus three units; rate ratio 0.85. 95% CI 0.82-0.88). At follow-up six months later, the primary composite outcome remained similar (17.4 percent in the restrictive group versus 17.1 percent in the liberal group; OR 1.02, 95% CI 0.87-1.18), and mortality did not differ [78]. A meta-analysis of randomized trials investigating restrictive versus liberal strategies for RBC transfusion in cardiac surgical patients noted similar outcomes for mortality, myocardial infarction, stroke, renal failure, or infection with either strategy [79]. Also, fewer RBC transfusions were reported with a restrictive transfusion strategy in some trials [45,80,81]. However, in one meta-analysis of randomized trials that included both noncardiac and cardiac surgical patients, a trend toward lower mortality was noted if a liberal rather than a restrictive transfusion threshold was employed [82]. This isolated, non-significant finding is insufficient to justify liberal transfusion in the absence of a clinical indication.

In observational studies of cardiac surgical patients, an association between RBC transfusions and adverse postoperative outcomes has been noted (eg, mortality, renal failure, stroke), but these findings are not consistent, nor apparent in the prospective studies discussed above [83,84]. Massive transfusions may pose a greater risk [38,85]. Further discussion is available in a separate topic. (See "Early noncardiac complications of coronary artery bypass graft surgery", section on 'Blood transfusion'.)

Fresh frozen plasma — We use goal-directed protocols or algorithms to guide transfusion decisions based on specific abnormalities of hemostasis (eg, PT, aPTT, or INR >1.5 times normal value in the setting of ongoing bleeding) [41,86] (see 'Use of transfusion algorithms' above). Transfusion of FFP has many of the risks associated with RBC transfusion, and is not transfused in the absence of significant laboratory evidence of coagulopathy and active bleeding [3,40,86].

Transfusion of FFP is also included in massive transfusion protocols, usually in a 1:1 ratio with transfusion of RBC units, which is relevant to a massively bleeding cardiac surgery patient [87]. (See "Intraoperative transfusion of blood products in adults", section on 'Plasma'.)

Platelets — Transfusion of platelets is reserved for patients with platelet count <100,000/mm3 (or platelet dysfunction due to residual anti-platelet drug effect) if there is ongoing clinically significant microvascular bleeding [40]. Patients who are taking antiplatelet medication, particularly P2Y12 receptor inhibitors such as clopidogrel, prasugrel, and ticagrelor, are at higher risk for such bleeding due to platelet dysfunction. (See "Intraoperative transfusion of blood products in adults", section on 'Platelets' and "Early noncardiac complications of coronary artery bypass graft surgery", section on 'Antiplatelet agents'.)

Thrombocytopenia is common in the immediate postbypass period due to a combination of hemodilution, platelet loss due to persistent surgical bleeding, platelet adherence to the CPB circuit surface, consumption due to coagulation activation, and accelerated clearance caused by thrombin-mediated activation. Rarely, thrombocytopenia is a manifestation of acute disseminated intravascular coagulation. (See "Blood management and anticoagulation for cardiopulmonary bypass", section on 'Effects of cardiopulmonary bypass on hemostasis' and "Evaluation and management of disseminated intravascular coagulation (DIC) in adults", section on 'Acute DIC'.)

Management of thrombocytopenia and thrombosis due to heparin-induced thrombocytopenia (HIT) in the postoperative period is discussed separately. (See "Management of heparin-induced thrombocytopenia (HIT) during cardiac or vascular surgery".)

Cryoprecipitate — Cryoprecipitate can be used to treat hypofibrinogenemia (eg, <100 mg/dL) in a patient with intractable bleeding and coagulopathy when fibrinogen concentrate is not available (see 'Fibrinogen concentrate versus cryoprecipitate' below). One unit of cryoprecipitate contains all of the fibrinogen present in one unit of whole blood (approximately 200 to 400 mg) in a volume of 10 to 20 mL (table 6). Each unit raises the plasma fibrinogen concentration by approximately 7 to 10 mg/dL. For adult patients, cryoprecipitate is typically prepared as a pooled product combining individual cryoprecipitate units derived from 5 to 10 blood donors in a final volume of approximately 50 to 200 mL. Due to the low volume of antibodies in each unit, cryoprecipitate does not need to be ABO matched in adults. (See "Clinical use of Cryoprecipitate".)

Compared with fibrinogen concentrate (see 'Fibrinogen concentrate versus cryoprecipitate' below), cryoprecipitate is more readily available with lower cost in the United States, and is used more commonly in both the United States and the United Kingdom. Also, bleeding and coagulopathy are often multifactorial in the immediate postbypass period. For this reason, many clinicians prefer cryoprecipitate to treat significant postbypass coagulopathy since each unit is derived from one unit of FFP and thus replaces many hemostatic factors (eg, fibrinogen, factor VIII, factor XIII, von Willebrand factor [vWF], fibronectin) (table 7). However, cryoprecipitate is not available on mainland Europe.

Dosing with a 5 unit bag of cryoprecipitate will increase fibrinogen levels by approximately 35 to 50 mg/dL in a 70 kg adult, although this increase may be less if the patient is actively hemorrhaging [88]. As with fibrinogen concentrate, the plasma fibrinogen level is monitored, and repeat doses are administered to maintain the level above the appropriate threshold. (See "Intraoperative transfusion of blood products in adults", section on 'Cryoprecipitate'.)

Risks of cryoprecipitate transfusion are similar to those for FFP, although transfusion-associated circulatory overload (TACO) is less likely with cryoprecipitate due to lower total transfused volume. (See "Clinical use of Cryoprecipitate", section on 'Risks and adverse events'.)

Use of hemostatic agents — Hemostatic agents may be used to treat excessive bleeding in the postbypass period.

Antifibrinolytic agents — Timing for discontinuing administration of a prophylactic antifibrinolytic agent (eg, epsilon-aminocaproic acid [EACA] or tranexamic acid [TXA]) in the postbypass or postoperative period varies among centers. However, continuing administration is reasonable for patients with persistent bleeding [89,90]. (See "Blood management and anticoagulation for cardiopulmonary bypass", section on 'Antifibrinolytic administration'.)

Fibrinogen concentrate versus cryoprecipitate — We administer a fibrinogen concentrate, if available, to treat hypofibrinogenemia (eg, <100 mg/dL) in cardiac surgical patients with clinically significant bleeding and coagulopathy [40,91-99]. Fibrinogen concentrate can be used for treatment of hypofibrinogenemia rather than using cryoprecipitate or FFP (see 'Cryoprecipitate' above and 'Fresh frozen plasma' above). Fibrinogen concentrate is a pasteurized and pooled product that has a small volume and offers less risk of infection transmission and immunological complications of blood transfusion [99]. It is also a pharmaceutical product that can be stored and reconstituted at the point of care.

Fibrinogen concentrates (eg, RiaSTAP, Haemocomplettan, FIBRYGA) are prepared from pooled human plasma and are available as lyophilized powders (approximately 1 g [1000 mg]/vial) that are reconstituted in a small volume. Dose is calculated according to the target fibrinogen concentration; administration of 70 mg/kg increases fibrinogen concentration by approximately 100 mg/dL [100]. Dosing is typically monitored with measurements of plasma fibrinogen levels as well as POC testing to avoid hypercoagulability. (See "Intraoperative transfusion of blood products in adults", section on 'Point-of-care tests' and "Disorders of fibrinogen", section on 'Fibrinogen concentrate: Dosing and monitoring'.)  

Meta-analyses have found that administration of fibrinogen concentrate reduces transfusions in many types of cardiac surgical procedures, but effects on other outcomes such as survival are unclear:

A 2019 trial randomly assigned 735 cardiac surgical patients with clinically significant bleeding and documented hypofibrinogenemia (fibrinogen level <150 to 200 mg/dL) to receive 4 g of fibrinogen concentrate or 10 units of cryoprecipitate after CPB [101]. Both groups received a similar number of units of allogeneic blood component transfusions including RBCs, FFP, and platelets. The number of units of transfused allogeneic blood components including RBCs, FFP, and platelets was similar for patients receiving either treatment. Differences between the groups were not significant for other outcomes including mortality (9.4 percent in the fibrinogen concentrate group versus 7.4 percent in the cryoprecipitate group; hazard ratio [HR] 1.28; 95% CI 0.77-2.12) and thromboembolic complications (7.0 percent in the fibrinogen concentrate group versus 9.6 percent in the cryoprecipitate group; OR 0.70; 95% CI 0.42-1.20).

A 2018 meta-analysis of randomized trials (exclusively cardiac surgical patients) found that fibrinogen concentrate administered either prophylactically before CPB or during bleeding after CPB reduced the risk of RBC transfusion compared with placebo or other hemostatic treatments (risk ratio 0.64, 95% CI 0.49-0.83), but mortality was not reduced (597 patients; eight trials) [98]. Conclusions were limited by the small number of clinical events within each trial.

In two randomized trials in patients undergoing high-risk complex cardiac surgical procedures, prophylactic use of fibrinogen concentrate was not effective in reducing blood loss or transfusion of allogeneic blood products compared with placebo [102,103].

Administration of fibrinogen concentrate to treat severe perioperative bleeding is more common in Europe than in the United States, in part because cryoprecipitate is not typically available in Europe. The European Society of Anesthesia guidelines suggest use of fibrinogen concentrate to maintain a target fibrinogen concentration >150 to 200 mg/dL [104,105]. Similarly, the Hemostasis and Transfusion Scientific subcommittee of the European Association of Cardiothoracic Anaesthesiology suggests administration of fibrinogen concentrate in cardiac surgical patients with microvascular bleeding after CPB to maintain physiologic fibrinogen activity based on viscoelastic coagulation testing [99]. Reduced serum concentrations of fibrinogen may predict bleeding since it is the first substrate component of the coagulation cascade to be consumed below critical levels in hemorrhaging patients [106]. Proponents argue that the baseline concentration of fibrinogen is relatively low, and there are no fibrinogen stores to be mobilized [91,107]. Thus, fibrinogen is the first coagulation protein to become critically low during intraoperative bleeding, and it may be the most critical coagulation component required for local hemostasis [88,106]. However, guidelines do not suggest aiming for supranormal fibrinogen levels [99]. Notably, administration of fibrinogen concentrate as a single agent will only raise the fibrinogen level and will not address other coagulation factor deficiencies. For this reason, efficacy compared with placebo has not been shown when fibrinogen concentrate is administered prophylactically in patients with expected significant intraoperative bleeding during complex cardiac surgical procedures [102,103]. While administration of fibrinogen concentrate will not always resolve coagulopathic bleeding, we aggressively correct acquired hypofibrinogenemia in patients with clinically significant bleeding as a component of a multifactorial transfusion algorithm [58]. Some clinicians prefer cryoprecipitate in this setting since each unit contains many hemostatic factors (eg, fibrinogen, factor VIII, factor XIII, von Willebrand factor [vWF], fibronectin), as noted above. (See 'Cryoprecipitate' above.)

Although thromboembolic complications can occur, particularly in patients with a thrombotic fibrinogen variant (or pregnancy), most studies have not reported increased risk of thrombotic events [96,98,99]. Monitoring to avoid overcorrection of fibrinogen deficiency minimizes this risk.

Prothrombin complex concentrate (PCC)

PCCs – A 4-factor unactivated prothrombin complex concentrate (PCC) is typically used for emergency cardiac surgery in patients chronically taking warfarin or another vitamin K antagonist [40,108]. All PCCs contain factors II, IX and X. Those that do not contain appreciable factor VII are known as 3-factor PCCs, while those containing factor VII are labeled as 4-factor PCCs (table 8). Advantages of PCCs over FFP include rapid administration in a small volume, resulting in more rapid reversal of the anticoagulant effect and avoidance of volume overload and transfusion reactions [91,93,108-113]. (See "Plasma derivatives and recombinant DNA-produced coagulation factors", section on 'PCCs'.)

For patients with intractable coagulopathy and diffuse bleeding or for patients intolerant of high FFP transfusion volume, off-label intraoperative use of unactivated 4-factor or 3-factor PCC products after cardiac surgery has been evaluated in observational studies [40,91,109,110,114-119]. However, other primary causes such as thrombocytopenia or low levels of fibrinogen due to hemodilution, platelet dysfunction, or surgical sources of bleeding should be sought and treated before considering a PCC product for such intractable coagulopathy [109,120]. Also, consider that PCC does contain antithrombin III, while FFP does. Finally, risks of PCC administration include thromboembolic events, which may be more likely with repeat or excessive dosing of both 4-factor and 3-factor PCCs, and may extend well into the postoperative period [121].

Data regarding intraoperative safety of PCC products in cardiac surgical patients are limited [104,109,119,122,123]:

A 2019 meta-analysis that included 861 patients in four nonrandomized studies in cardiac surgical patients noted a reduced risk of RBC transfusion with administration of perioperative PCC compared with administration of FFP (OR 2.22, 95% CI 1.45-3.40) [119]. There were no differences in the risk of in-hospital mortality, stroke, or acute kidney injury between patients receiving PCC versus FFP (although there was a trend toward a higher risk of renal replacement therapy in the PCC group).

A subsequently published retrospective study analyzed 114 consecutive patients who received a low dose of 3-factor PCC (ie, 15.8 ± 7.1 international units/kg) for refractory bleeding during complex cardiac surgical procedure as part of a transfusion algorithm guided by standard and POC laboratory testing (see 'Use of transfusion algorithms' above) [123]. Administration of the 3-factor PCC reduced transfusions of blood products compared with the period before PCC administration. However, four patients receiving PCC suffered an ischemic stroke within 30 days of surgery (3.8 percent), while seven had a venous thromboembolic event (6.5 percent) [123].

As more patients are entered into registries for 4-factor and 3-factor PCC product use, risk assessment for such dose-related adverse events will be more certain.

Activated PCCs – In contrast with unactivated PCCs, activated PCC products such as factor eight inhibitor bypassing activity (FEIBA) contain activated factor VII (table 8) [109]. These agents have a greater prothrombotic risk compared with unactivated PCC products, and are only rarely used. (See "Plasma derivatives and recombinant DNA-produced coagulation factors", section on 'PCCs'.)

Recombinant activated factor VII (rFVIIa) — Recombinant activated factor VII (rFVIIa) is licensed for prevention of surgical bleeding in patients with hemophilia. In rare instances when intractable life-threatening bleeding persists after cardiac surgery with CPB, rFVIIa has been administered and may achieve bleeding cessation and reduce transfusion requirements in that setting [40,124-126]. However, high thromboembolism rates >20 percent (including myocardial infarction) with mortality >30 percent have been described in retrospective evaluations of registries for refractory bleeding cases [127-129]. (See "Recombinant factor VIIa: Administration and adverse effects", section on 'Off-label uses' and "Perioperative blood management: Strategies to minimize transfusions", section on 'Recombinant activated factor VII'.)

Although the optimal dose of rFVIIa for off-label use is unknown, relatively low doses may achieve bleeding cessation and are preferred in most cardiac surgical patients with life-threatening bleeding [40]. One study reported that a median dose of 13.3 mcg/kg resulted in low overall blood transfusions in this setting [125]. A cautious dosing strategy is necessary in the rare instances when rFVIIa is employed. For example, small incremental doses of 10 mcg/kg can be administered approximately every 15 minutes, while recognizing that thromboembolic complications will likely increase with dose escalation, or in the presence of stagnant flow or devices such as extracorporeal membrane oxygenation (ECMO). Stocking the smaller 1 mg vials of rFVIIa facilitates this incremental approach.

Similar to off-label use of PCCs, other primary causes of coagulopathy should always be sought and treated before administering rFVIIa. Failure to treat the primary coagulation defect increases the likelihood of an inadequate response to the initial dose of rFVIIa [127,128,130], and may encourage use of higher doses that are more likely to cause thrombosis [129,131]. (See "Recombinant factor VIIa: Administration and adverse effects", section on 'General approach to administration'.)

Desmopressin (DDAVP) — We administer intravenous (IV) desmopressin (DDAVP) 0.3 mcg/kg when persistent microvascular bleeding is evident after CPB in selected patients with acquired platelet defects due to uremia, or to those with acquired von Willebrand syndrome (eg, due to chronic aortic stenosis or presence of a left ventricular assist device) [91,132-135]. DDAVP should be infused slowly over 15 to 30 minutes to avoid vasodilation. (See "Platelet dysfunction in uremia", section on 'Desmopressin (DDAVP)' and "Acquired von Willebrand syndrome", section on 'Treatment of acute bleeding'.)

Very limited data suggest that IV DDAVP may be beneficial to reduce blood loss in some cardiac surgical patients with intractable microvascular bleeding due to platelet dysfunction that may be caused by hypothermia, acidosis, aspirin use, and/or the effects of CPB [40,136-139]. However, studies are not consistent [140,141]. Two 2017 meta-analyses of the efficacy of DDAVP in patients undergoing cardiac or noncardiac surgery noted only small reductions in perioperative blood loss and volume of RBC transfusions compared with placebo, but quality of evidence was low [138,139]. Thus, generalized perioperative use in cardiac surgical patients is not warranted [40,54,91].

Possible adverse side effects of DDAVP include hypertension, hypotension, and flushing, as well as fluid overload, hyponatremia (which may cause seizures if close attention to free water restriction is not used), and rare thrombotic events (table 9). Tachyphylaxis occurs after the initial dose and during the initial three to five days of administration.

EARLY POSTOPERATIVE MANAGEMENT

Management of bleeding and coagulopathy — Close monitoring for bleeding continues in the postoperative period. Medical causes of bleeding should be investigated and treated [142].

Return to the operating room for surgical re-exploration and intervention may be necessary based on the rate, and presumed location of bleeding, as well as potential for a surgical cause [143,144]. (See "Perioperative blood management: Strategies to minimize transfusions", section on 'Close monitoring for bleeding'.)

Management of anemia — Postoperative anemia is common due to exacerbation of pre-existing anemia, blood loss during surgery, and excessive postoperative phlebotomies [86]. Management strategies are discussed separately (algorithm 2). (See "Perioperative blood management: Strategies to minimize transfusions", section on 'Management of postoperative anemia'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Transfusion and patient blood management" and "Society guideline links: Management of cardiopulmonary bypass".)

SUMMARY AND RECOMMENDATIONS

Systemic heparin anticoagulation is reversed with administration of protamine. (See 'Administration and dosing of protamine' above.)

Timing of initial administration – Neutralization of heparin is initiated after weaning from cardiopulmonary bypass (CPB), but before aortic decannulation. An initial "test dose" of protamine 10 mg is administered to allow early detection of serious hemodynamic changes indicative of a protamine reaction. The remainder of the protamine dose is administered slowly, typically over a 5 to 15 minute period.

Initial dosing – Calculation of protamine dosing for heparin reversal is based on a point-of-care (POC) titration to existing heparin in the blood, or by administering 0.7 mg protamine per 100 units of the initial pre-bypass heparin dose. Additional protamine is administered according to assay measurements of residual heparin, or an additional 25 to 100 mg of protamine may be administered if evidence of residual heparin effect is noted on the activated clotting time (ACT) measurement or with viscoelastic testing. In most cases, there is no reason to exceed 1 mg protamine per 100 units of total administered heparin; in practice, we rarely administer more than 250 mg of protamine.

Postbypass and postoperative protamine administration – We administer a protamine infusion of 25 to 50 mg/hour over two to four postoperative hours to avoid "heparin rebound." Release of heparin from the fatty tissues into the intravascular space (ie, "heparin rebound") may occur after the initial protamine dose has been metabolized in patients who received a large dose of heparin.

Vasodilation due to direct and indirect effects of protamine commonly occur, particularly if administration is rapid. Such reactions are treated with administration of vasopressors as needed (table 1). More severe reactions (ie, anaphylaxis and acute pulmonary vasoconstriction) are uncommon; treatment is described in the table (table 2). (See 'Management of protamine reactions' above.)

Active warming techniques to achieve and maintain normothermia are employed to minimize coagulopathy and blood loss. (See 'Maintenance of normothermia' above.)

Causes of excessive bleeding during and after weaning from CPB include inadequate surgical hemostasis, loss of platelets and coagulation factors due to persistent surgical bleeding, effects of hemodilution, presence of residual heparin, hypothermic coagulopathy, and coagulopathy due to hyperfibrinolysis and platelet activation and consumption induced by the extracorporeal circuit. Meticulous surgical techniques and systematic intraoperative checking of potential surgical sites of bleeding is critically important in the postbypass period. (See 'Management of bleeding' above and 'Fastidious surgical hemostasis' above and 'Maintenance of normothermia' above.)

We use a goal-directed protocol or algorithm to guide transfusion decisions, based on measurement of hemoglobin (Hgb) or hematocrit (Hct) as well as assessment of specific abnormalities of hemostasis using standard laboratory tests (algorithm 1), and/or point-of care tests of hemostasis, similar to the practice guidelines of several professional societies. Use of such transfusion protocols and algorithms to guide decision-making in cardiac surgery can avoid or reduce unnecessary transfusions of blood products. (See 'Use of transfusion algorithms' above.)

Transfusion of blood components is based on the following parameters (see 'Transfusion of blood products' above):

Red blood cells (RBCs) – We typically transfuse RBCs for Hgb <7 to 8 g/dL (or Hct < 21 to 24 percent). However, transfusion decisions are individualized, taking into account patient-related factors (eg, age, severity of illness, cardiac function, risk for critical end-organ ischemia), the clinical setting (massive or active blood loss), laboratory parameters (eg, low mixed venous oxygen saturation), or evidence of myocardial ischemia. (See 'Red blood cells' above.)

Fresh frozen plasma (FFP) – FFP is transfused when persistent bleeding is accompanied by a prothrombin time (PT), activated partial thromboplastin time (aPTT), or international normalized ratio (INR) >1.5 times normal value. (See 'Fresh frozen plasma' above.)

Platelets – Transfusion of platelets is reserved for patients with platelet count <100,000/mm3 and ongoing clinically significant microvascular bleeding. (See 'Platelets' above.)

Cryoprecipitate – Cryoprecipitate can be used to treat hypofibrinogenemia (eg, <100 mg/dL) in a patient with intractable bleeding and coagulopathy after CPB, if fibrinogen concentrate is not available. Some clinicians prefer cryoprecipitate in this setting since each unit is derived from one unit of FFP and is a concentrate of many hemostatic factors (eg, fibrinogen, factor VIII, factor XIII, von Willebrand factor [vWF], fibronectin). (See 'Cryoprecipitate' above and 'Fibrinogen concentrate versus cryoprecipitate' above.)

Hemostatic agents may be used to treat intractable bleeding and coagulopathy after CPB in selected patients:

Fibrinogen concentrate versus cryoprecipitate – When hypofibrinogenemia is present in a patient with clinically significant bleeding and coagulopathy, a fibrinogen concentrate can be administered. Fibrinogen concentrate or cryoprecipitate is preferred rather than FFP to treat hypofibrinogenemia to avoid volume overload. Fibrinogen concentrate also has little risk of infection transmission or immunological complications of blood transfusion. However, administration of fibrinogen concentrate will only raise the fibrinogen level and will not address other coagulation factor deficiencies. (See 'Fibrinogen concentrate versus cryoprecipitate' above and 'Cryoprecipitate' above.)

Prothrombin complex concentrate (PCC) – For patients with intractable coagulopathy and diffuse bleeding after CPB, or for patients intolerant of high FFP transfusion volume, off-label intraoperative use of unactivated 4-factor or 3-factor PCC products may be considered. However, other primary causes of bleeding such as thrombocytopenia, hypofibrinogenemia due to hemodilution, platelet dysfunction, or surgical sources of bleeding should be sought and treated before considering a PCC product. Risks include thromboembolic events, which may be more likely with repeat or excessive dosing of both 4-factor and 3-factor PCCs, and may extend well into the postoperative period. (See 'Prothrombin complex concentrate (PCC)' above.)

Recombinant activated factor VII (rFVIIa) – In rare instances when intractable life-threatening bleeding persists after cardiac surgery, rFVIIa has been administered to achieve bleeding cessation. However, retrospective evaluations have noted high thromboembolism rates after administration. Thus, other primary causes of coagulopathy should always be sought and treated before considering rFVIIa. (See 'Recombinant activated factor VII (rFVIIa)' above.)

Desmopressin (DDAVP) – We administer intravenous (IV) DDAVP 0.3 mcg/kg when persistent microvascular bleeding is evident after CPB in selected patients with acquired platelet defects due to uremia or those with acquired von Willebrand disease (eg, due to chronic aortic stenosis or presence of a left ventricular assist device). (See 'Desmopressin (DDAVP)' above.)

REFERENCES

  1. Sniecinski RM, Chandler WL. Activation of the hemostatic system during cardiopulmonary bypass. Anesth Analg 2011; 113:1319.
  2. Despotis GJ, Avidan MS, Hogue CW Jr. Mechanisms and attenuation of hemostatic activation during extracorporeal circulation. Ann Thorac Surg 2001; 72:S1821.
  3. Wahba A, Milojevic M, Boer C, et al. 2019 EACTS/EACTA/EBCP guidelines on cardiopulmonary bypass in adult cardiac surgery. Eur J Cardiothorac Surg 2020; 57:210.
  4. Welsby IJ, Newman MF, Phillips-Bute B, et al. Hemodynamic changes after protamine administration: association with mortality after coronary artery bypass surgery. Anesthesiology 2005; 102:308.
  5. Wakefield TW, Hantler CB, Wrobleski SK, et al. Effects of differing rates of protamine reversal of heparin anticoagulation. Surgery 1996; 119:123.
  6. Despotis GJ, Gravlee G, Filos K, Levy J. Anticoagulation monitoring during cardiac surgery: a review of current and emerging techniques. Anesthesiology 1999; 91:1122.
  7. Shore-Lesserson L, Baker RA, Ferraris VA, et al. The Society of Thoracic Surgeons, The Society of Cardiovascular Anesthesiologists, and The American Society of ExtraCorporeal Technology: Clinical Practice Guidelines-Anticoagulation During Cardiopulmonary Bypass. Anesth Analg 2018; 126:413.
  8. Abuelkasem E, Mazzeffi MA, Henderson RA, et al. Clinical Impact of Protamine Titration-Based Heparin Neutralization in Patients Undergoing Coronary Bypass Grafting Surgery. J Cardiothorac Vasc Anesth 2019; 33:2153.
  9. Guo Y, Tang J, Du L, et al. Protamine dosage based on two titrations reduces blood loss after valve replacement surgery: a prospective, double-blinded, randomized study. Can J Cardiol 2012; 28:547.
  10. Vonk AB, Veerhoek D, van den Brom CE, et al. Individualized heparin and protamine management improves rotational thromboelastometric parameters and postoperative hemostasis in valve surgery. J Cardiothorac Vasc Anesth 2014; 28:235.
  11. Koster A, Börgermann J, Gummert J, et al. Protamine overdose and its impact on coagulation, bleeding, and transfusions after cardiopulmonary bypass: results of a randomized double-blind controlled pilot study. Clin Appl Thromb Hemost 2014; 20:290.
  12. Wang J, Ma HP, Zheng H. Blood loss after cardiopulmonary bypass, standard vs titrated protamine: a meta-analysis. Neth J Med 2013; 71:123.
  13. Boer C, Meesters MI, Veerhoek D, Vonk ABA. Anticoagulant and side-effects of protamine in cardiac surgery: a narrative review. Br J Anaesth 2018; 120:914.
  14. Dunning J, Versteegh M, Fabbri A, et al. Guideline on antiplatelet and anticoagulation management in cardiac surgery. Eur J Cardiothorac Surg 2008; 34:73.
  15. Mochizuki T, Olson PJ, Szlam F, et al. Protamine reversal of heparin affects platelet aggregation and activated clotting time after cardiopulmonary bypass. Anesth Analg 1998; 87:781.
  16. Ni Ainle F, Preston RJ, Jenkins PV, et al. Protamine sulfate down-regulates thrombin generation by inhibiting factor V activation. Blood 2009; 114:1658.
  17. Carr ME Jr, Carr SL. At high heparin concentrations, protamine concentrations which reverse heparin anticoagulant effects are insufficient to reverse heparin anti-platelet effects. Thromb Res 1994; 75:617.
  18. Shore-Lesserson L, Baker RA, Ferraris VA, et al. The Society of Thoracic Surgeons, The Society of Cardiovascular Anesthesiologists, and The American Society of ExtraCorporeal Technology: Clinical Practice Guidelines-Anticoagulation During Cardiopulmonary Bypass. Ann Thorac Surg 2018; 105:650.
  19. Teoh KH, Young E, Blackall MH, et al. Can extra protamine eliminate heparin rebound following cardiopulmonary bypass surgery? J Thorac Cardiovasc Surg 2004; 128:211.
  20. Jia Z, Tian G, Ren Y, et al. Pharmacokinetic model of unfractionated heparin during and after cardiopulmonary bypass in cardiac surgery. J Transl Med 2015; 13:45.
  21. Butterworth J, Lin YA, Prielipp RC, et al. Rapid disappearance of protamine in adults undergoing cardiac operation with cardiopulmonary bypass. Ann Thorac Surg 2002; 74:1589.
  22. Frick PG, Brögli H. The mechansim of heparin rebound after extracorporeal circulation for open cardiac surgery. Surgery 1966; 59:721.
  23. Galeone A, Rotunno C, Guida P, et al. Monitoring incomplete heparin reversal and heparin rebound after cardiac surgery. J Cardiothorac Vasc Anesth 2013; 27:853.
  24. Taneja R, Marwaha G, Sinha P, et al. Elevated activated partial thromboplastin time does not correlate with heparin rebound following cardiac surgery. Can J Anaesth 2009; 56:489.
  25. Horrow JC. Protamine allergy. J Cardiothorac Anesth 1988; 2:225.
  26. Horrow JC. Protamine: a review of its toxicity. Anesth Analg 1985; 64:348.
  27. Levy JH, Bartz RR. Protamine, is something fishy about it? The spectre of anaphylaxis continues. J Cardiothorac Vasc Anesth 2019; 33:487.
  28. Weiss ME, Nyhan D, Peng ZK, et al. Association of protamine IgE and IgG antibodies with life-threatening reactions to intravenous protamine. N Engl J Med 1989; 320:886.
  29. Lowenstein E, Zapol WM. Protamine reactions, explosive mediator release, and pulmonary vasoconstriction. Anesthesiology 1990; 73:373.
  30. Valchanov K, Falter F, George S, et al. Three Cases of Anaphylaxis to Protamine: Management of Anticoagulation Reversal. J Cardiothorac Vasc Anesth 2019; 33:482.
  31. Lock R, Hessel EA 2nd. Probable reversal of protamine reactions by heparin administration. J Cardiothorac Anesth 1990; 4:604.
  32. Comunale ME, Maslow A, Robertson LK, et al. Effect of site of venous protamine administration, previously alleged risk factors, and preoperative use of aspirin on acute protamine-induced pulmonary vasoconstriction. J Cardiothorac Vasc Anesth 2003; 17:309.
  33. Morel DR, Zapol WM, Thomas SJ, et al. C5a and thromboxane generation associated with pulmonary vaso- and broncho-constriction during protamine reversal of heparin. Anesthesiology 1987; 66:597.
  34. Levy JH, Adkinson NF Jr. Anaphylaxis during cardiac surgery: implications for clinicians. Anesth Analg 2008; 106:392.
  35. Del Duca D, Sheth SS, Clarke AE, et al. Use of methylene blue for catecholamine-refractory vasoplegia from protamine and aprotinin. Ann Thorac Surg 2009; 87:640.
  36. Albuquerque AA, Margarido EA, Menardi AC, et al. Methylene Blue to Treat Protamine-induced Anaphylaxis Reactions. An Experimental Study in Pigs. Braz J Cardiovasc Surg 2016; 31:226.
  37. Viaro F, Dalio MB, Evora PR. Catastrophic cardiovascular adverse reactions to protamine are nitric oxide/cyclic guanosine monophosphate dependent and endothelium mediated: should methylene blue be the treatment of choice? Chest 2002; 122:1061.
  38. Turan A, Yang D, Bonilla A, et al. Morbidity and mortality after massive transfusion in patients undergoing non-cardiac surgery. Can J Anaesth 2013; 60:761.
  39. Bennett-Guerrero E, Zhao Y, O'Brien SM, et al. Variation in use of blood transfusion in coronary artery bypass graft surgery. JAMA 2010; 304:1568.
  40. Raphael J, Mazer CD, Subramani S, et al. Society of Cardiovascular Anesthesiologists Clinical Practice Improvement Advisory for Management of Perioperative Bleeding and Hemostasis in Cardiac Surgery Patients. Anesth Analg 2019; 129:1209.
  41. Society of Thoracic Surgeons Blood Conservation Guideline Task Force, Ferraris VA, Brown JR, et al. 2011 update to the Society of Thoracic Surgeons and the Society of Cardiovascular Anesthesiologists blood conservation clinical practice guidelines. Ann Thorac Surg 2011; 91:944.
  42. Society of Thoracic Surgeons Blood Conservation Guideline Task Force, Ferraris VA, Ferraris SP, et al. Perioperative blood transfusion and blood conservation in cardiac surgery: the Society of Thoracic Surgeons and The Society of Cardiovascular Anesthesiologists clinical practice guideline. Ann Thorac Surg 2007; 83:S27.
  43. Dhir A, Tempe DK. Anemia and Patient Blood Management in Cardiac Surgery-Literature Review and Current Evidence. J Cardiothorac Vasc Anesth 2018; 32:2726.
  44. Pleym H, Wahba A, Videm V, et al. Increased fibrinolysis and platelet activation in elderly patients undergoing coronary bypass surgery. Anesth Analg 2006; 102:660.
  45. Hajjar LA, Vincent JL, Galas FR, et al. Transfusion requirements after cardiac surgery: The TRACS randomized controlled trial. JAMA 2010; 304:1559.
  46. Biancari F, Kinnunen EM, Kiviniemi T, et al. Meta-analysis of the Sources of Bleeding after Adult Cardiac Surgery. J Cardiothorac Vasc Anesth 2018; 32:1618.
  47. Biancari F, Mikkola R, Heikkinen J, et al. Estimating the risk of complications related to re-exploration for bleeding after adult cardiac surgery: a systematic review and meta-analysis. Eur J Cardiothorac Surg 2012; 41:50.
  48. Martini WZ. Coagulopathy by hypothermia and acidosis: mechanisms of thrombin generation and fibrinogen availability. J Trauma 2009; 67:202.
  49. Wolberg AS, Meng ZH, Monroe DM 3rd, Hoffman M. A systematic evaluation of the effect of temperature on coagulation enzyme activity and platelet function. J Trauma 2004; 56:1221.
  50. Valeri CR, Khabbaz K, Khuri SF, et al. Effect of skin temperature on platelet function in patients undergoing extracorporeal bypass. J Thorac Cardiovasc Surg 1992; 104:108.
  51. Rajagopalan S, Mascha E, Na J, Sessler DI. The effects of mild perioperative hypothermia on blood loss and transfusion requirement. Anesthesiology 2008; 108:71.
  52. Reed RL 2nd, Johnson TD, Hudson JD, Fischer RP. The disparity between hypothermic coagulopathy and clotting studies. J Trauma 1992; 33:465.
  53. Lester ELW, Fox EE, Holcomb JB, et al. The impact of hypothermia on outcomes in massively transfused patients. J Trauma Acute Care Surg 2019; 86:458.
  54. American Society of Anesthesiologists Task Force on Perioperative Blood Management. Practice guidelines for perioperative blood management: an updated report by the American Society of Anesthesiologists Task Force on Perioperative Blood Management*. Anesthesiology 2015; 122:241.
  55. Mueller MM, Van Remoortel H, Meybohm P, et al. Patient Blood Management: Recommendations From the 2018 Frankfurt Consensus Conference. JAMA 2019; 321:983.
  56. Kozek-Langenecker SA, Ahmed AB, Afshari A, et al. Management of severe perioperative bleeding: guidelines from the European Society of Anaesthesiology: First update 2016. Eur J Anaesthesiol 2017; 34:332.
  57. Curry NS, Davenport R, Pavord S, et al. The use of viscoelastic haemostatic assays in the management of major bleeding: A British Society for Haematology Guideline. Br J Haematol 2018; 182:789.
  58. Ghadimi K, Levy JH, Welsby IJ. Perioperative management of the bleeding patient. Br J Anaesth 2016; 117:iii18.
  59. Karkouti K, Callum J, Wijeysundera DN, et al. Point-of-Care Hemostatic Testing in Cardiac Surgery: A Stepped-Wedge Clustered Randomized Controlled Trial. Circulation 2016; 134:1152.
  60. Weber CF, Görlinger K, Meininger D, et al. Point-of-care testing: a prospective, randomized clinical trial of efficacy in coagulopathic cardiac surgery patients. Anesthesiology 2012; 117:531.
  61. 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; 92:178.
  62. Spalding GJ, Hartrumpf M, Sierig T, et al. Cost reduction of perioperative coagulation management in cardiac surgery: value of "bedside" thrombelastography (ROTEM). Eur J Cardiothorac Surg 2007; 31:1052.
  63. Girdauskas E, Kempfert J, Kuntze T, et al. Thromboelastometrically guided transfusion protocol during aortic surgery with circulatory arrest: a prospective, randomized trial. J Thorac Cardiovasc Surg 2010; 140:1117.
  64. Hanke AA, Herold U, Dirkmann D, et al. Thromboelastometry Based Early Goal-Directed Coagulation Management Reduces Blood Transfusion Requirements, Adverse Events, and Costs in Acute Type A Aortic Dissection: A Pilot Study. Transfus Med Hemother 2012; 39:121.
  65. Karkouti K, McCluskey SA, Callum J, et al. Evaluation of a novel transfusion algorithm employing point-of-care coagulation assays in cardiac surgery: a retrospective cohort study with interrupted time-series analysis. Anesthesiology 2015; 122:560.
  66. Görlinger K, Fries D, Dirkmann D, et al. Reduction of Fresh Frozen Plasma Requirements by Perioperative Point-of-Care Coagulation Management with Early Calculated Goal-Directed Therapy. Transfus Med Hemother 2012; 39:104.
  67. Fabbro M 2nd, Winkler AM, Levy JH. Technology: Is There Sufficient Evidence to Change Practice in Point-of-Care Management of Coagulopathy? J Cardiothorac Vasc Anesth 2017; 31:1849.
  68. Kuiper GJAJM, van Egmond LT, Henskens YMC, et al. Shifts of Transfusion Demand in Cardiac Surgery After Implementation of Rotational Thromboelastometry-Guided Transfusion Protocols: Analysis of the HEROES-CS (HEmostasis Registry of patiEntS in Cardiac Surgery) Observational, Prospective Open Cohort Database. J Cardiothorac Vasc Anesth 2019; 33:307.
  69. Le Quellec S, Bordet JC, Negrier C, Dargaud Y. Comparison of current platelet functional tests for the assessment of aspirin and clopidogrel response. A review of the literature. Thromb Haemost 2016; 116:638.
  70. Wikkelsø A, Wetterslev J, Møller AM, Afshari A. Thromboelastography (TEG) or thromboelastometry (ROTEM) to monitor haemostatic treatment versus usual care in adults or children with bleeding. Cochrane Database Syst Rev 2016; :CD007871.
  71. Serraino GF, Murphy GJ. Routine use of viscoelastic blood tests for diagnosis and treatment of coagulopathic bleeding in cardiac surgery: updated systematic review and meta-analysis. Br J Anaesth 2017; 118:823.
  72. Wikkelsø A, Wetterslev J, Møller AM, Afshari A. Thromboelastography (TEG) or rotational thromboelastometry (ROTEM) to monitor haemostatic treatment in bleeding patients: a systematic review with meta-analysis and trial sequential analysis. Anaesthesia 2017; 72:519.
  73. Deppe AC, Weber C, Zimmermann J, et al. Point-of-care thromboelastography/thromboelastometry-based coagulation management in cardiac surgery: a meta-analysis of 8332 patients. J Surg Res 2016; 203:424.
  74. Westbrook AJ, Olsen J, Bailey M, et al. Protocol based on thromboelastograph (TEG) out-performs physician preference using laboratory coagulation tests to guide blood replacement during and after cardiac surgery: a pilot study. Heart Lung Circ 2009; 18:277.
  75. Görlinger K, Dirkmann D, Hanke AA, et al. First-line therapy with coagulation factor concentrates combined with point-of-care coagulation testing is associated with decreased allogeneic blood transfusion in cardiovascular surgery: a retrospective, single-center cohort study. Anesthesiology 2011; 115:1179.
  76. Patel NN, Murphy GJ. Transfusion triggers in cardiac surgery: Where do we go from here? Can J Anaesth 2018; 65:868.
  77. Mazer CD, Whitlock RP, Fergusson DA, et al. Restrictive or liberal red-cell transfusion for cardiac surgery. N Engl J Med 2017; 377:2133.
  78. Mazer CD, Whitlock RP, Fergusson DA, et al. Six-Month Outcomes after Restrictive or Liberal Transfusion for Cardiac Surgery. N Engl J Med 2018; 379:1224.
  79. Shehata N, Mistry N, da Costa BR, et al. Restrictive compared with liberal red cell transfusion strategies in cardiac surgery: a meta-analysis. Eur Heart J 2019; 40:1081.
  80. Murphy GJ, Pike K, Rogers CA, et al. Liberal or restrictive transfusion after cardiac surgery. N Engl J Med 2015; 372:997.
  81. Koch CG, Sessler DI, Mascha EJ, et al. A randomized clinical trial of red blood cell transfusion triggers in cardiac surgery. Ann Thorac Surg 2017; 104:1243.
  82. Chong MA, Krishnan R, Cheng D, Martin J. Should Transfusion Trigger Thresholds Differ for Critical Care Versus Perioperative Patients? A Meta-Analysis of Randomized Trials. Crit Care Med 2018; 46:252.
  83. LaPar DJ, Hawkins RB, McMurry TL, et al. Preoperative anemia versus blood transfusion: Which is the culprit for worse outcomes in cardiac surgery? J Thorac Cardiovasc Surg 2018; 156:66.
  84. Tantawy H, Li A, Dai F, et al. Association of red blood cell transfusion and short- and longer-term mortality after coronary artery bypass graft surgery. J Cardiothorac Vasc Anesth 2018; 32:1225.
  85. Dixon B, Santamaria JD, Reid D, et al. The association of blood transfusion with mortality after cardiac surgery: cause or confounding? (CME). Transfusion 2013; 53:19.
  86. Burns CD, Brown JP, Corwin HL, et al. Special Report From the Society for the Advancement of Blood Management: The Choosing Wisely Campaign. Anesth Analg 2019; 129:1381.
  87. Delaney M, Stark PC, Suh M, et al. Massive Transfusion in Cardiac Surgery: The Impact of Blood Component Ratios on Clinical Outcomes and Survival. Anesth Analg 2017; 124:1777.
  88. Levy JH, Welsby I, Goodnough LT. Fibrinogen as a therapeutic target for bleeding: a review of critical levels and replacement therapy. Transfusion 2014; 54:1389.
  89. Lier H, Maegele M, Shander A. Tranexamic Acid for Acute Hemorrhage: A Narrative Review of Landmark Studies and a Critical Reappraisal of Its Use Over the Last Decade. Anesth Analg 2019; 129:1574.
  90. Faraoni D, Levy JH. Tranexamic Acid for Acute Hemorrhage: When Is Enough Evidence Enough? Anesth Analg 2019; 129:1459.
  91. Pagano D, Milojevic M, Meesters MI, et al. 2017 EACTS/EACTA Guidelines on patient blood management for adult cardiac surgery. Eur J Cardiothorac Surg 2018; 53:79.
  92. Ranucci M, Baryshnikova E, Crapelli GB, et al. Randomized, double-blinded, placebo-controlled trial of fibrinogen concentrate supplementation after complex cardiac surgery. J Am Heart Assoc 2015; 4:e002066.
  93. Ghadimi K, Welsby IJ. Pro: Factor Concentrates are Essential for Hemostasis in Complex Cardiac Surgery. J Cardiothorac Vasc Anesth 2018; 32:558.
  94. Rahe-Meyer N, Solomon C, Hanke A, et al. Effects of fibrinogen concentrate as first-line therapy during major aortic replacement surgery: a randomized, placebo-controlled trial. Anesthesiology 2013; 118:40.
  95. Wikkelsø A, Lunde J, Johansen M, et al. Fibrinogen concentrate in bleeding patients. Cochrane Database Syst Rev 2013; :CD008864.
  96. Fominskiy E, Nepomniashchikh VA, Lomivorotov VV, et al. Efficacy and Safety of Fibrinogen Concentrate in Surgical Patients: A Meta-Analysis of Randomized Controlled Trials. J Cardiothorac Vasc Anesth 2016; 30:1196.
  97. Maeda T, Miyata S, Usui A, et al. Safety of Fibrinogen Concentrate and Cryoprecipitate in Cardiovascular Surgery: Multicenter Database Study. J Cardiothorac Vasc Anesth 2019; 33:321.
  98. Li JY, Gong J, Zhu F, et al. Fibrinogen Concentrate in Cardiovascular Surgery: A Meta-analysis of Randomized Controlled Trials. Anesth Analg 2018; 127:612.
  99. Erdoes G, Koster A, Meesters MI, et al. The role of fibrinogen and fibrinogen concentrate in cardiac surgery: an international consensus statement from the Haemostasis and Transfusion Scientific Subcommittee of the European Association of Cardiothoracic Anaesthesiology. Anaesthesia 2019; 74:1589.
  100. Hanna JM, Keenan JE, Wang H, et al. Use of human fibrinogen concentrate during proximal aortic reconstruction with deep hypothermic circulatory arrest. J Thorac Cardiovasc Surg 2016; 151:376.
  101. Callum J, Farkouh ME, Scales DC, et al. Effect of Fibrinogen Concentrate vs Cryoprecipitate on Blood Component Transfusion After Cardiac Surgery: The FIBRES Randomized Clinical Trial. JAMA 2019; 322:1966.
  102. Bilecen S, de Groot JA, Kalkman CJ, et al. Effect of Fibrinogen Concentrate on Intraoperative Blood Loss Among Patients With Intraoperative Bleeding During High-Risk Cardiac Surgery: A Randomized Clinical Trial. JAMA 2017; 317:738.
  103. Rahe-Meyer N, Levy JH, Mazer CD, et al. Randomized evaluation of fibrinogen vs placebo in complex cardiovascular surgery (REPLACE): a double-blind phase III study of haemostatic therapy. Br J Anaesth 2016; 117:41.
  104. Kozek-Langenecker SA, Afshari A, Albaladejo P, et al. Management of severe perioperative bleeding: guidelines from the European Society of Anaesthesiology. Eur J Anaesthesiol 2013; 30:270.
  105. Levy JH, Goodnough LT. How I use fibrinogen replacement therapy in acquired bleeding. Blood 2015; 125:1387.
  106. Blome M, Isgro F, Kiessling AH, et al. Relationship between factor XIII activity, fibrinogen, haemostasis screening tests and postoperative bleeding in cardiopulmonary bypass surgery. Thromb Haemost 2005; 93:1101.
  107. Hiippala ST, Myllylä GJ, Vahtera EM. Hemostatic factors and replacement of major blood loss with plasma-poor red cell concentrates. Anesth Analg 1995; 81:360.
  108. Levy JH, Douketis J, Steiner T, et al. Prothrombin Complex Concentrates for Perioperative Vitamin K Antagonist and Non-vitamin K Anticoagulant Reversal. Anesthesiology 2018; 129:1171.
  109. Ghadimi K, Levy JH, Welsby IJ. Prothrombin Complex Concentrates for Bleeding in the Perioperative Setting. Anesth Analg 2016; 122:1287.
  110. Smith MM, Ashikhmina E, Brinkman NJ, Barbara DW. Perioperative Use of Coagulation Factor Concentrates in Patients Undergoing Cardiac Surgery. J Cardiothorac Vasc Anesth 2017; 31:1810.
  111. Goldhammer JE, Herman CR. A Split From Conventional Blood Component Therapy? J Cardiothorac Vasc Anesth 2018; 32:168.
  112. Sun GH, Patel V, Moreno-Duarte I, et al. Intraoperative Administration of 4-Factor Prothrombin Complex Concentrate Reduces Blood Requirements in Cardiac Transplantation. J Cardiothorac Vasc Anesth 2018; 32:161.
  113. Bhatt HV, Subramaniam K. PRO: Prothrombin Complex Concentrate Should Be Used in Preference to Fresh Frozen Plasma for Hemostasis in Cardiac Surgical Patients. J Cardiothorac Vasc Anesth 2018; 32:1062.
  114. Treml B, Oswald E, Schenk B. Reversing anticoagulation in the hemorrhaging patient. Curr Opin Anaesthesiol 2019; 32:206.
  115. Fitzgerald J, Lenihan M, Callum J, et al. Use of prothrombin complex concentrate for management of coagulopathy after cardiac surgery: a propensity score matched comparison to plasma. Br J Anaesth 2018; 120:928.
  116. Cappabianca G, Mariscalco G, Biancari F, et al. Safety and efficacy of prothrombin complex concentrate as first-line treatment in bleeding after cardiac surgery. Crit Care 2016; 20:5.
  117. Ortmann E, Besser MW, Sharples LD, et al. An exploratory cohort study comparing prothrombin complex concentrate and fresh frozen plasma for the treatment of coagulopathy after complex cardiac surgery. Anesth Analg 2015; 121:26.
  118. Arnékian V, Camous J, Fattal S, et al. Use of prothrombin complex concentrate for excessive bleeding after cardiac surgery. Interact Cardiovasc Thorac Surg 2012; 15:382.
  119. Roman M, Biancari F, Ahmed AB, et al. Prothrombin Complex Concentrate in Cardiac Surgery: A Systematic Review and Meta-Analysis. Ann Thorac Surg 2019; 107:1275.
  120. Song HK, Tibayan FA, Kahl EA, et al. Safety and efficacy of prothrombin complex concentrates for the treatment of coagulopathy after cardiac surgery. J Thorac Cardiovasc Surg 2014; 147:1036.
  121. Franchini M, Lippi G. Prothrombin complex concentrates: an update. Blood Transfus 2010; 8:149.
  122. Fries D. The early use of fibrinogen, prothrombin complex concentrate, and recombinant-activated factor VIIa in massive bleeding. Transfusion 2013; 53 Suppl 1:91S.
  123. Hashmi NK, Ghadimi K, Srinivasan AJ, et al. Three-factor prothrombin complex concentrates for refractory bleeding after cardiovascular surgery within an algorithmic approach to haemostasis. Vox Sang 2019; 114:374.
  124. Goodnough LT, Levy JH. The Judicious Use of Recombinant Factor VIIa. Semin Thromb Hemost 2016; 42:125.
  125. Baral P, Cotter E, Gao G, et al. Characteristics Associated With Mortality in 372 Patients Receiving Low-Dose Recombinant Factor VIIa (rFVIIa) for Cardiac Surgical Bleeding. J Cardiothorac Vasc Anesth 2019; 33:2133.
  126. Hessel EA 2nd. What's New in Cardiopulmonary Bypass. J Cardiothorac Vasc Anesth 2019; 33:2296.
  127. Karkouti K, Beattie WS, Arellano R, et al. Comprehensive Canadian review of the off-label use of recombinant activated factor VII in cardiac surgery. Circulation 2008; 118:331.
  128. Lee AI, Campigotto F, Rawn JD, et al. Clinical significance of coagulation studies in predicting response to activated recombinant Factor VII in cardiac surgery patients. Br J Haematol 2012; 157:397.
  129. Levi M, Levy JH, Andersen HF, Truloff D. Safety of recombinant activated factor VII in randomized clinical trials. N Engl J Med 2010; 363:1791.
  130. Andersen ND, Bhattacharya SD, Williams JB, et al. Intraoperative use of low-dose recombinant activated factor VII during thoracic aortic operations. Ann Thorac Surg 2012; 93:1921.
  131. Gill R, Herbertson M, Vuylsteke A, et al. Safety and efficacy of recombinant activated factor VII: a randomized placebo-controlled trial in the setting of bleeding after cardiac surgery. Circulation 2009; 120:21.
  132. Tiede A, Rand JH, Budde U, et al. How I treat the acquired von Willebrand syndrome. Blood 2011; 117:6777.
  133. Sadler JE. Aortic stenosis, von Willebrand factor, and bleeding. N Engl J Med 2003; 349:323.
  134. Vincentelli A, Susen S, Le Tourneau T, et al. Acquired von Willebrand syndrome in aortic stenosis. N Engl J Med 2003; 349:343.
  135. Steinlechner B, Zeidler P, Base E, et al. Patients with severe aortic valve stenosis and impaired platelet function benefit from preoperative desmopressin infusion. Ann Thorac Surg 2011; 91:1420.
  136. Hanke AA, Dellweg C, Kienbaum P, et al. Effects of desmopressin on platelet function under conditions of hypothermia and acidosis: an in vitro study using multiple electrode aggregometry*. Anaesthesia 2010; 65:688.
  137. Ying CL, Tsang SF, Ng KF. The potential use of desmopressin to correct hypothermia-induced impairment of primary haemostasis--an in vitro study using PFA-100. Resuscitation 2008; 76:129.
  138. Desborough MJ, Oakland K, Brierley C, et al. Desmopressin use for minimising perioperative blood transfusion. Cochrane Database Syst Rev 2017; 7:CD001884.
  139. Desborough MJ, Oakland KA, Landoni G, et al. Desmopressin for treatment of platelet dysfunction and reversal of antiplatelet agents: a systematic review and meta-analysis of randomized controlled trials. J Thromb Haemost 2017; 15:263.
  140. de Prost D, Barbier-Boehm G, Hazebroucq J, et al. Desmopressin has no beneficial effect on excessive postoperative bleeding or blood product requirements associated with cardiopulmonary bypass. Thromb Haemost 1992; 68:106.
  141. Orlov D, McCluskey SA, Callum J, et al. Utilization and Effectiveness of Desmopressin Acetate After Cardiac Surgery Supplemented With Point-of-Care Hemostatic Testing: A Propensity-Score-Matched Analysis. J Cardiothorac Vasc Anesth 2017; 31:883.
  142. Santise G, Nardella S, Migliano F, et al. The HAS-BLED Score is Associated With Major Bleeding in Patients After Cardiac Surgery. J Cardiothorac Vasc Anesth 2019; 33:1601.
  143. Shander A, Javidroozi M, Perelman S, et al. From bloodless surgery to patient blood management. Mt Sinai J Med 2012; 79:56.
  144. Muñoz M, Acheson AG, Bisbe E, et al. An international consensus statement on the management of postoperative anaemia after major surgical procedures. Anaesthesia 2018; 73:1418.
Topic 122792 Version 7.0

References

1 : Activation of the hemostatic system during cardiopulmonary bypass.

2 : Mechanisms and attenuation of hemostatic activation during extracorporeal circulation.

3 : 2019 EACTS/EACTA/EBCP guidelines on cardiopulmonary bypass in adult cardiac surgery.

4 : Hemodynamic changes after protamine administration: association with mortality after coronary artery bypass surgery.

5 : Effects of differing rates of protamine reversal of heparin anticoagulation.

6 : Anticoagulation monitoring during cardiac surgery: a review of current and emerging techniques.

7 : The Society of Thoracic Surgeons, The Society of Cardiovascular Anesthesiologists, and The American Society of ExtraCorporeal Technology: Clinical Practice Guidelines-Anticoagulation During Cardiopulmonary Bypass.

8 : Clinical Impact of Protamine Titration-Based Heparin Neutralization in Patients Undergoing Coronary Bypass Grafting Surgery.

9 : Protamine dosage based on two titrations reduces blood loss after valve replacement surgery: a prospective, double-blinded, randomized study.

10 : Individualized heparin and protamine management improves rotational thromboelastometric parameters and postoperative hemostasis in valve surgery.

11 : Protamine overdose and its impact on coagulation, bleeding, and transfusions after cardiopulmonary bypass: results of a randomized double-blind controlled pilot study.

12 : Blood loss after cardiopulmonary bypass, standard vs titrated protamine: a meta-analysis.

13 : Anticoagulant and side-effects of protamine in cardiac surgery: a narrative review.

14 : Guideline on antiplatelet and anticoagulation management in cardiac surgery.

15 : Protamine reversal of heparin affects platelet aggregation and activated clotting time after cardiopulmonary bypass.

16 : Protamine sulfate down-regulates thrombin generation by inhibiting factor V activation.

17 : At high heparin concentrations, protamine concentrations which reverse heparin anticoagulant effects are insufficient to reverse heparin anti-platelet effects.

18 : The Society of Thoracic Surgeons, The Society of Cardiovascular Anesthesiologists, and The American Society of ExtraCorporeal Technology: Clinical Practice Guidelines-Anticoagulation During Cardiopulmonary Bypass.

19 : Can extra protamine eliminate heparin rebound following cardiopulmonary bypass surgery?

20 : Pharmacokinetic model of unfractionated heparin during and after cardiopulmonary bypass in cardiac surgery.

21 : Rapid disappearance of protamine in adults undergoing cardiac operation with cardiopulmonary bypass.

22 : The mechansim of heparin rebound after extracorporeal circulation for open cardiac surgery.

23 : Monitoring incomplete heparin reversal and heparin rebound after cardiac surgery.

24 : Elevated activated partial thromboplastin time does not correlate with heparin rebound following cardiac surgery.

25 : Protamine allergy.

26 : Protamine: a review of its toxicity.

27 : Protamine, is something fishy about it? The spectre of anaphylaxis continues.

28 : Association of protamine IgE and IgG antibodies with life-threatening reactions to intravenous protamine.

29 : Protamine reactions, explosive mediator release, and pulmonary vasoconstriction.

30 : Three Cases of Anaphylaxis to Protamine: Management of Anticoagulation Reversal.

31 : Probable reversal of protamine reactions by heparin administration.

32 : Effect of site of venous protamine administration, previously alleged risk factors, and preoperative use of aspirin on acute protamine-induced pulmonary vasoconstriction.

33 : C5a and thromboxane generation associated with pulmonary vaso- and broncho-constriction during protamine reversal of heparin.

34 : Anaphylaxis during cardiac surgery: implications for clinicians.

35 : Use of methylene blue for catecholamine-refractory vasoplegia from protamine and aprotinin.

36 : Methylene Blue to Treat Protamine-induced Anaphylaxis Reactions. An Experimental Study in Pigs.

37 : Catastrophic cardiovascular adverse reactions to protamine are nitric oxide/cyclic guanosine monophosphate dependent and endothelium mediated: should methylene blue be the treatment of choice?

38 : Morbidity and mortality after massive transfusion in patients undergoing non-cardiac surgery.

39 : Variation in use of blood transfusion in coronary artery bypass graft surgery.

40 : Society of Cardiovascular Anesthesiologists Clinical Practice Improvement Advisory for Management of Perioperative Bleeding and Hemostasis in Cardiac Surgery Patients.

41 : 2011 update to the Society of Thoracic Surgeons and the Society of Cardiovascular Anesthesiologists blood conservation clinical practice guidelines.

42 : Perioperative blood transfusion and blood conservation in cardiac surgery: the Society of Thoracic Surgeons and The Society of Cardiovascular Anesthesiologists clinical practice guideline.

43 : Anemia and Patient Blood Management in Cardiac Surgery-Literature Review and Current Evidence.

44 : Increased fibrinolysis and platelet activation in elderly patients undergoing coronary bypass surgery.

45 : Transfusion requirements after cardiac surgery: The TRACS randomized controlled trial.

46 : Meta-analysis of the Sources of Bleeding after Adult Cardiac Surgery.

47 : Estimating the risk of complications related to re-exploration for bleeding after adult cardiac surgery: a systematic review and meta-analysis.

48 : Coagulopathy by hypothermia and acidosis: mechanisms of thrombin generation and fibrinogen availability.

49 : A systematic evaluation of the effect of temperature on coagulation enzyme activity and platelet function.

50 : Effect of skin temperature on platelet function in patients undergoing extracorporeal bypass.

51 : The effects of mild perioperative hypothermia on blood loss and transfusion requirement.

52 : The disparity between hypothermic coagulopathy and clotting studies.

53 : The impact of hypothermia on outcomes in massively transfused patients.

54 : Practice guidelines for perioperative blood management: an updated report by the American Society of Anesthesiologists Task Force on Perioperative Blood Management*.

55 : Patient Blood Management: Recommendations From the 2018 Frankfurt Consensus Conference.

56 : Management of severe perioperative bleeding: guidelines from the European Society of Anaesthesiology: First update 2016.

57 : The use of viscoelastic haemostatic assays in the management of major bleeding: A British Society for Haematology Guideline.

58 : Perioperative management of the bleeding patient.

59 : Point-of-Care Hemostatic Testing in Cardiac Surgery: A Stepped-Wedge Clustered Randomized Controlled Trial.

60 : Point-of-care testing: a prospective, randomized clinical trial of efficacy in coagulopathic cardiac surgery patients.

61 : Comparison of structured use of routine laboratory tests or near-patient assessment with clinical judgement in the management of bleeding after cardiac surgery.

62 : Cost reduction of perioperative coagulation management in cardiac surgery: value of "bedside" thrombelastography (ROTEM).

63 : Thromboelastometrically guided transfusion protocol during aortic surgery with circulatory arrest: a prospective, randomized trial.

64 : Thromboelastometry Based Early Goal-Directed Coagulation Management Reduces Blood Transfusion Requirements, Adverse Events, and Costs in Acute Type A Aortic Dissection: A Pilot Study.

65 : Evaluation of a novel transfusion algorithm employing point-of-care coagulation assays in cardiac surgery: a retrospective cohort study with interrupted time-series analysis.

66 : Reduction of Fresh Frozen Plasma Requirements by Perioperative Point-of-Care Coagulation Management with Early Calculated Goal-Directed Therapy.

67 : Technology: Is There Sufficient Evidence to Change Practice in Point-of-Care Management of Coagulopathy?

68 : Shifts of Transfusion Demand in Cardiac Surgery After Implementation of Rotational Thromboelastometry-Guided Transfusion Protocols: Analysis of the HEROES-CS (HEmostasis Registry of patiEntS in Cardiac Surgery) Observational, Prospective Open Cohort Database.

69 : Comparison of current platelet functional tests for the assessment of aspirin and clopidogrel response. A review of the literature.

70 : Thromboelastography (TEG) or thromboelastometry (ROTEM) to monitor haemostatic treatment versus usual care in adults or children with bleeding.

71 : Routine use of viscoelastic blood tests for diagnosis and treatment of coagulopathic bleeding in cardiac surgery: updated systematic review and meta-analysis.

72 : Thromboelastography (TEG) or rotational thromboelastometry (ROTEM) to monitor haemostatic treatment in bleeding patients: a systematic review with meta-analysis and trial sequential analysis.

73 : Point-of-care thromboelastography/thromboelastometry-based coagulation management in cardiac surgery: a meta-analysis of 8332 patients.

74 : Protocol based on thromboelastograph (TEG) out-performs physician preference using laboratory coagulation tests to guide blood replacement during and after cardiac surgery: a pilot study.

75 : First-line therapy with coagulation factor concentrates combined with point-of-care coagulation testing is associated with decreased allogeneic blood transfusion in cardiovascular surgery: a retrospective, single-center cohort study.

76 : Transfusion triggers in cardiac surgery: Where do we go from here?

77 : Restrictive or liberal red-cell transfusion for cardiac surgery.

78 : Six-Month Outcomes after Restrictive or Liberal Transfusion for Cardiac Surgery.

79 : Restrictive compared with liberal red cell transfusion strategies in cardiac surgery: a meta-analysis.

80 : Liberal or restrictive transfusion after cardiac surgery.

81 : A randomized clinical trial of red blood cell transfusion triggers in cardiac surgery.

82 : Should Transfusion Trigger Thresholds Differ for Critical Care Versus Perioperative Patients? A Meta-Analysis of Randomized Trials.

83 : Preoperative anemia versus blood transfusion: Which is the culprit for worse outcomes in cardiac surgery?

84 : Association of red blood cell transfusion and short- and longer-term mortality after coronary artery bypass graft surgery.

85 : The association of blood transfusion with mortality after cardiac surgery: cause or confounding? (CME).

86 : Special Report From the Society for the Advancement of Blood Management: The Choosing Wisely Campaign.

87 : Massive Transfusion in Cardiac Surgery: The Impact of Blood Component Ratios on Clinical Outcomes and Survival.

88 : Fibrinogen as a therapeutic target for bleeding: a review of critical levels and replacement therapy.

89 : Tranexamic Acid for Acute Hemorrhage: A Narrative Review of Landmark Studies and a Critical Reappraisal of Its Use Over the Last Decade.

90 : Tranexamic Acid for Acute Hemorrhage: When Is Enough Evidence Enough?

91 : 2017 EACTS/EACTA Guidelines on patient blood management for adult cardiac surgery.

92 : Randomized, double-blinded, placebo-controlled trial of fibrinogen concentrate supplementation after complex cardiac surgery.

93 : Pro: Factor Concentrates are Essential for Hemostasis in Complex Cardiac Surgery.

94 : Effects of fibrinogen concentrate as first-line therapy during major aortic replacement surgery: a randomized, placebo-controlled trial.

95 : Fibrinogen concentrate in bleeding patients.

96 : Efficacy and Safety of Fibrinogen Concentrate in Surgical Patients: A Meta-Analysis of Randomized Controlled Trials.

97 : Safety of Fibrinogen Concentrate and Cryoprecipitate in Cardiovascular Surgery: Multicenter Database Study.

98 : Fibrinogen Concentrate in Cardiovascular Surgery: A Meta-analysis of Randomized Controlled Trials.

99 : The role of fibrinogen and fibrinogen concentrate in cardiac surgery: an international consensus statement from the Haemostasis and Transfusion Scientific Subcommittee of the European Association of Cardiothoracic Anaesthesiology.

100 : Use of human fibrinogen concentrate during proximal aortic reconstruction with deep hypothermic circulatory arrest.

101 : Effect of Fibrinogen Concentrate vs Cryoprecipitate on Blood Component Transfusion After Cardiac Surgery: The FIBRES Randomized Clinical Trial.

102 : Effect of Fibrinogen Concentrate on Intraoperative Blood Loss Among Patients With Intraoperative Bleeding During High-Risk Cardiac Surgery: A Randomized Clinical Trial.

103 : Randomized evaluation of fibrinogen vs placebo in complex cardiovascular surgery (REPLACE): a double-blind phase III study of haemostatic therapy.

104 : Management of severe perioperative bleeding: guidelines from the European Society of Anaesthesiology.

105 : How I use fibrinogen replacement therapy in acquired bleeding.

106 : Relationship between factor XIII activity, fibrinogen, haemostasis screening tests and postoperative bleeding in cardiopulmonary bypass surgery.

107 : Hemostatic factors and replacement of major blood loss with plasma-poor red cell concentrates.

108 : Prothrombin Complex Concentrates for Perioperative Vitamin K Antagonist and Non-vitamin K Anticoagulant Reversal.

109 : Prothrombin Complex Concentrates for Bleeding in the Perioperative Setting.

110 : Perioperative Use of Coagulation Factor Concentrates in Patients Undergoing Cardiac Surgery.

111 : A Split From Conventional Blood Component Therapy?

112 : Intraoperative Administration of 4-Factor Prothrombin Complex Concentrate Reduces Blood Requirements in Cardiac Transplantation.

113 : PRO: Prothrombin Complex Concentrate Should Be Used in Preference to Fresh Frozen Plasma for Hemostasis in Cardiac Surgical Patients.

114 : Reversing anticoagulation in the hemorrhaging patient.

115 : Use of prothrombin complex concentrate for management of coagulopathy after cardiac surgery: a propensity score matched comparison to plasma.

116 : Safety and efficacy of prothrombin complex concentrate as first-line treatment in bleeding after cardiac surgery.

117 : An exploratory cohort study comparing prothrombin complex concentrate and fresh frozen plasma for the treatment of coagulopathy after complex cardiac surgery.

118 : Use of prothrombin complex concentrate for excessive bleeding after cardiac surgery.

119 : Prothrombin Complex Concentrate in Cardiac Surgery: A Systematic Review and Meta-Analysis.

120 : Safety and efficacy of prothrombin complex concentrates for the treatment of coagulopathy after cardiac surgery.

121 : Prothrombin complex concentrates: an update.

122 : The early use of fibrinogen, prothrombin complex concentrate, and recombinant-activated factor VIIa in massive bleeding.

123 : Three-factor prothrombin complex concentrates for refractory bleeding after cardiovascular surgery within an algorithmic approach to haemostasis.

124 : The Judicious Use of Recombinant Factor VIIa.

125 : Characteristics Associated With Mortality in 372 Patients Receiving Low-Dose Recombinant Factor VIIa (rFVIIa) for Cardiac Surgical Bleeding.

126 : What's New in Cardiopulmonary Bypass.

127 : Comprehensive Canadian review of the off-label use of recombinant activated factor VII in cardiac surgery.

128 : Clinical significance of coagulation studies in predicting response to activated recombinant Factor VII in cardiac surgery patients.

129 : Safety of recombinant activated factor VII in randomized clinical trials.

130 : Intraoperative use of low-dose recombinant activated factor VII during thoracic aortic operations.

131 : Safety and efficacy of recombinant activated factor VII: a randomized placebo-controlled trial in the setting of bleeding after cardiac surgery.

132 : How I treat the acquired von Willebrand syndrome.

133 : Aortic stenosis, von Willebrand factor, and bleeding.

134 : Acquired von Willebrand syndrome in aortic stenosis.

135 : Patients with severe aortic valve stenosis and impaired platelet function benefit from preoperative desmopressin infusion.

136 : Effects of desmopressin on platelet function under conditions of hypothermia and acidosis: an in vitro study using multiple electrode aggregometry*.

137 : The potential use of desmopressin to correct hypothermia-induced impairment of primary haemostasis--an in vitro study using PFA-100.

138 : Desmopressin use for minimising perioperative blood transfusion.

139 : Desmopressin for treatment of platelet dysfunction and reversal of antiplatelet agents: a systematic review and meta-analysis of randomized controlled trials.

140 : Desmopressin has no beneficial effect on excessive postoperative bleeding or blood product requirements associated with cardiopulmonary bypass.

141 : Utilization and Effectiveness of Desmopressin Acetate After Cardiac Surgery Supplemented With Point-of-Care Hemostatic Testing: A Propensity-Score-Matched Analysis.

142 : The HAS-BLED Score is Associated With Major Bleeding in Patients After Cardiac Surgery.

143 : From bloodless surgery to patient blood management.

144 : An international consensus statement on the management of postoperative anaemia after major surgical procedures.