Skip to main content
Erschienen in: Thrombosis Journal 1/2023

Open Access 01.12.2023 | Review

Current and future strategies to monitor and manage coagulation in ECMO patients

verfasst von: Saeedreza Zeibi Shirejini, Josie Carberry, Zoe K. McQuilten, Aidan J. C. Burrell, Shaun D. Gregory, Christoph E. Hagemeyer

Erschienen in: Thrombosis Journal | Ausgabe 1/2023

Abstract

Extracorporeal membrane oxygenation (ECMO) can provide life-saving support for critically ill patients suffering severe respiratory and/or cardiac failure. However, thrombosis and bleeding remain common and complex problems to manage. Key causes of thrombosis in ECMO patients include blood contact to pro-thrombotic and non-physiological surfaces, as well as high shearing forces in the pump and membrane oxygenator. On the other hand, adverse effects of anticoagulant, thrombocytopenia, platelet dysfunction, acquired von Willebrand syndrome, and hyperfibrinolysis are all established as causes of bleeding. Finding safe and effective anticoagulants that balance thrombosis and bleeding risk remains challenging. This review highlights commonly used anticoagulants in ECMO, including their mechanism of action, monitoring methods, strengths and limitations. It further elaborates on existing anticoagulant monitoring strategies, indicating their target range, benefits and drawbacks. Finally, it introduces several highly novel approaches to real-time anticoagulation monitoring methods including sound, optical, fluorescent, and electrical measurement as well as their working principles and future directions for research.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACT
Activated clotting time
ACT-LR
Activated clotting time-low range
aPTT
Partial thromboplastin time
AT
Antithrombin
CPB
Cardiopulmonary bypass
DOAC
Direct oral anticoagulant
DTI
Direct thrombin inhibitors
DTT
Dilute thrombin time
ECLS
Extracorporeal life support
ECMO
Extracorporeal membrane oxygenation
ECT
Ecarin clotting time
FFP
Fresh frozen plasma
HIT
Heparin-induced thrombocytopenia
HSI
Hyperspectral imaging
LVAD
Left ventricular assist devices
PCC
Prothrombin complex concentrates
POC
Point-of-care
PT/INR
Prothrombin time/international normalized ratio
RBC
Red blood cell
ROTEM
Rotational thromboelastometry
TEG
Thromboelastography
UFH
Unfractionated heparin
VA-ECMO
Venoarterial extracorporeal membrane oxygenation
VV-ECMO
Venovenous extracorporeal membrane oxygenation
VKA
Vitamin K antagonists
WHO
World health organization

Introduction

Extracorporeal membrane oxygenation (ECMO) is a treatment option for patients with advanced cardiorespiratory failure and can be utilized in both children and adults [1]. ECMO is a life-saving heart–lung machine that delivers oxygen to patients with refractory severe respiratory or cardiac failure as they await a transplant or recover from a serious illness for a few days or weeks [2]. In an ECMO circuit, the deoxygenated blood drained from the venous circulation is pumped to the oxygenator via a pump to exchange carbon dioxide with oxygen. The blood is then returned to either the venous (VV-ECMO) or arterial (VA-ECMO) circulation [3]. The ECMO circuit components (Fig. 1) include a pump, membrane oxygenator, heat exchanger, venous cannula, arterial or venous infusion canula, tubing, and connectors [4].
The most frequent complications and one of the common causes of death for patients on ECMO are thrombus formation and bleeding [5, 6]. The risk of bleeding is related to patient-specific and treatment-related factors associated with ECMO. According to Virchow's triad, which explains the thrombosis etiologies as hypercoagulability, vessel wall injury, and blood flow stasis, the ECMO circuit contains non-biological surfaces, regions of very high shear stress and regions of prolonged blood residence time, all of which act to promote thrombus formation at a level requiring systemic anticoagulation, which in turn increases bleeding complications [7].
ECMO patients are frequently critically ill, increasing the risk of bleeding complications [8]. The bleeding rate during ECMO is 20.8–39.6% [6, 9] with the cannula site (13.2%), gastrointestinal tract (5.5%), lungs (6.1%), and central nervous system (3.9%) being the most prevalent sites [10]. ECMO patients are also at risk of thrombosis complications, including ischemic stroke, right ventricular thrombus [11], left ventricular thrombus [12], and pulmonary embolism [13]. The rate of thrombosis formation in patients undergoing ECMO is 10–46.1% of patients depending on the circuit type and age of the patient in various centres [14]. Finding the balance between bleeding and thrombosis necessitates continuous monitoring of various parameters including coagulation factors, fibrinogen, and platelets. In this review, conventional and recently developed anticoagulants used in ECMO are presented, including their mechanism of action, monitoring methods, strengths and limitations. It expands on existing anticoagulant monitoring systems, indicating their target range, advantages, and disadvantages. It also introduces various unique real-time coagulation monitoring techniques, as well as their operating principles and future research prospects.

Anticoagulation during extracorporeal circulation

Firstly, anticoagulants utilized in ECMO are summarized, including their benefits and limitations to provide a comprehensive viewpoint for future research. In addition, this review takes a fresh view on anticoagulant monitoring strategies by classifying them according to their purpose (Fig. 2).

Anticoagulant agents

Anticoagulants commonly used for anticoagulation in ECMO patients include heparin, direct thrombin inhibitors (DTIs) (bivalirudin, argatroban, lepirudin), factor Xa inhibitors (danaparoid and fondaparinux), direct oral anticoagulants (DOAC) (DTI (dabigatran), Xabans (rivaroxaban, apixaban, darexaban, edoxaban, and betrixaban)), factor-XIIa inhibitors, nafamostat mesylate (NM), and warfarin so that heparin and DTIs are the most prevalent anticoagulants used in ECMO patients. Although heparin is the most commonly used in clinical applications, heparin resistance is a major concern in ECMO. It is defined as a situation where the ability of heparin to inhibit thrombin (factor IIa) and fibrin formation is reduced such that the correlation between dose and response is lost and increasing the heparin dosage will not result in the desired anticoagulation effect [7, 15]. Moreover, its usage is associated with the rare but life threatening immune-mediated disorder Heparin-induced thrombocytopenia (HIT), specified by thrombocytopenia and a paradoxical prothrombotic state in heparin treatment [16]. Alternative anticoagulants, on the other hand, offer excellent potential for usage in HIT patients, but present some other challenges. The working mechanism, advantages, disadvantages, and other aspects of different anticoagulants used during ECMO support including heparin and its alternatives are provided in Table 1.
Table 1
Anticoagulant Agents
Anticoagulant
Mechanism
Typical dosing
Monitoring
Half-life
Elimination
Reversal agent (Antidote)
Administration
Advantages
Disadvantages
Ref
Heparin
Enhance the AT activity to inactivate thrombin; Inhibiting anti-FXa and AT
A bolus initial dose of 40–100 U/kg followed by a continuous infusion of 10–50 U/kg/hr, targeted to an aPTT or Anti-Xa level
Anti-factor Xa, ACT, aPTT, ROTEM/TEG, AT assay
90 min
Renal and hepatic
Protamine
Intravenous (IV)
● Well known mechanism of action
● Easy to monitor
● Easy titration
● Ease of reversibility with protamine
● Inexpensive
● Non-linear dose–response relationship
● HIT induction
● Heparin resistance
● Depends on the level of AT
[61]
Bivalirudin
Reversible direct thrombin inhibitor
It can be used with or without a bolus; A bolus initial dose of 0–0.75 mg/kg and a maintenance dose of between 0.025 and 0.5 mg/kg/h
Plasma level of drug, ECT, aPTT, diluted thrombin time (dTT), ACT
25 min
Renal (20%) and hepatic (80%)
None
IV
● No HIT induction
● Short half-life
● Independent of AT levels
● Bivalirudin resistance requiring dose escalation
● Interferes with fibrin production, platelet aggregation, and factor XII activation
● Prolonged half-life and drug accumulation in renal failure
[62]
Argatroban
Reversible direct thrombin inhibitor
Typically, no bolus loading is required (However, some centres use 100–200 \(\mathrm{\mu g}/\mathrm{kg}\))
A maintenance infusion of rate between 0.1 and 2 \(\mathrm{\mu g}/\mathrm{kg}/\mathrm{min}\)
aPTT, diluted thrombin time, ACT, ECT, ROTEM/TEG
39 to 51 min; depending on hepatic function
Hepatic
None
IV
● No HIT induction
● Fewer transfusions than heparin
● Independent of AT levels
● Increased dose required to maintain therapeutic range over time
● Less familiar to most clinicians
● Long onset of action
● Interferes with fibrin production, platelet aggregation and factor XII activation
● Prolongs the aPTT, ACT, PT/INR results
[63]
Danaparoid
Factor Xa inhibitor
200–300 U/hr
Anti‐factor Xa assay
19‐25 h
Renal
None
IV and subcutaneous
● Recommended for HIT situation
● Some anti-thrombin activities
● Has been unavailable for several years due to manufacturing issues
[64]
Fondaparinux
Factor Xa inhibitor
2.5–10 mg/day
Anti‐factor Xa assay
17–21 h
Renal
None
IV and subcutaneous
● High level of activity
● Recommended for HIT situation
● Its activity is 20 times greater than the danaparoid
● Does not inhibit thrombin
● Major bleeding in HIT patients
● Restricted usage in renal dysfunction situation
● Not for use while actively undergoing ECMO
[65]
Nafamostat mesylate
Inhibits many procoagulant factors including thrombin, factors XIIa, Xa, and antifibrinolytic and anti-platelet action
0.26–0.93 mg/kg/hr
ACT, aPTT
8 min
Renal
None
IV
● Short half-life
● Limited published data on its safety and efficacy
[66]
Warfarin
Inhibits vitamin K-dependent clotting factors (II, VII, IX, X)
Initial dose of 10 mg and maintenance dosage of 1–2 mg per day
INR
20–60 h
Mainly hepatic and minor enzymatic
Vitamin K, prothrombin complex concentrates (PCC) and fresh frozen plasma (FFP)
IV or orally
● Long-term anticoagulation after ECMO
● Easy administration
● Reversibility
● Not for use while actively undergoing ECMO
● Not a good choice for acute HIT situation
● High drug interactions and dose adjustment required
[67]
Dabigatran
Reversible direct thrombin inhibitor
75–150 mg per day
ACT, aPTT, ECT and thrombin time (TT)
12–17 h, up to 34 h in renal failure
Renal
Idarucizumab (Praxbind)
Oral
● Does not require frequent laboratory monitoring due to its low drug–drug and drug–food interactions
● Reversibility
● Induces dyspepsia
● High cost
● Limited published data on its safety and efficacy
● Limited application in ECMO due to long onset of action
[68]
Rivaroxaban
Factor Xa inhibitor
15–20 mg per day
Prothrombin time, Anti‐factor Xa assay
5–9 h in healthy young people and 11 to 13 h in elderly people
Both renal and hepatic
Andexanet alfa
Oral
● Does not require frequent laboratory monitoring
● Reversibility
● Limited information about using DOACs in ECMO patients due to long onset of action and prolonged half-life
[69]
AT Antithrombin, TT Thrombin Time, dTT Diluted thrombin Time, DTI Direct Thrombin Inhibitor, DOAC Direct Oral Anticoagulant, NM Nafamostat mesylate, IV Intravenous, HIT Heparin-Induced Thrombocytopenia, FBC Full Blood Count, aPTT Partial Thromboplastin Time, PT/INR Prothrombin time/international normalized ratio, ECT Ecarin Clotting Time, POC Point-of-Care, ACT Activated Clotting Time, ROTEM Rotational Thromboelastometry, TEG Thromboelastography)

Heparin

UFH is the most frequently used anticoagulant in patients undergoing ECMO due to its advantages including its low cost, titratability, and easy reversibility by protamine. Heparin inhibits thrombin by binding to antithrombin (AT). AT has low anticoagulation activity but when conjugated with heparin, its anticoagulation activity increases 1000–2000-fold [17]. Heparin resistance is the main concern in ECMO, which is defined as a situation where the heparin ability to inhibit thrombin and fibrin formation is reduced so that heparin dosage response is not correlated with the injected amount of heparin and increasing the heparin dosage will not result in the desired anticoagulation effect [7]. More heparin injections are required under these conditions to achieve desired activated clotting time (ACT) values, which may result in bleeding. The situation is significantly worse in newborns as their AT level is lower than that of adults [18].
Heparin use is associated with immune-mediated side effects known HIT and specified by thrombocytopenia and a paradoxical prothrombotic state in heparin treatment. While it has been reported that HIT is a rare phenomenon in neonates [19, 20], it occurs in 0.8–7% of the adult ECMO patients [21, 22]. It has been demonstrated that 30–60% of HIT patients experience thrombotic complications [23, 24]. Therefore, in order to address the aforementioned issues, it is necessary to provide alternative anticoagulants to heparin, such as bivalirudin [20].

Bivalirudin

DTIs, unlike UFH, do not rely on AT to act as an anticoagulant, but instead they directly inhibit both free circulating and fibrin-bound thrombin. Bivalirudin is a reversible thrombin-binding synthetic bivalent analogue of hirudin with excellent pharmacological profiles [25]. Since bivalirudin can inhibit plasma thrombin, clot-bound thrombin, and collagen-induced platelet activation without forming a complex with the cofactor AT III, it has a much higher bioavailability than heparin [26, 27]. It has a short half-life of approximately 25 min in patients with normal renal function, making it suitable for rapid titration [28, 29]. It is mostly metabolized by the liver through proteolytic cleavage, but it also partially cleared by the kidney (20%), so the dose should be adjusted during renal dysfunction as it prolongs its half-life [30, 31]. Bivalirudin has been used to prevent clotting during ECMO in both HIT patients and non-HIT patients [3234]. It is administered intravenously in doses ranging from 0.025 to 0.48 mg/kg/hour, with an action time of 2–4 min [35]. Bivalirudin efficacy has been shown to correlate well with both ACT and aPTT results [36, 37]. The researchers also compared the bivalirudin and UHF aPTT results and discovered that using bivalirudin yields more stable aPTT results [34, 38]. There is no unified approach for bivalirudin infusion, so it can be used with or without an initial bolus of bivalirudin with initial loading ranging from 0.04 to 2.5 mg/kg followed by continuous infusion [34, 39]. In particular, Koster et al. used bivalirudin for HIT patients with a bolus of 0.5 mg/kg followed by a continuous infusion of 0.5 mg/kg/h to maintain an ACT of 200–220 s [33]. In another study, Jyoti et al. [39], were able to achieve a target ACT of 200–220 s with an injection rate of 0.1–0.2 mg/kg/h and no bolus dosage of bivalirudin. The dose of bivalirudin is maintained at 0.03–0.2 mg/kg/h to maintain therapeutic targets, with [19, 32, 33] or without [34] an additional initial amount of 0.5 mg/kg. In a meta-analysis, it has been reported that in-hospital mortality, major bleeding events and pump-related thrombosis were less frequent in DTI compared to heparin [40].
There are some considerations before bivalirudin usage in ECMO. First, it affects the renal clearance process in patients with impaired renal function, resulting in drug accumulation. Therefore, lower dosage of bivalirudin is required for patients with renal dysfunction [41]. Moreover, since bivalirudin is rapidly metabolized where the blood is in stasis, it is not a viable option in venoarterial ECMO [42]. Another limitation of the bivalirudin is that there is no antidote in case of overdose or bleeding, which makes bleeding management challenging. Bivalirudin resistance may exist in the absence of a clear etiology [43]. APTT, ECT, and plasma-diluted thrombin time tests are typically used for monitoring anticoagulant effect of bivalirudin [44, 45].

Argatroban

Argatroban is a synthetic direct thrombin inhibitor with a half-life of 39–51 min [46] and is not recommended for patients with severe hepatic dysfunction since it is metabolized in the liver, therefore, renal failure is not a concern. One of the primary issues in ECLS that limits DTIs adoption is a lack of pharmacologic antidote. However, due to their short half-lives, if bleeding occurs, the injection of DTIs can be stopped or reduced to stop the bleeding. Argatroban has been utilised as an alternative to UHF in cases of suspected HIT in adults, pediatrics, and neonates on ECMO. Its maintenance dose is 0.1–0.65 \(\mathrm{\mu g}/\mathrm{kg}/\mathrm{min}\) [47, 48], and centres use a 100–200 \(\mathrm{\mu g}/\mathrm{kg}\) initial bolus dose [35].

Fondaparinux

Fondaparinux is a factor Xa inhibitor that has been indicated to be effective as an anticoagulant agent in severe acute HIT [49]. Parlar et al. [50] used fondaparinux daily subcutaneous injections (2.5 mg per day) in ECMO loop for a patient with HIT and found no adverse effects. Compared with DTIs which require the aPTT or the ECT monitoring methods, anti-FXa assays are more reliable for fondaparinux monitoring since anti-FXa assays do not rely on patient factors. To delineate, the aPTT and the ECT are influenced by the prothrombin level of the HIT patients which is often low and results in falsely long aPTT and, consequently, in inappropriate dose of the anticoagulant [51].

Nafamostat mesylate

Nafamostat mesylate is a synthetic serine‐protease inhibitor that inhibits many procoagulant factors, including thrombin, plasmin, trypsin, kallikrein, factors XIIa and Xa [52]. In the literature, there are conflicting findings for the use of NM in ECMO. Lim et al. [53] investigated thromboembolic or bleeding complications during ECMO using heparin and NM. According to their findings, bleeding complications were more common in patients receiving NM, while thromboembolic problems were comparable in both cases. Other studies, on the other hand, claim that it is an appropriate alternative to heparin that reduces the risk of bleeding in ECMO patients [54, 55]. Like other anticoagulants, there is no unified approach for the dosage rate but typically it falls into the range of 0.26–0.93 mg/kg/hr [5456].

Warfarin

Warfarin is an oral anticoagulant that inhibits the utilisation of vitamin K (factors II, VII, IX, and X). The main advantage of this method is its ease of administration and reversibility [57]. When patients have been adequately anticoagulated with DTIs and require long-term anticoagulation after the acute period of HIT, they are typically switched to vitamin K antagonists (VKA) such as warfarin and phenprocoumon [58]. Warfarin dosage should be determined based on the patient's response to the drug, and it can be monitored using international normalised ratio (INR) analysis to keep its results in therapeutic range [2, 3, 59]. Lee et al. describe the successful use of ECMO as a bridge-to-recovery therapy in a patient suffering from fatal warfarin-exacerbated DAH [60].

Anticoagulation monitoring methods

Given the importance of anticoagulant monitoring and dose adjustment, it is vital to determine the appropriate approach for each anticoagulant and its dosage. Partial thromboplastin time (aPTT), anti-factor Xa assay, D-dimer, PT/INR, Full blood counts (FBC), Fibrinogen, ECT, activated clotting time (ACT), and viscoelastic tests (ROTEM/TEG) are discussed in this section. Also, recently developed novel real-time monitoring methods including sound, optical, fluorescent, and electrical measurements methods are presented. Table 2 summarises the sample type, purpose, target range, advantages, and disadvantages of different techniques.
Table 2
Conventional anticoagulation monitoring methods
Method
Sample type
Advantages
Disadvantages
Purpose
Desired range
Ref
aPTT
Plasma
● Widely available
● Well known method
● Easy to interpret
● Not affected by platelet count
● Nonspecific to heparin
● Time-consuming and user-dependent
● Can be affected by various parameters, such as drugs, hematocrit, abnormalities in coagulation factors, fibrinogen, high C-reactive protein
● Extended clotting time in the presence of lupus anticoagulant
To monitor anticoagulant effect
25–90 s or 1.5–2.5 times baseline (depending on the device, anticoagulant, and utilized method)
[70]
Anti-factor Xa assay
Plasma
● Sensitive to UFH
● Independent of coagulopathy, thrombocytopenia, or dilution
● Correlates better than ACT and aPTT with heparin concentration
● Affected by hyperlipidemia and hyperbilirubinemia, haemolysis, lipaemia, AT, and high plasma-free haemoglobin
● Not easily accessible
● Costly
● Time-consuming
● Only measures inhibition while not showing the amount of fibrin and thrombin generated
To monitor anticoagulant effect
0.3–0.7 IU/mL
[71]
ACT
Whole blood
● Point-of-care test
● Low cost and fast
● Simple operation
● Better correlate with high concentrations of heparin
Small sample volume
● User and instrument dependent results
● Lack of specificity
Sensitive to various parameters including platelet dysfunction, platelet inhibitors (e.g., GP IIb/IIIa), temperature, haematocrit, blood thrombocyte activity, haemodilution, anemia, hypothermia, coagulation factors deficiencies, hypofibrinogenemia, fibrinogen, thrombocytopenia, AT level, oral coagulants, and activator type
To assess anticoagulant effect
180–220 s
[72]
D-dimer
Whole blood or plasma
● Prognostic value for oxygenator failure
● High sensitivity
● Time-consuming
● Relatively expensive
● Moderate specificity
To monitor fibrin formation and fibrinolysis in the circuit and patient’s body
0.28–1 mg/L
[73]
PT/INR
Plasma
● Indicator of various factors including factors I, II, V, VII, and X
● Interfere with DTIs
● Deficits in common factors I, II, V, and X can prolong PT
To assess underlying coagulability and to monitor Vitamin K antagonists
 < 1.5 for heparin-treated patients, > 2–3 for other anticoagulants
[74]
ECT
Plasma
● Simple operation
● Not sensitive to heparin
● Rely on both prothrombin and fibrinogen
● Lack of standardization and uniformity
● Results affected by DTIs
To monitor the effect of DTIs
300–500 s
[75]
Viscoelastic tests (ROTEM/TEG)
Whole blood
● Point-of-care test
● Distinguishing clotting factor deficiency, platelet dysfunction, and hyperfibrinolysis
● Poor specificity
● Low sensitivity to hyperfibrinolysis
● Sensitive to vibration
● Extended clotting time in the presence of lupus anticoagulant
● Lack of standardization and uniformity
To assess clotting time (anticoagulant effect), thrombocytopenia, hypofibrinogenemia, and fibrinolysis
Not established
 
FBC Full Blood Count, aPTT Partial Thromboplastin Time, PT/INR Prothrombin time/international normalized ratio, DTI Direct thrombin inhibitors, ECT Ecarin Clotting Time, ACT Activated Clotting Time, ROTEM Rotational Thromboelastometry, TEG Thromboelastography), UFH Unfractionated heparin

Activated partial thromboplastin time

APTT is an anticoagulant monitoring technique that is most commonly used to assess the effect of heparin and bivalirudin [76]. It is defined as the time required for calcium-free plasma to generate clots after being exposed to fibrin-activating reagents and calcium. Clot formation can be detected using a variety of analytical methods, including optical, mechanical, and electrochemical techniques [77]. This method involves combining citrated plasma, a phospholipid, calcium, and a contact pathway activator (silica, celite, kaolin, ellagic acid, polyphenolic acid) to trigger clot formation [78]. The normal range is defined in most laboratories as 25–90 s (an aPTT level of 1.5–2.5 times baseline is recommended for anticoagulation monitoring) but it varies from clinic to clinic and is determined by the instrument and reagents used and it is critical not to extrapolate data from one ECMO centre to another without knowing the method and assay used [79, 80]. Bates et al. investigated the relationship between aPTT and anti-factor Xa assay heparin level using four different automated coagulometers and six commercial aPTT reagents. Their findings revealed that, while there is a good correlation (r = 0.64 to 0.95) between aPTT anti-factor Xa assay results, the aPTT values at 0.3 IU/mL plasma heparin concentration determined by anti-factor Xa assay will range from 48 to 108 s, depending on the instrument and reagent utilised [81].
Another concern with this method is that it can be influenced by parameters, including drugs, hematocrit, acute phase reactants, abnormalities in coagulation factors, high C-reactive protein, hyperbilirubinemia, hyperlipidemia and lupus anticoagulant [82] so that deficiencies in common pathway factors I, II, V, and X, as well as contact pathway components such as high-molecular-weight kininogen, prekallikrein, and factors VIII, IX, XI, and XII, and lupus anticoagulants can prolong the aPTT results [78].

Anti-factor Xa assay

Anti-factor Xa is a functional chromogenic assay for coagulation monitoring and evaluating the effective anticoagulant concentration. The anti-Xa assay is specific to heparinoid’s action and is unaffected by deficits in other coagulation factors and can be used with or without exogenous AT. In the former method, the sample is treated with sufficient AT, so that the rate-limiting reagent, which is heparin, can inhibit Xa and produce a precise measurement of heparin in the patient sample. In this method, a specific amount of coagulation factor Xa conjugated with chromophore and AT is added to patient plasma containing heparin [83]. Following that, as a result of the chromogenic reaction, AT and heparin form an inhibitory complex that inactivates factor Xa. The activated factor X is then introduced to the sample, which cleaves the chromophore compound, and the amount of released chromophore is measured using spectroscopy. As the amount of remaining factor Xa in the sample is inversely proportional to the original amount of heparin, the colour change will be greater as less heparin/AT complex interacts with factor Xa, indicating a lower drug level. The relationship between AT and heparin is critical because, even with a high level of heparin, a deficiency in AT causes more unbound factor Xa, implying lower heparin levels. On the other hand, kits without AT do not add extra AT and give a more precise measurement of in-vivo anticoagulation because the patient's AT and heparin levels are both rate-limiting reagents. However, this approach has the problem of being unable to differentiate between AT deficiency and inadequate heparin [84, 85].
The widely accepted target range for anti-factor Xa levels during ECMO is 0.3–0.7 IU/mL [86, 87]. Unlike the ACT and aPTT methods, this method is unaffected by coagulopathy, thrombocytopenia, or dilution and best represents the overall heparin anticoagulation level. However, some parameters which can occur in patients on ECMO, such as hyperbilirubinemia, haemolysis, lipaemia, hyperlipidemia, and plasma-free haemoglobin affect anti-Xa assay results [88, 89]. Anti-Xa activity was observed to be significantly lower in ECMO samples with plasma free haemoglobin levels of 50 mg/dL or above when compared to normal samples: 0.33 (0.25–0.42) versus 0.4 (0.31–0.48) IU/mL [86, 87, 90]. It was stated that anti-Xa assay correlates better with heparin concentration than ACT and aPTT [9193]. Nankervis et al. studied 12 neonates on ECMO to determine the appropriate heparin dosage based on ACT and anti-Xa results. Their findings show a strong correlation between anti-Xa test and heparin dosage (r = 0.75; p < 0.0001), however ACT results do not correlate with either anti-Xa or heparin dosage [94]. Also, in a cohort study, Bembea et al. [95] discovered a moderate correlation between anti-factor Xa results and heparin injection dosage (r = 0.33) in 34 extracorporeal life support (ECLS) pediatric patients, but a poor correlation with ACT (r = 0.02) and aPTT (r = 0.17) for all patients. It is worth noting that patients on ECMO who are being monitored by anti-Xa levels compared to ACT have fewer blood draws for monitoring, a longer duration between circuit changes, and lower transfusions and dosages of activated factor VII [93, 96].
While anti-factor Xa assay can measure the heparin effect, it cannot reflect other coagulation parameters or the patient's overall hemostatic condition [97]. In fact, the anti-Xa method only measures inhibition, not the amount of fibrin and thrombin produced in the patient's body. Moreover, compared with ACT and aPTT, anti-factor Xa assay more expensive and not easily accessible in all laboratories and hospitals and its results are affected by hypertriglyceridemia (triglyceride level > 360 mg/dL) and hyperbilirubinemia (bilirubin level > 6.6 mg/dL) [98].

Activated clotting time

ACT is the widely used and well-established point-of-care (POC) method for anticoagulation monitoring. In this method, whole blood is transferred to a tube coated with various activators, such as glass, celite or kaoline, ellagic acid, diatomaceous earth, silica, calcium, and phospholipids, to stimulate the contact activation pathway and the coagulation response (fibrin clot formation) is measured over time in seconds. The mobility of a magnet during clot formation, or the variations in the velocity of blood movement as it begins to clot, is measured and recorded over time. When compared to laboratory-based methods, the advantage of ACT is that it can be conducted as a whole-blood test on a bedside machine, requires a little sample volume, is low-cost, and can be done by unacquainted individuals [99]. The amount of sample required for testing varies between 10 \(\mathrm{\mu L}\) and 2 mL depending on the ACT machine. However, several factors including hemodilution, platelet dysfunction, hypothermia, anemia, hypofibrinogenemia, thrombocytopenia, platelet inhibitors (e.g., GP IIb/IIIa), and coagulation factor deficiencies can affect ACT results [100, 101].
There are currently no standardized target range for ACT, however the range of 140–240 s are commonly used in clinical practice [7, 102]. The aPTT approach works well for Unfractionated heparin (UFH) values between 0.1 and 1 U/mL, but the ACT method works better for UFH concentrations between 1 and 5 U/mL. Therefore, based on the heparin level used in ECMO, ACT shows a poor correlation with heparin concentration, while aPTT yields acceptable results in neonates and adults [103105]. Several studies have been conducted to investigate the relationship between ACT, aPTT, and anti-Xa activity [106, 107]. Khaja et al. discovered that in neonates, aPTT has a stronger correlation with anti-XA than ACT [105]. Although ACT is a good indicator for high heparin concentrations in cardiopulmonary bypass (CPB), low-range ACT (ACT-LR) was proposed for use in the lower heparin dosage range (150–200 IU/kg) used in ECMO [61, 105]. It is worth noting that most hospitals use ACT or ACT-LR methods for routine coagulation monitoring in ECMO.

D-dimer

The D-dimer protein is the cleaved product of the fibrinolysis process. Therefore, the presence of D-dimers in the blood signals clot lysis. D-dimer levels are higher in patients with disseminated intravascular coagulation (DIC) caused by sepsis, deep vein thrombosis (DVT), pulmonary embolism, or other thrombotic disorders. The D-dimer level, which describes the cross-linked fibrin degradation products generated by reactive fibrinolysis, has also been shown to be a marker for determining oxygenator functionality and tracking oxygenator malfunction [108]. In a retrospective study it was found that D-dimer level increased significantly within 3 days before exchange from 15 [920] to 30 [2135] mg/dL (P = 0.002) and declined significantly within 1 day thereafter to 13 [717] mg/dL (P = 0.003) [109]. Although the oxygen transferred rate is commonly used to calculate the exchange time for the oxygenator, the D-dimer level, and particularly a steadily increasing level in the absence of an alternative explanation, is an alternative method for calculating the appropriate time for membrane exchange.

Prothrombin time and international normalized ratio

Prothrombin time (PT) is one of the main clinical coagulation monitoring methods. Prothrombin is a protein made by the liver that aids in blood clotting formation. This method involves adding thromboplastin (a mixture of tissue factor, calcium, and phospholipid) to patient plasma and measuring the time it takes for the plasma to clot [110]. The PT results are sensitive to thromboplastin components so using different thromboplastin reagents results in different Prothrombin times, even with the same plasma sample. Therefore, to have consistent and unified results, the World Health Organization (WHO) proposed the international normalized ratio (INR), which has since become the standard method for PT reporting [111]. In this way, INR represents the ratio of PT divided by a control PT value determined by using a WHO-developed international reference thromboplastin reagent. The target INR for a heparin-treated patient should be less than 1.5. Researchers also discovered that the PT/INR value for other anticoagulants including VKA (e.g., warfarin), DTIs, or the oral Factor Xa inhibitors, is in the range of 2–3 or higher [112]. DTIs interfere with the prothrombin time and deficits in common factors I, II, V, and X can prolong PT results [113]. INR was developed to account for discrepancies in laboratory PT reagents and to standardise vitamin K antagonists (VKA), such as warfarin therapy monitoring [78].

Ecarin clotting time

Ecarin is a metalloprotease isolated from the venom of a saw-scaled viper echis carinatus and a specific activator of prothrombin that can be used to measure the activity of DTIs, such as argatroban and dabigatran [114, 115]. In this method, clotting time is measured after the addition of ecarin solution to diluted plasma. The conversion of fibrinogen to fibrin is used to measure the proteolytic activity of non-inhibited meizothrombin. ECT was successfully implemented by Teruya et al. for bivalirudin concentration monitoring in ECMO [62]. Although this method is not widely used in ECMO, it has been demonstrated that ECT results have a strong correlation with bivalirudin concentration [37]. Furthermore, Alouidor et al. recently proposed a point-of-care ECT analysis device with a good correlation with bivalirudin and dabigatran concentrations that only requires 5 \(\mathrm{\mu L}\) of whole blood [116]. Finally, it seems that more research into this approach in ECMO settings with various anticoagulants is required.

Viscoelastic tests

Thromboelastography (TEG) and rotational thromboelastometry (ROTEM) are point-of-care viscoelastic tests used in ECMO patients to monitor coagulation in the presence of a various stimulating agents [117119]. Unlike other methods, such as ACT, aPTT, and PT/INR, which only show the endpoint (whether or not a thrombus has formed), this assay can reveal information about different aspects of the coagulation cascade, the clot formation dynamics, clot strength and clot lysis, and fibrinolysis [120122]. The dynamic changes in the viscoelasticity of whole blood during clotting under low shear stress are recorded. A torsion wire or pin is used to measure the strength of the formed clot. The wire in TEG is stationary and the cup oscillates while in ROTEM, the cup is stationary while the pin oscillates [123]. The oscillations are hampered as the clot forms, and both the TEG and ROTEM devices detect and convert changes in oscillations into the numerous measurements evaluated. In this method, a pair of samples with and without heparinase addition are tested to investigate hemostasis in the presence of UFH [124]. The effectiveness of UFH is then determined by comparing the clotting time in both samples, which is a good indicator in cases where heparin resistance is a concern. The analysis duration for different parameters varies, so coagulation factors can be assessed in 5 min, and fibrinolysis analysis takes 60–90 min. While viscoelastic tests provide useful information about the coagulation system, it does not provide data on the level of von Willebrand factor (VWF), which is a good indicator of potential bleeding or hemostasis condition [125]. Although the use of viscoelastic tests in ECMO patients has increased in recent years, it is still not widely available in most centres [126] which can be due to a lack of generalised therapeutic ranges in the neonatal population [17].

Novel real-time anticoagulation monitoring methods

This section presents recently developed novel real-time monitoring methods such as sound, optical, fluorescent, and electrical measurements. Furthermore, a brief explanation of each approach and its application in ECMO circuits is provided.

Sound monitoring method

Due to the high shear rates in the ECMO pump, it is one of the suspect points for clot formation. Measuring the sound generated by the pump is a promising method for detecting clot formation in the pump and preventing potential patient and pump damage. Fuchs et al. [127] discovered a link between sound signals and clots in the pump's inlet and outlet while using ECMO. The frequency spectrum changes while blood clots move through the pump at different pump speeds, which is a reliable indicator of clot formation inside the pump (Fig. 3). It has been illustrated that the acoustic analysis method is a low-cost, simple, and easy-to-access method that can provide reliable results and detect thrombosis formation in the pump in the early stages of thrombosis formation. Although this is a non-invasive technology, it can only be used to monitor thrombosis in the pump because the analysis is based on the sound made by the pump [128].

Optical method

Optical methods are non-invasive and real-time thrombus monitoring techniques that detect the light intensity scattered by blood at different wavelengths [129]. Fujiwara et al. [130] utilized a hyperspectral imaging (HSI) technique to visualize thrombosis formation within a levitated centrifugal pump, which is commonly used in ECMO devices (Fig. 4). Two groups of pigs were employed in this study undergoing ECMO and LVAD. The purpose of this study was to detect thrombosis inside the centrifugal ECMO pump and the source of the thrombosis. In this way, multiple real-time images of the inside of the pump were acquired with the HS camera over the wavelength range of 608–752 nm. Within 24 h of blood circulation, thrombosis was discovered, arising from both the inside and outside of the pump. Thrombosis generated outside the pump was identified to form around the inlet cannula and junction between the pump inlet and tubing. It is worth noting that optical methods have some limitations. First, the installation and adjustment of the distance for hyperspectral camera focusing should be considered. Furthermore, due to the limitations on bending radius, optical fibres are fragile.
In another study, Morita et al. [129] used a micro-optical monitoring method with an extracorporeal circuit to detect thrombus in fresh porcine blood. Their setup composed of two micro-optical thrombus sensors which detect scattered light at two wavelengths, 660 and 855 nm. One optical sensor was placed on the rotary pump (to monitor clotting) and the other on the flow channel (less suspicious thrombus formation point, as reference point). In this vein, each LED emits 660 and 855 nm light into the blood flow, which is absorbed or scattered by blood components (most notably red blood cells (RBCs)) and then detected by a photodiode. To monitor thrombus formation in the pump, the ratio of light intensity at each wavelength for both sensors was eventually measured. They also depicted that the proposed micro-optical sensor has no installation limitations and allows researchers to install additional micro-sensors at various points where thrombus formation is suspected.

Fluorescence method

Fluorescence microscopy is another monitoring method for thrombin formation, which measures the onset of thrombus formation, which produces fibrin in the blood. By fluorescently labelling fibrinogen, it is possible to evaluate the microscopic clot formation process [131]. Therefore, the clotting time can be calculated by measuring the variation and distribution of fluorescent intensity over time and, consequently, the influence of heparin concentration on blood clotting time. Considering that high shear rate regions are more suspect for clot formation, Sun et al. [132] utilized a whole blood flow cytometry assay to measure platelet activation in a fresh human blood sample in the centrifugal pump and oxygenator in the ECMO circuit. It was found that platelet activation and adhesion on fibrinogen increases after 4 h of running ECMO which can result in thrombosis formation. On the other hand, GPIbα and GPVI platelet receptors population decreases over time weakening platelet adhesion to collagen and VWF, resulting in bleeding complications (Fig. 5).

Electrical measurement

Electrical impedance measurements can be used to monitor blood coagulation in ECMO systems. Based on single or multiple-frequency electrical impedance measurements, it has been demonstrated that there is a relationship between thrombus formation and electrical resistivity/permittivity [133]. Red blood cells (RBCs) have phospholipid on their surfaces, they can activate factor IX causing coagulation. Therefore, it can be deduced that RBC aggregability is related to thrombus formation. Using multiple-frequency electrical impedance spectroscopy, Li et al. [134] investigated RBC aggregability in an extracorporeal circulation system with pulsatile flow. Their results revealed that in coagulating blood, RBCs aggregability decreases, indicating thrombus formation. ACT and fibrinogen were examined to assess the relationship between aggregability and blood coagulation, and their results show a decrease in their level over time, similar to RBC aggregability (Fig. 6). It is also worth noting that electrical impedance measurement can be readily applied to low-cost, compact, and simple POC blood coagulation testing devices. However, the limitation of this method is that it is invasive because the electrodes must come into contact with the blood.

Future perspectives

It is essential to find appropriate anticoagulants and related monitoring methods to balance between thrombosis and bleeding with ECMO. Factor XII inhibitors are promising anticoagulant agents which are currently under investigation in in-vivo ECMO models. It has been demonstrated that coating the surface with FXIIa‐directed corn trypsin inhibitor prevents blood clotting when catheters are inserted into rabbit jugular veins [135]. Also, it has been illustrated that knocking out FXII enhances the time to catheter occlusion. Recently, researchers found that human antibody 3F7 binds to FXIIa, preventing blood clotting within the ECMO circuit [136]. Antibodies have an advantage over other anticoagulants in that they do not increase the risk of bleeding. Although it has only been tested in animal ECMO circuits, FXII-directed therapy appears to be a promising method for clot prevention on ECMO support [137].
It has recently been demonstrated in a variety of clinical settings that using low-dose anticoagulation is an effective strategy to decrease bleeding complications and enhance survival rate while patients are receiving ECMO, as opposed to when a standard dosage is used [138, 139]. Although thrombosis is still a concern, recent studies suggest that there is no difference in thrombosis complications and hospital mortality between low-dose and therapeutic anticoagulant dosage [140]. While several studies have now demonstrated the benefits of using low-dose anticoagulation in adults to reduce bleeding while still preventing major thrombosis, there is still a lack of research on the efficacy and safety of using low-dose anticoagulation in pediatric patients undergoing ECMO. As a result, extensive clinical research and trials are required to determine the optimal low anticoagulant dosage in both adults and pediatric patients.
In another approach, shear-sensitive drug delivery systems may protect the ECMO circuit from coagulation and thrombosis formation by modifying commonly used generalized drug delivery which results in bleeding complications. Localized drug delivery method is a novel approach which relies on material deformation or disaggregation to elicit drug release [141]. Nanoparticles can be used for nanotherapeutic targeting drug delivery systems that, like platelets, can activate (rupture) under high shear stress and release the drug at the site of action to prevent blood clotting [142]. This characteristic enables the development of shear-sensitive nanoparticles capable of releasing antithrombosis drugs only at high shear stress points, such as the cannulas, pump and oxygenator while remaining intact at lower shear rates. This is beneficial because it not only reduces the amount of anticoagulant used but also aids in localized drug delivery, which only releases the drug where it is required, potentially resulting in less bleeding.
Recently, an anticoagulant-free method based on surface modification of the ECMO circuit was developed to decrease bleeding and thrombosis complications [143]. This method is based on the application of appropriate surface coating techniques to make the surface of the tubing, membrane oxygenator, and pump biocompatible and avoid thrombosis formation. Several innovative approaches, such as polymer coating, nitric oxide (NO) coating, and tethered liquid perfluorocarbon coating, are presented that can improve ECMO hemocompatibility. When considering surface coating technologies, it is also crucial to consider gas exchange rate, pressure drop, and shear stress inside the membrane oxygenator. These features must be incorporated to increase the stability and durability of the materials suitable for biocompatible surface coatings.
Various real-time analysis methods for thrombosis monitoring are presented in this review. Although these techniques do not often show the coagulation status quantitatively, there are low-cost, compact, and user-friendly POC equipment available that can be used at the patient's bedside. POC diagnostics are promising techniques for evaluating anticoagulation impact when compared to conventional standard laboratory-based assays [144]. Laboratory-based testing is time-consuming, costly, and user-dependent. POC devices have several advantages, including reliability, quick response, and reduced sample consumption [145]. Some conventional coagulation monitoring methods, such as d-dimer, aPTT, PT/INR, and ECT, have recently been developed as POC devices [146]. Although the benefits of POC devices are well established, and various POC devices have been introduced, there is always a need to study novel devices to improve the accuracy and reliability of the results. Microfluidic devices, in particular, can be exploited for anticoagulation monitoring due to benefits such as low cost, simple operation, quick analysis, and reduced sample consumption. Furthermore, because microfluidic systems are high throughput, several samples can be analysed simultaneously with a small amount of sample in a single integrated platform [147]. Although microfluidic platforms have been developed and are currently being used in research laboratories, they are rarely marketed. Commercialisation of low-cost and high-throughput microfluidic devices for monitoring the coagulation pathway would provide unique advantages in the clinical management of patients receiving ECMO.

Conclusion

Although heparin is still the gold standard anticoagulation agent utilized in ECMO, heparin resistance and HIT are major clinical limitations. Next-generation anticoagulants also have their own shortcomings, which may include: short half-life, lack of antidots and restrictive monitoring methods that hamper broad clinical implementation. Several anticoagulant monitoring methods have been developed to track the patient's coagulation condition and adjust the anticoagulant dosage; some monitoring methods correlate better with specific anticoagulant agents than others. To establish an optimal balance of anticoagulation and bleeding requires a variety of approaches, rather than a single monitoring method, to monitor anticoagulation impact. Existing research efforts should be directed towards the development of novel anticoagulants, surface engineering to modify non-biocompatible surfaces, targeted drug delivery systems, and the development of real-time and POC monitoring methods that will result in fewer thrombosis and bleeding complications.

Acknowledgements

The authors would like to recognize the financial assistance provided by the National Health and Medical Research Council (NHMRC) and Monash University.

Declarations

Not applicable.
All authors have approved the manuscript for submission.

Competing interests

The authors declare no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Combes A, Price S, Slutsky AS, Brodie D. Temporary circulatory support for cardiogenic shock. The Lancet. 2020;396(10245):199–212.CrossRef Combes A, Price S, Slutsky AS, Brodie D. Temporary circulatory support for cardiogenic shock. The Lancet. 2020;396(10245):199–212.CrossRef
2.
Zurück zum Zitat Fanelli V, Giani M, Grasselli G, Mojoli F, Martucci G, Grazioli L, et al. Extracorporeal membrane oxygenation for COVID-19 and influenza H1N1 associated acute respiratory distress syndrome: a multicenter retrospective cohort study. Crit Care. 2022;26(1):34.CrossRef Fanelli V, Giani M, Grasselli G, Mojoli F, Martucci G, Grazioli L, et al. Extracorporeal membrane oxygenation for COVID-19 and influenza H1N1 associated acute respiratory distress syndrome: a multicenter retrospective cohort study. Crit Care. 2022;26(1):34.CrossRef
3.
Zurück zum Zitat Frerou A, Lesouhaitier M, Gregoire M, Uhel F, Gacouin A, Reizine F, et al. Venoarterial extracorporeal membrane oxygenation induces early immune alterations. Crit Care. 2021;25(1):9.CrossRef Frerou A, Lesouhaitier M, Gregoire M, Uhel F, Gacouin A, Reizine F, et al. Venoarterial extracorporeal membrane oxygenation induces early immune alterations. Crit Care. 2021;25(1):9.CrossRef
4.
Zurück zum Zitat Chan CHH, Ki KK, Zhang M, Asnicar C, Cho HJ, Ainola C, et al. Extracorporeal membrane oxygenation-induced hemolysis: An in vitro study to appraise causative factors. Membranes. 2021;11(5):313. Chan CHH, Ki KK, Zhang M, Asnicar C, Cho HJ, Ainola C, et al. Extracorporeal membrane oxygenation-induced hemolysis: An in vitro study to appraise causative factors. Membranes. 2021;11(5):313.
5.
Zurück zum Zitat Heuts S, Makhoul M, Mansouri AN, Taccone FS, Obeid A, Belliato M, et al. Defining and understanding the “extra-corporeal membrane oxygenation gap” in the veno-venous configuration: timing and causes of death. Artif Organs. 2022;46(3):349–61.CrossRef Heuts S, Makhoul M, Mansouri AN, Taccone FS, Obeid A, Belliato M, et al. Defining and understanding the “extra-corporeal membrane oxygenation gap” in the veno-venous configuration: timing and causes of death. Artif Organs. 2022;46(3):349–61.CrossRef
6.
Zurück zum Zitat Vaquer S, de Haro C, Peruga P, Oliva JC, Artigas A. Systematic review and meta-analysis of complications and mortality of veno-venous extracorporeal membrane oxygenation for refractory acute respiratory distress syndrome. Ann Intensive Care. 2017;7(1):1–13. Vaquer S, de Haro C, Peruga P, Oliva JC, Artigas A. Systematic review and meta-analysis of complications and mortality of veno-venous extracorporeal membrane oxygenation for refractory acute respiratory distress syndrome. Ann Intensive Care. 2017;7(1):1–13.
7.
Zurück zum Zitat Bembea MM, Annich G, Rycus P, Oldenburg G, Berkowitz I, Pronovost P. Variability in anticoagulation management of patients on extracorporeal membrane oxygenation: an international survey. Pediatr Crit Care Med. 2013;14(2): e77.CrossRef Bembea MM, Annich G, Rycus P, Oldenburg G, Berkowitz I, Pronovost P. Variability in anticoagulation management of patients on extracorporeal membrane oxygenation: an international survey. Pediatr Crit Care Med. 2013;14(2): e77.CrossRef
8.
Zurück zum Zitat Schünemann HJ, Cushman M, Burnett AE, Kahn SR, Beyer-Westendorf J, Spencer FA, et al. American Society of Hematology 2018 guidelines for management of venous thromboembolism: prophylaxis for hospitalized and nonhospitalized medical patients. Blood Adv. 2018;2(22):3198–225.CrossRef Schünemann HJ, Cushman M, Burnett AE, Kahn SR, Beyer-Westendorf J, Spencer FA, et al. American Society of Hematology 2018 guidelines for management of venous thromboembolism: prophylaxis for hospitalized and nonhospitalized medical patients. Blood Adv. 2018;2(22):3198–225.CrossRef
9.
Zurück zum Zitat Zangrillo A, Landoni G, Biondi-Zoccai G, Greco M, Greco T, Frati G, et al. A meta-analysis of complications and mortality of extracorporeal membrane oxygenation. Crit Care Resusc. 2013;15(3):172–8. Zangrillo A, Landoni G, Biondi-Zoccai G, Greco M, Greco T, Frati G, et al. A meta-analysis of complications and mortality of extracorporeal membrane oxygenation. Crit Care Resusc. 2013;15(3):172–8.
10.
Zurück zum Zitat Thiagarajan RR, Barbaro RP, Rycus PT, Mcmullan DM, Conrad SA, Fortenberry JD, et al. Extracorporeal life support organization registry international report 2016. ASAIO J. 2017;63(1):60–7.CrossRef Thiagarajan RR, Barbaro RP, Rycus PT, Mcmullan DM, Conrad SA, Fortenberry JD, et al. Extracorporeal life support organization registry international report 2016. ASAIO J. 2017;63(1):60–7.CrossRef
11.
Zurück zum Zitat Bhat AG, Golchin A, Pasupula DK, Hernandez-Montfort JA. Right sided intracardiac thrombosis during veno-arterial extracorporeal membrane oxygenation: a case report and literature review. Case Rep Crit Care. 2019;2019:8594681. Bhat AG, Golchin A, Pasupula DK, Hernandez-Montfort JA. Right sided intracardiac thrombosis during veno-arterial extracorporeal membrane oxygenation: a case report and literature review. Case Rep Crit Care. 2019;2019:8594681.
12.
Zurück zum Zitat Weber C, Deppe AC, Sabashnikov A, Slottosch I, Kuhn E, Eghbalzadeh K, et al. Left ventricular thrombus formation in patients undergoing femoral veno-arterial extracorporeal membrane oxygenation. Perfusion. 2018;33(4):283–8.CrossRef Weber C, Deppe AC, Sabashnikov A, Slottosch I, Kuhn E, Eghbalzadeh K, et al. Left ventricular thrombus formation in patients undergoing femoral veno-arterial extracorporeal membrane oxygenation. Perfusion. 2018;33(4):283–8.CrossRef
13.
Zurück zum Zitat Parzy G, Daviet F, Persico N, Rambaud R, Scemama U, Adda M, et al. Prevalence and risk factors for thrombotic complications following venovenous extracorporeal membrane oxygenation: a CT scan study. Crit Care Med. 2020;48(2):192–9.CrossRef Parzy G, Daviet F, Persico N, Rambaud R, Scemama U, Adda M, et al. Prevalence and risk factors for thrombotic complications following venovenous extracorporeal membrane oxygenation: a CT scan study. Crit Care Med. 2020;48(2):192–9.CrossRef
14.
Zurück zum Zitat Abruzzo A, Gorantla V, Thomas SE. Venous thromboembolic events in the setting of extracorporeal membrane oxygenation support in adults: a systematic review. Thromb Res. 2022;212:58–71.CrossRef Abruzzo A, Gorantla V, Thomas SE. Venous thromboembolic events in the setting of extracorporeal membrane oxygenation support in adults: a systematic review. Thromb Res. 2022;212:58–71.CrossRef
15.
Zurück zum Zitat Levy JH, Connors JM. Heparin resistance - clinical perspectives and management strategies. N Engl J Med. 2021;385(9):826–32.CrossRef Levy JH, Connors JM. Heparin resistance - clinical perspectives and management strategies. N Engl J Med. 2021;385(9):826–32.CrossRef
16.
Zurück zum Zitat Fisser C, Winkler M, Malfertheiner MV, Philipp A, Foltan M, Lunz D, et al. Argatroban versus heparin in patients without heparin-induced thrombocytopenia during venovenous extracorporeal membrane oxygenation: a propensity-score matched study. Crit Care. 2021;25(1):160.CrossRef Fisser C, Winkler M, Malfertheiner MV, Philipp A, Foltan M, Lunz D, et al. Argatroban versus heparin in patients without heparin-induced thrombocytopenia during venovenous extracorporeal membrane oxygenation: a propensity-score matched study. Crit Care. 2021;25(1):160.CrossRef
17.
Zurück zum Zitat Northrop MS, Sidonio RF, Phillips SE, Smith AH, Daphne HC, Pietsch JB, et al. The use of an extracorporeal membrane oxygenation anticoagulation laboratory protocol is associated with decreased blood product use, decreased hemorrhagic complications, and increased circuit life. Pediatr Crit Care Med. 2015;16(1):66–74.CrossRef Northrop MS, Sidonio RF, Phillips SE, Smith AH, Daphne HC, Pietsch JB, et al. The use of an extracorporeal membrane oxygenation anticoagulation laboratory protocol is associated with decreased blood product use, decreased hemorrhagic complications, and increased circuit life. Pediatr Crit Care Med. 2015;16(1):66–74.CrossRef
18.
Zurück zum Zitat Monagle P, Chan AKC, Goldenberg NA, Ichord RN, Journeycake JM, Nowak-Göttl U, et al. Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(2 Suppl):e737S-e801S. Monagle P, Chan AKC, Goldenberg NA, Ichord RN, Journeycake JM, Nowak-Göttl U, et al. Antithrombotic therapy in neonates and children: Antithrombotic Therapy and Prevention of Thrombosis, 9th ed: American College of Chest Physicians Evidence-Based Clinical Practice Guidelines. Chest. 2012;141(2 Suppl):e737S-e801S.
19.
Zurück zum Zitat Pollak U, Yacobobich J, Tamary H, Dagan O, Manor-Shulman O. Heparin-induced thrombocytopenia and extracorporeal membrane oxygenation: a case report and review of the literature. J Extra Corpor Technol. 2011;43(1):5–12. Pollak U, Yacobobich J, Tamary H, Dagan O, Manor-Shulman O. Heparin-induced thrombocytopenia and extracorporeal membrane oxygenation: a case report and review of the literature. J Extra Corpor Technol. 2011;43(1):5–12.
20.
Zurück zum Zitat Scott LK, Grier LR, Conrad SA. Heparin-induced thrombocytopenia in a pediatric patient receiving extracorporeal support and treated with argatroban. Pediatr Crit Care Med. 2006;7(3):255–7.CrossRef Scott LK, Grier LR, Conrad SA. Heparin-induced thrombocytopenia in a pediatric patient receiving extracorporeal support and treated with argatroban. Pediatr Crit Care Med. 2006;7(3):255–7.CrossRef
21.
Zurück zum Zitat Glick D, Dzierba AL, Abrams D, Muir J, Eisenberger A, Diuguid D, et al. Clinically suspected heparin-induced thrombocytopenia during extracorporeal membrane oxygenation. J Crit Care. 2015;30(6):1190–4.CrossRef Glick D, Dzierba AL, Abrams D, Muir J, Eisenberger A, Diuguid D, et al. Clinically suspected heparin-induced thrombocytopenia during extracorporeal membrane oxygenation. J Crit Care. 2015;30(6):1190–4.CrossRef
22.
Zurück zum Zitat Sokolovic M, Pratt AK, Vukicevic V, Sarumi M, Johnson LS, Shah NS. Platelet count trends and prevalence of heparin-induced thrombocytopenia in a cohort of extracorporeal membrane oxygenator patients. Crit Care Med. 2016;44(11):e1031–7.CrossRef Sokolovic M, Pratt AK, Vukicevic V, Sarumi M, Johnson LS, Shah NS. Platelet count trends and prevalence of heparin-induced thrombocytopenia in a cohort of extracorporeal membrane oxygenator patients. Crit Care Med. 2016;44(11):e1031–7.CrossRef
23.
Zurück zum Zitat Greinacher A, Farner B, Kroll H, Kohlmann T, Warkentin TE, Eichler P. Clinical features of heparin-induced thrombocytopenia including risk factors for thrombosis. a retrospective analysis of 408 patients. Thromb Haemost. 2005;94(1):132–5. Greinacher A, Farner B, Kroll H, Kohlmann T, Warkentin TE, Eichler P. Clinical features of heparin-induced thrombocytopenia including risk factors for thrombosis. a retrospective analysis of 408 patients. Thromb Haemost. 2005;94(1):132–5.
24.
Zurück zum Zitat Warkentin TE, Kelton JG. A 14-year study of heparin-induced thrombocytopenia. Am J Med. 1996;101(5):502–7.CrossRef Warkentin TE, Kelton JG. A 14-year study of heparin-induced thrombocytopenia. Am J Med. 1996;101(5):502–7.CrossRef
25.
Zurück zum Zitat Gates R, Yost P, Parker B. The use of bivalirudin for cardiopulmonary bypass anticoagulation in pediatric heparin-induced thrombocytopenia patients. Artif Organs. 2010;34(8):667–9. Gates R, Yost P, Parker B. The use of bivalirudin for cardiopulmonary bypass anticoagulation in pediatric heparin-induced thrombocytopenia patients. Artif Organs. 2010;34(8):667–9.
26.
Zurück zum Zitat Erdoes G, Ortmann E, Martinez Lopez De Arroyabe B, Reid C, Koster A. Role of Bivalirudin for Anticoagulation in Adult Perioperative Cardiothoracic Practice. J Cardiothorac Vasc Anesth. 2020;34(8):2207–14. Erdoes G, Ortmann E, Martinez Lopez De Arroyabe B, Reid C, Koster A. Role of Bivalirudin for Anticoagulation in Adult Perioperative Cardiothoracic Practice. J Cardiothorac Vasc Anesth. 2020;34(8):2207–14.
27.
Zurück zum Zitat Walker EA, Roberts AJ, Louie EL, Dager WE. Bivalirudin dosing requirements in adult patients on extracorporeal life support with or without continuous renal replacement therapy. Asaio j. 2019;65(2):134–8.CrossRef Walker EA, Roberts AJ, Louie EL, Dager WE. Bivalirudin dosing requirements in adult patients on extracorporeal life support with or without continuous renal replacement therapy. Asaio j. 2019;65(2):134–8.CrossRef
28.
Zurück zum Zitat Hirsh J, O’Donnell M, Weitz JI. New anticoagulants. Blood. 2005;105(2):453–63.CrossRef Hirsh J, O’Donnell M, Weitz JI. New anticoagulants. Blood. 2005;105(2):453–63.CrossRef
29.
Zurück zum Zitat Hirsh J, O’Donnell M, Eikelboom JW. Beyond unfractionated heparin and warfarin: current and future advances. Circulation. 2007;116(5):552–60.CrossRef Hirsh J, O’Donnell M, Eikelboom JW. Beyond unfractionated heparin and warfarin: current and future advances. Circulation. 2007;116(5):552–60.CrossRef
30.
Zurück zum Zitat Berei TJ, Lillyblad MP, Wilson KJ, Garberich RF, Hryniewicz KM. Evaluation of systemic heparin versus bivalirudin in adult patients supported by extracorporeal membrane oxygenation. Asaio j. 2018;64(5):623–9.CrossRef Berei TJ, Lillyblad MP, Wilson KJ, Garberich RF, Hryniewicz KM. Evaluation of systemic heparin versus bivalirudin in adult patients supported by extracorporeal membrane oxygenation. Asaio j. 2018;64(5):623–9.CrossRef
31.
Zurück zum Zitat Horwitz JR, Cofer BR, Warner BW, Cheu HW, Lally KP. A multicenter trial of 6-aminocaproic acid (Amicar) in the prevention of bleeding in infants on ECMO. J Pediatr Surg. 1998;33(11):1610–3.CrossRef Horwitz JR, Cofer BR, Warner BW, Cheu HW, Lally KP. A multicenter trial of 6-aminocaproic acid (Amicar) in the prevention of bleeding in infants on ECMO. J Pediatr Surg. 1998;33(11):1610–3.CrossRef
32.
Zurück zum Zitat Pappalardo F, Maj G, Scandroglio A, Sampietro F, Zangrillo A, Koster A. Bioline heparin-coated ECMO with bivalirudin anticoagulation in a patient with acute heparin-induced thrombocytopenia: the immune reaction appeared to continue unabated. Perfusion. 2009;24(2):135–7.CrossRef Pappalardo F, Maj G, Scandroglio A, Sampietro F, Zangrillo A, Koster A. Bioline heparin-coated ECMO with bivalirudin anticoagulation in a patient with acute heparin-induced thrombocytopenia: the immune reaction appeared to continue unabated. Perfusion. 2009;24(2):135–7.CrossRef
33.
Zurück zum Zitat Koster A, Weng Y, Böttcher W, Gromann T, Kuppe H, Hetzer R. Successful use of bivalirudin as anticoagulant for ECMO in a patient with acute HIT. Ann Thorac Surg. 2007;83(5):1865–7.CrossRef Koster A, Weng Y, Böttcher W, Gromann T, Kuppe H, Hetzer R. Successful use of bivalirudin as anticoagulant for ECMO in a patient with acute HIT. Ann Thorac Surg. 2007;83(5):1865–7.CrossRef
34.
Zurück zum Zitat Ranucci M, Ballotta A, Kandil H, Isgrò G, Carlucci C, Baryshnikova E, et al. Bivalirudin-based versus conventional heparin anticoagulation for postcardiotomy extracorporeal membrane oxygenation. Crit Care. 2011;15(6):R275.CrossRef Ranucci M, Ballotta A, Kandil H, Isgrò G, Carlucci C, Baryshnikova E, et al. Bivalirudin-based versus conventional heparin anticoagulation for postcardiotomy extracorporeal membrane oxygenation. Crit Care. 2011;15(6):R275.CrossRef
35.
Zurück zum Zitat Wong JJ-M, Lam JCM, Mok YH, Lee JH. Anticoagulation in extracorporeal membrane oxygenation. J Emergency Crit Care Med. 2018;2:12.CrossRef Wong JJ-M, Lam JCM, Mok YH, Lee JH. Anticoagulation in extracorporeal membrane oxygenation. J Emergency Crit Care Med. 2018;2:12.CrossRef
36.
Zurück zum Zitat Nagle EL, Dager WE, Duby JJ, Roberts AJ, Kenny LE, Murthy MS, et al. Bivalirudin in pediatric patients maintained on extracorporeal life support. Pediatr Crit Care Med. 2013;14(4):e182–8.CrossRef Nagle EL, Dager WE, Duby JJ, Roberts AJ, Kenny LE, Murthy MS, et al. Bivalirudin in pediatric patients maintained on extracorporeal life support. Pediatr Crit Care Med. 2013;14(4):e182–8.CrossRef
37.
Zurück zum Zitat Casserly IP, Kereiakes DJ, Gray WA, Gibson PH, Lauer MA, Reginelli JP, et al. Point-of-care ecarin clotting time versus activated clotting time in correlation with bivalirudin concentration. Thromb Res. 2004;113(2):115–21.CrossRef Casserly IP, Kereiakes DJ, Gray WA, Gibson PH, Lauer MA, Reginelli JP, et al. Point-of-care ecarin clotting time versus activated clotting time in correlation with bivalirudin concentration. Thromb Res. 2004;113(2):115–21.CrossRef
38.
Zurück zum Zitat Pieri M, Agracheva N, Bonaveglio E, Greco T, De Bonis M, Covello RD, et al. Bivalirudin versus heparin as an anticoagulant during extracorporeal membrane oxygenation: a case-control study. J Cardiothorac Vasc Anesth. 2013;27(1):30–4.CrossRef Pieri M, Agracheva N, Bonaveglio E, Greco T, De Bonis M, Covello RD, et al. Bivalirudin versus heparin as an anticoagulant during extracorporeal membrane oxygenation: a case-control study. J Cardiothorac Vasc Anesth. 2013;27(1):30–4.CrossRef
39.
Zurück zum Zitat Jyoti A, Maheshwari A, Daniel E, Motihar A, Bhathiwal RS, Sharma D. Bivalirudin in venovenous extracorporeal membrane oxygenation. J Extra Corpor Technol. 2014;46(1):94–7. Jyoti A, Maheshwari A, Daniel E, Motihar A, Bhathiwal RS, Sharma D. Bivalirudin in venovenous extracorporeal membrane oxygenation. J Extra Corpor Technol. 2014;46(1):94–7.
40.
Zurück zum Zitat M’Pembele R, Roth S, Metzger A, Nucaro A, Stroda A, Polzin A, et al. Evaluation of clinical outcomes in patients treated with heparin or direct thrombin inhibitors during extracorporeal membrane oxygenation: a systematic review and meta-analysis. Thromb J. 2022;20(1):42.CrossRef M’Pembele R, Roth S, Metzger A, Nucaro A, Stroda A, Polzin A, et al. Evaluation of clinical outcomes in patients treated with heparin or direct thrombin inhibitors during extracorporeal membrane oxygenation: a systematic review and meta-analysis. Thromb J. 2022;20(1):42.CrossRef
41.
Zurück zum Zitat Tsu LV, Dager WE. Bivalirudin dosing adjustments for reduced renal function with or without hemodialysis in the management of heparin-induced thrombocytopenia. Ann Pharmacother. 2011;45(10):1185–92.CrossRef Tsu LV, Dager WE. Bivalirudin dosing adjustments for reduced renal function with or without hemodialysis in the management of heparin-induced thrombocytopenia. Ann Pharmacother. 2011;45(10):1185–92.CrossRef
42.
Zurück zum Zitat Ranucci M. Bivalirudin and post-cardiotomy ECMO: a word of caution. Crit Care. 2012;16(3):427.CrossRef Ranucci M. Bivalirudin and post-cardiotomy ECMO: a word of caution. Crit Care. 2012;16(3):427.CrossRef
43.
Zurück zum Zitat Cardinale M, Ha M, Liu MH, Reardon DP. Direct Thrombin Inhibitor Resistance and Possible Mechanisms. Hosp Pharm. 2016;51(11):922–7.CrossRef Cardinale M, Ha M, Liu MH, Reardon DP. Direct Thrombin Inhibitor Resistance and Possible Mechanisms. Hosp Pharm. 2016;51(11):922–7.CrossRef
44.
Zurück zum Zitat Schmid C. Extracorporeal Circulation and Myocardial Protection in Adult Cardiac Surgery. Cardiac Surgery: Springer; 2017. p. 111–52. Schmid C. Extracorporeal Circulation and Myocardial Protection in Adult Cardiac Surgery. Cardiac Surgery: Springer; 2017. p. 111–52.
45.
Zurück zum Zitat Koster A, Weng Y, Böttcher W, Gromann T, Kuppe H, Hetzer R. Successful use of bivalirudin as anticoagulant for ECMO in a patient with acute HIT. Ann Thorac Surg. 2007;83(5):1865–7.CrossRef Koster A, Weng Y, Böttcher W, Gromann T, Kuppe H, Hetzer R. Successful use of bivalirudin as anticoagulant for ECMO in a patient with acute HIT. Ann Thorac Surg. 2007;83(5):1865–7.CrossRef
46.
Zurück zum Zitat van der Staak FH, de Haan AF, Geven WB, Festen C. Surgical repair of congenital diaphragmatic hernia during extracorporeal membrane oxygenation: hemorrhagic complications and the effect of tranexamic acid. J Pediatr Surg. 1997;32(4):594–9.CrossRef van der Staak FH, de Haan AF, Geven WB, Festen C. Surgical repair of congenital diaphragmatic hernia during extracorporeal membrane oxygenation: hemorrhagic complications and the effect of tranexamic acid. J Pediatr Surg. 1997;32(4):594–9.CrossRef
47.
Zurück zum Zitat Young G, Yonekawa KE, Nakagawa P, Nugent DJ. Argatroban as an alternative to heparin in extracorporeal membrane oxygenation circuits. Perfusion. 2004;19(5):283–8.CrossRef Young G, Yonekawa KE, Nakagawa P, Nugent DJ. Argatroban as an alternative to heparin in extracorporeal membrane oxygenation circuits. Perfusion. 2004;19(5):283–8.CrossRef
48.
Zurück zum Zitat Phillips MR, Khoury AI, Ashton RF, Cairns BA, Charles AG. The dosing and monitoring of argatroban for heparin-induced thrombocytopenia during extracorporeal membrane oxygenation: a word of caution. Anaesth Intensive Care. 2014;42(1):97–8.CrossRef Phillips MR, Khoury AI, Ashton RF, Cairns BA, Charles AG. The dosing and monitoring of argatroban for heparin-induced thrombocytopenia during extracorporeal membrane oxygenation: a word of caution. Anaesth Intensive Care. 2014;42(1):97–8.CrossRef
49.
Zurück zum Zitat Warkentin TE, Cook RJ, Marder VJ, Sheppard JA, Moore JC, Eriksson BI, et al. Anti-platelet factor 4/heparin antibodies in orthopedic surgery patients receiving antithrombotic prophylaxis with fondaparinux or enoxaparin. Blood. 2005;106(12):3791–6.CrossRef Warkentin TE, Cook RJ, Marder VJ, Sheppard JA, Moore JC, Eriksson BI, et al. Anti-platelet factor 4/heparin antibodies in orthopedic surgery patients receiving antithrombotic prophylaxis with fondaparinux or enoxaparin. Blood. 2005;106(12):3791–6.CrossRef
50.
Zurück zum Zitat Parlar AI, Sayar U, Cevirme D, Yuruk MA, Mataraci I. Successful use of fondaparinux in a patient with heparin-induced thrombocytopenia while on extracorporeal membrane oxygenation after mitral valve redo surgery. Int J Artif Organs. 2014;37(4):344–7.CrossRef Parlar AI, Sayar U, Cevirme D, Yuruk MA, Mataraci I. Successful use of fondaparinux in a patient with heparin-induced thrombocytopenia while on extracorporeal membrane oxygenation after mitral valve redo surgery. Int J Artif Organs. 2014;37(4):344–7.CrossRef
51.
Zurück zum Zitat Greinacher A, Warkentin TE. The direct thrombin inhibitor hirudin. Thromb Haemost. 2008;99(5):819–29. Greinacher A, Warkentin TE. The direct thrombin inhibitor hirudin. Thromb Haemost. 2008;99(5):819–29.
52.
Zurück zum Zitat Ota T, Okada K, Kano H, Okita Y. Cardiopulmonary bypass using nafamostat mesilate for patients with infective endocarditis and recent intracranial hemorrhage. Interact Cardiovasc Thorac Surg. 2007;6(3):270–3.CrossRef Ota T, Okada K, Kano H, Okita Y. Cardiopulmonary bypass using nafamostat mesilate for patients with infective endocarditis and recent intracranial hemorrhage. Interact Cardiovasc Thorac Surg. 2007;6(3):270–3.CrossRef
53.
Zurück zum Zitat Lim JY, Kim JB, Choo SJ, Chung CH, Lee JW, Jung SH. Anticoagulation during extracorporeal membrane oxygenation; nafamostat mesilate versus heparin. Ann Thorac Surg. 2016;102(2):534–9.CrossRef Lim JY, Kim JB, Choo SJ, Chung CH, Lee JW, Jung SH. Anticoagulation during extracorporeal membrane oxygenation; nafamostat mesilate versus heparin. Ann Thorac Surg. 2016;102(2):534–9.CrossRef
54.
Zurück zum Zitat Park JH, Her C, Min HK, Kim DK, Park SH, Jang HJ. Nafamostat mesilate as a regional anticoagulant in patients with bleeding complications during extracorporeal membrane oxygenation. Int J Artif Organs. 2015;38(11):595–9.CrossRef Park JH, Her C, Min HK, Kim DK, Park SH, Jang HJ. Nafamostat mesilate as a regional anticoagulant in patients with bleeding complications during extracorporeal membrane oxygenation. Int J Artif Organs. 2015;38(11):595–9.CrossRef
55.
Zurück zum Zitat Han SJ, Kim HS, Kim KI, Whang SM, Hong KS, Lee WK, et al. Use of nafamostat mesilate as an anticoagulant during extracorporeal membrane oxygenation. J Korean Med Sci. 2011;26(7):945–50.CrossRef Han SJ, Kim HS, Kim KI, Whang SM, Hong KS, Lee WK, et al. Use of nafamostat mesilate as an anticoagulant during extracorporeal membrane oxygenation. J Korean Med Sci. 2011;26(7):945–50.CrossRef
56.
Zurück zum Zitat Nagaya M, Futamura M, Kato J, Niimi N, Fukuta S. Application of a new anticoagulant (Nafamostat Mesilate) to control hemorrhagic complications during extracorporeal membrane oxygenation–a preliminary report. J Pediatr Surg. 1997;32(4):531–5.CrossRef Nagaya M, Futamura M, Kato J, Niimi N, Fukuta S. Application of a new anticoagulant (Nafamostat Mesilate) to control hemorrhagic complications during extracorporeal membrane oxygenation–a preliminary report. J Pediatr Surg. 1997;32(4):531–5.CrossRef
57.
58.
Zurück zum Zitat Warkentin TE, Greinacher A. Heparin-induced thrombocytopenia: recognition, treatment, and prevention: the Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy. Chest. 2004;126(3 Suppl):311s-37s. Warkentin TE, Greinacher A. Heparin-induced thrombocytopenia: recognition, treatment, and prevention: the Seventh ACCP Conference on Antithrombotic and Thrombolytic Therapy. Chest. 2004;126(3 Suppl):311s-37s.
59.
Zurück zum Zitat Hirsh J, Fuster V, Ansell J, Halperin JL. american heart association/american college of cardiology foundation guide to warfarin therapy. Circulation. 2003;107(12):1692–711.CrossRef Hirsh J, Fuster V, Ansell J, Halperin JL. american heart association/american college of cardiology foundation guide to warfarin therapy. Circulation. 2003;107(12):1692–711.CrossRef
60.
Zurück zum Zitat Lee JH, Kim SW. Successful management of warfarin-exacerbated diffuse alveolar hemorrhage using an extracorporeal membrane oxygenation. Multidiscip Respir Med. 2013;8(1):16.CrossRef Lee JH, Kim SW. Successful management of warfarin-exacerbated diffuse alveolar hemorrhage using an extracorporeal membrane oxygenation. Multidiscip Respir Med. 2013;8(1):16.CrossRef
61.
Zurück zum Zitat Nguyen TP, Phan XT, Huynh DQ, Viet Truong HT, Hai Le YN, Nguyen TM, et al. Monitoring Unfractionated Heparin in Adult Patients Undergoing Extracorporeal Membrane Oxygenation (ECMO): ACT, APTT, or ANTI-XA? Crit Care Res Pract. 2021;2021:5579936. Nguyen TP, Phan XT, Huynh DQ, Viet Truong HT, Hai Le YN, Nguyen TM, et al. Monitoring Unfractionated Heparin in Adult Patients Undergoing Extracorporeal Membrane Oxygenation (ECMO): ACT, APTT, or ANTI-XA? Crit Care Res Pract. 2021;2021:5579936.
62.
Zurück zum Zitat Teruya J, Bruzdoski K, Hensch L, Hui SKR, Kostousov V. How to best monitor bivalirudin anticoagulant effect for ECMO and VAD—Comparison of four assay methods. Int J Lab Hematol. 2022;44(3):589–94. Teruya J, Bruzdoski K, Hensch L, Hui SKR, Kostousov V. How to best monitor bivalirudin anticoagulant effect for ECMO and VAD—Comparison of four assay methods. Int J Lab Hematol. 2022;44(3):589–94.
63.
Zurück zum Zitat Fisser C, Winkler M, Malfertheiner MV, Philipp A, Foltan M, Lunz D, et al. Argatroban versus heparin in patients without heparin-induced thrombocytopenia during venovenous extracorporeal membrane oxygenation: a propensity-score matched study. Crit Care. 2021;25(1):1–10.CrossRef Fisser C, Winkler M, Malfertheiner MV, Philipp A, Foltan M, Lunz D, et al. Argatroban versus heparin in patients without heparin-induced thrombocytopenia during venovenous extracorporeal membrane oxygenation: a propensity-score matched study. Crit Care. 2021;25(1):1–10.CrossRef
64.
Zurück zum Zitat Myllylahti L, Pitkänen H, Magnani H, Lassila R. Experience of danaparoid to treat vaccine-induced immune thrombocytopenia and thrombosis, VITT. Thromb J. 2022;20(1):4.CrossRef Myllylahti L, Pitkänen H, Magnani H, Lassila R. Experience of danaparoid to treat vaccine-induced immune thrombocytopenia and thrombosis, VITT. Thromb J. 2022;20(1):4.CrossRef
65.
Zurück zum Zitat Serasli E, Antoniadou M, Tsara V, Kalpakidis V, Megalopoulos A, Trellopoulos G, et al. Successful management of acute thromboembolic disease complicated with heparin induced thrombocytopenia type II (HIT II): a case series. Thromb J. 2008;6(1):9.CrossRef Serasli E, Antoniadou M, Tsara V, Kalpakidis V, Megalopoulos A, Trellopoulos G, et al. Successful management of acute thromboembolic disease complicated with heparin induced thrombocytopenia type II (HIT II): a case series. Thromb J. 2008;6(1):9.CrossRef
66.
Zurück zum Zitat Moon K, Hong K-W, Bae I-G. Treatment effect of nafamostat mesylate in patients with COVID-19 pneumonia: study protocol for a randomized controlled trial. Trials. 2021;22(1):832.CrossRef Moon K, Hong K-W, Bae I-G. Treatment effect of nafamostat mesylate in patients with COVID-19 pneumonia: study protocol for a randomized controlled trial. Trials. 2021;22(1):832.CrossRef
67.
Zurück zum Zitat Lippi G, Favaloro EJ. Laboratory monitoring of warfarin in the era of direct oral anticoagulants. The Lancet Haematology. 2015;2(6):e223–4.CrossRef Lippi G, Favaloro EJ. Laboratory monitoring of warfarin in the era of direct oral anticoagulants. The Lancet Haematology. 2015;2(6):e223–4.CrossRef
68.
Zurück zum Zitat Tralhão A, Aguiar C, Ferreira J, Rebocho MJ, Santos E, Martins D, et al. Dabigatran reversal with idarucizumab in a patient undergoing heart transplantation: first European report. Thromb J. 2017;15(1):23.CrossRef Tralhão A, Aguiar C, Ferreira J, Rebocho MJ, Santos E, Martins D, et al. Dabigatran reversal with idarucizumab in a patient undergoing heart transplantation: first European report. Thromb J. 2017;15(1):23.CrossRef
69.
Zurück zum Zitat Harder S. Pharmacokinetic and pharmacodynamic evaluation of rivaroxaban: considerations for the treatment of venous thromboembolism. Thromb J. 2014;12(1):22.CrossRef Harder S. Pharmacokinetic and pharmacodynamic evaluation of rivaroxaban: considerations for the treatment of venous thromboembolism. Thromb J. 2014;12(1):22.CrossRef
70.
Zurück zum Zitat Berk ZB, Shah A, Sun W, Griffith BP, Wu ZJ. An ex vivo comparison of partial thromboplastin time and activated clotting time for heparin anticoagulation in an ovine model. Artif Organs. 2022;46(3):501–5.CrossRef Berk ZB, Shah A, Sun W, Griffith BP, Wu ZJ. An ex vivo comparison of partial thromboplastin time and activated clotting time for heparin anticoagulation in an ovine model. Artif Organs. 2022;46(3):501–5.CrossRef
71.
Zurück zum Zitat Honore PM, Barreto Gutierrez L, Kugener L, Redant S, Attou R, Gallerani A, et al. Use of multiple laboratory tests including anti-factor Xa to optimally manage anticoagulation during ECMO. Crit Care. 2020;24(1):427.CrossRef Honore PM, Barreto Gutierrez L, Kugener L, Redant S, Attou R, Gallerani A, et al. Use of multiple laboratory tests including anti-factor Xa to optimally manage anticoagulation during ECMO. Crit Care. 2020;24(1):427.CrossRef
72.
Zurück zum Zitat Villalba CAF, Brogan TV, McMullan DM, Yalon L, Jordan DI, Chandler WL. Conversion from activated clotting time to anti-Xa heparin activity assay for heparin monitoring during extracorporeal membrane oxygenation. Crit Care Med. 2020;48(12):e1179–84.CrossRef Villalba CAF, Brogan TV, McMullan DM, Yalon L, Jordan DI, Chandler WL. Conversion from activated clotting time to anti-Xa heparin activity assay for heparin monitoring during extracorporeal membrane oxygenation. Crit Care Med. 2020;48(12):e1179–84.CrossRef
73.
Zurück zum Zitat Chandel A, Patolia S, Looby M, Bade N, Khangoora V, King CS. Association of D-dimer and fibrinogen with hypercoagulability in COVID-19 requiring extracorporeal membrane oxygenation. J Intensive Care Med. 2021;36(6):689–95.CrossRef Chandel A, Patolia S, Looby M, Bade N, Khangoora V, King CS. Association of D-dimer and fibrinogen with hypercoagulability in COVID-19 requiring extracorporeal membrane oxygenation. J Intensive Care Med. 2021;36(6):689–95.CrossRef
74.
Zurück zum Zitat Onundarson PT, Francis CW, Indridason OS, Arnar DO, Bjornsson ES, Magnusson MK, et al. Fiix-prothrombin time versus standard prothrombin time for monitoring of warfarin anticoagulation: a single centre, double-blind, randomised, non-inferiority trial. The Lancet Haematology. 2015;2(6):e231–40.CrossRef Onundarson PT, Francis CW, Indridason OS, Arnar DO, Bjornsson ES, Magnusson MK, et al. Fiix-prothrombin time versus standard prothrombin time for monitoring of warfarin anticoagulation: a single centre, double-blind, randomised, non-inferiority trial. The Lancet Haematology. 2015;2(6):e231–40.CrossRef
75.
Zurück zum Zitat Gosselin RC, Douxfils J. Ecarin based coagulation testing. Am J Hematol. 2020;95(7):863–9.CrossRef Gosselin RC, Douxfils J. Ecarin based coagulation testing. Am J Hematol. 2020;95(7):863–9.CrossRef
76.
Zurück zum Zitat Olson JD, Arkin CF, Brandt JT, Cunningham MT, Giles A, Koepke JA, et al. College of American Pathologists Conference XXXI on laboratory monitoring of anticoagulant therapy: laboratory monitoring of unfractionated heparin therapy. Arch Pathol Lab Med. 1998;122(9):782–98. Olson JD, Arkin CF, Brandt JT, Cunningham MT, Giles A, Koepke JA, et al. College of American Pathologists Conference XXXI on laboratory monitoring of anticoagulant therapy: laboratory monitoring of unfractionated heparin therapy. Arch Pathol Lab Med. 1998;122(9):782–98.
77.
Zurück zum Zitat Mohammadi Aria M, Erten A, Yalcin O. Technology advancements in blood coagulation measurements for point-of-care diagnostic testing. Front Bioeng Biotechnol. 2019;7:395.CrossRef Mohammadi Aria M, Erten A, Yalcin O. Technology advancements in blood coagulation measurements for point-of-care diagnostic testing. Front Bioeng Biotechnol. 2019;7:395.CrossRef
78.
Zurück zum Zitat Ignjatovic V. Activated partial thromboplastin time. Methods Mol Biol. 2013;992:111–20.CrossRef Ignjatovic V. Activated partial thromboplastin time. Methods Mol Biol. 2013;992:111–20.CrossRef
79.
Zurück zum Zitat Baglin T, Barrowcliffe TW, Cohen A, Greaves M. Guidelines on the use and monitoring of heparin. Br J Haematol. 2006;133(1):19–34.CrossRef Baglin T, Barrowcliffe TW, Cohen A, Greaves M. Guidelines on the use and monitoring of heparin. Br J Haematol. 2006;133(1):19–34.CrossRef
80.
Zurück zum Zitat Ogawa F, Oi Y, Nakajima K, Matsumura R, Nakagawa T, Miyagawa T, et al. An evaluation of venous thromboembolism by whole-body enhanced CT scan for critical COVID-19 pneumonia with markedly rises of coagulopathy related factors: a case series study. Thromb J. 2021;19(1):26.CrossRef Ogawa F, Oi Y, Nakajima K, Matsumura R, Nakagawa T, Miyagawa T, et al. An evaluation of venous thromboembolism by whole-body enhanced CT scan for critical COVID-19 pneumonia with markedly rises of coagulopathy related factors: a case series study. Thromb J. 2021;19(1):26.CrossRef
81.
Zurück zum Zitat Bates SM, Weitz JI, Johnston M, Hirsh J, Ginsberg JS. Use of a fixed activated partial thromboplastin time ratio to establish a therapeutic range for unfractionated heparin. Arch Intern Med. 2001;161(3):385–91.CrossRef Bates SM, Weitz JI, Johnston M, Hirsh J, Ginsberg JS. Use of a fixed activated partial thromboplastin time ratio to establish a therapeutic range for unfractionated heparin. Arch Intern Med. 2001;161(3):385–91.CrossRef
82.
Zurück zum Zitat Price EA, Jin J, Nguyen HM, Krishnan G, Bowen R, Zehnder JL. Discordant aPTT and anti-Xa values and outcomes in hospitalized patients treated with intravenous unfractionated heparin. Ann Pharmacother. 2013;47(2):151–8.CrossRef Price EA, Jin J, Nguyen HM, Krishnan G, Bowen R, Zehnder JL. Discordant aPTT and anti-Xa values and outcomes in hospitalized patients treated with intravenous unfractionated heparin. Ann Pharmacother. 2013;47(2):151–8.CrossRef
83.
Zurück zum Zitat Kitchen S. Problems in laboratory monitoring of heparin dosage. Br J Haematol. 2000;111(2):397–406. Kitchen S. Problems in laboratory monitoring of heparin dosage. Br J Haematol. 2000;111(2):397–406.
84.
Zurück zum Zitat Newall F. Anti-factor Xa (anti-Xa) assay. Methods Mol Biol. 2013;992:265–72.CrossRef Newall F. Anti-factor Xa (anti-Xa) assay. Methods Mol Biol. 2013;992:265–72.CrossRef
85.
Zurück zum Zitat Gehrie E, Laposata M. Test of the month: the chromogenic antifactor Xa assay. Am J Hematol. 2012;87(2):194–6.CrossRef Gehrie E, Laposata M. Test of the month: the chromogenic antifactor Xa assay. Am J Hematol. 2012;87(2):194–6.CrossRef
86.
Zurück zum Zitat Khaja WA, Bilen O, Lukner RB, Edwards R, Teruya J. Evaluation of heparin assay for coagulation management in newborns undergoing ECMO. Am J Clin Pathol. 2010;134(6):950–4.CrossRef Khaja WA, Bilen O, Lukner RB, Edwards R, Teruya J. Evaluation of heparin assay for coagulation management in newborns undergoing ECMO. Am J Clin Pathol. 2010;134(6):950–4.CrossRef
87.
Zurück zum Zitat Ignjatovic V, Lai C, Summerhayes R, Mathesius U, Tawfilis S, Perugini MA, et al. Age-related differences in plasma proteins: how plasma proteins change from neonates to adults. PLoS ONE. 2011;6(2): e17213.CrossRef Ignjatovic V, Lai C, Summerhayes R, Mathesius U, Tawfilis S, Perugini MA, et al. Age-related differences in plasma proteins: how plasma proteins change from neonates to adults. PLoS ONE. 2011;6(2): e17213.CrossRef
88.
Zurück zum Zitat Vera-Aguilera J, Yousef H, Beltran-Melgarejo D, Teng TH, Jan R, Mok M, et al. Clinical scenarios for discordant anti-xa. Adv Hematol. 2016;2016:4054806.CrossRef Vera-Aguilera J, Yousef H, Beltran-Melgarejo D, Teng TH, Jan R, Mok M, et al. Clinical scenarios for discordant anti-xa. Adv Hematol. 2016;2016:4054806.CrossRef
89.
Zurück zum Zitat Ranucci M, Cotza M, Isgrò G, Carboni G, Ballotta A, Baryshnikova E. Anti-factor xa-based anticoagulation during extracorporeal membrane oxygenation: potential problems and possible solutions. Semin Thromb Hemost. 2020;46(4):419–27.CrossRef Ranucci M, Cotza M, Isgrò G, Carboni G, Ballotta A, Baryshnikova E. Anti-factor xa-based anticoagulation during extracorporeal membrane oxygenation: potential problems and possible solutions. Semin Thromb Hemost. 2020;46(4):419–27.CrossRef
90.
Zurück zum Zitat Kostousov V, Nguyen K, Hundalani SG, Teruya J. The influence of free hemoglobin and bilirubin on heparin monitoring by activated partial thromboplastin time and anti-Xa assay. Arch Pathol Lab Med. 2014;138(11):1503–6.CrossRef Kostousov V, Nguyen K, Hundalani SG, Teruya J. The influence of free hemoglobin and bilirubin on heparin monitoring by activated partial thromboplastin time and anti-Xa assay. Arch Pathol Lab Med. 2014;138(11):1503–6.CrossRef
91.
Zurück zum Zitat Liveris A, Bello RA, Friedmann P, Duffy MA, Manwani D, Killinger JS, et al. Anti-factor Xa assay is a superior correlate of heparin dose than activated partial thromboplastin time or activated clotting time in pediatric extracorporeal membrane oxygenation*. Pediatr Crit Care Med. 2014;15(2):e72–9.CrossRef Liveris A, Bello RA, Friedmann P, Duffy MA, Manwani D, Killinger JS, et al. Anti-factor Xa assay is a superior correlate of heparin dose than activated partial thromboplastin time or activated clotting time in pediatric extracorporeal membrane oxygenation*. Pediatr Crit Care Med. 2014;15(2):e72–9.CrossRef
92.
Zurück zum Zitat Delmas C, Jacquemin A, Vardon-Bounes F, Georges B, Guerrero F, Hernandez N, et al. Anticoagulation monitoring under ECMO support: a comparative study between the activated coagulation time and the anti-xa activity assay. J Intensive Care Med. 2020;35(7):679–86.CrossRef Delmas C, Jacquemin A, Vardon-Bounes F, Georges B, Guerrero F, Hernandez N, et al. Anticoagulation monitoring under ECMO support: a comparative study between the activated coagulation time and the anti-xa activity assay. J Intensive Care Med. 2020;35(7):679–86.CrossRef
93.
Zurück zum Zitat O’Meara LC, Alten JA, Goldberg KG, Timpa JG, Phillips J, Laney D, et al. Anti-xa directed protocol for anticoagulation management in children supported with extracorporeal membrane oxygenation. Asaio j. 2015;61(3):339–44.CrossRef O’Meara LC, Alten JA, Goldberg KG, Timpa JG, Phillips J, Laney D, et al. Anti-xa directed protocol for anticoagulation management in children supported with extracorporeal membrane oxygenation. Asaio j. 2015;61(3):339–44.CrossRef
94.
Zurück zum Zitat Nankervis CA, Preston TJ, Dysart KC, Wilkinson WD, Chicoine LG, Welty SE, et al. Assessing heparin dosing in neonates on venoarterial extracorporeal membrane oxygenation. Asaio j. 2007;53(1):111–4.CrossRef Nankervis CA, Preston TJ, Dysart KC, Wilkinson WD, Chicoine LG, Welty SE, et al. Assessing heparin dosing in neonates on venoarterial extracorporeal membrane oxygenation. Asaio j. 2007;53(1):111–4.CrossRef
95.
Zurück zum Zitat Bembea MM, Schwartz JM, Shah N, Colantuoni E, Lehmann CU, Kickler T, et al. Anticoagulation monitoring during pediatric extracorporeal membrane oxygenation. ASAIO Journal (American Society for Artificial Internal Organs: 1992). 2013;59(1):63. Bembea MM, Schwartz JM, Shah N, Colantuoni E, Lehmann CU, Kickler T, et al. Anticoagulation monitoring during pediatric extracorporeal membrane oxygenation. ASAIO Journal (American Society for Artificial Internal Organs: 1992). 2013;59(1):63.
96.
Zurück zum Zitat Niebler RA, Parker H, Hoffman GM. Impact of anticoagulation and circuit technology on complications during extracorporeal membrane oxygenation. Asaio j. 2019;65(3):270–6.CrossRef Niebler RA, Parker H, Hoffman GM. Impact of anticoagulation and circuit technology on complications during extracorporeal membrane oxygenation. Asaio j. 2019;65(3):270–6.CrossRef
97.
Zurück zum Zitat Colman E, Yin EB, Laine G, Chatterjee S, Saatee S, Herlihy JP, et al. Evaluation of a heparin monitoring protocol for extracorporeal membrane oxygenation and review of the literature. J Thorac Dis. 2019;11(8):3325–35.CrossRef Colman E, Yin EB, Laine G, Chatterjee S, Saatee S, Herlihy JP, et al. Evaluation of a heparin monitoring protocol for extracorporeal membrane oxygenation and review of the literature. J Thorac Dis. 2019;11(8):3325–35.CrossRef
98.
Zurück zum Zitat Vandiver JW, Vondracek TG. Antifactor Xa levels versus activated partial thromboplastin time for monitoring unfractionated heparin. Pharmacotherapy. 2012;32(6):546–58.CrossRef Vandiver JW, Vondracek TG. Antifactor Xa levels versus activated partial thromboplastin time for monitoring unfractionated heparin. Pharmacotherapy. 2012;32(6):546–58.CrossRef
99.
Zurück zum Zitat Horton S, Augustin S. Activated clotting time (ACT). Methods Mol Biol. 2013;992:155–67.CrossRef Horton S, Augustin S. Activated clotting time (ACT). Methods Mol Biol. 2013;992:155–67.CrossRef
100.
Zurück zum Zitat Finley A, Greenberg C. Heparin sensitivity and resistance: management during cardiopulmonary bypass. Anesth Analg. 2013;116(6):1210–22.CrossRef Finley A, Greenberg C. Heparin sensitivity and resistance: management during cardiopulmonary bypass. Anesth Analg. 2013;116(6):1210–22.CrossRef
101.
Zurück zum Zitat Baird CW, Zurakowski D, Robinson B, Gandhi S, Burdis-Koch L, Tamblyn J, et al. Anticoagulation and pediatric extracorporeal membrane oxygenation: impact of activated clotting time and heparin dose on survival. Ann Thorac Surg. 2007;83(3):912–9 discussion 9-20.CrossRef Baird CW, Zurakowski D, Robinson B, Gandhi S, Burdis-Koch L, Tamblyn J, et al. Anticoagulation and pediatric extracorporeal membrane oxygenation: impact of activated clotting time and heparin dose on survival. Ann Thorac Surg. 2007;83(3):912–9 discussion 9-20.CrossRef
102.
Zurück zum Zitat Maul TM, Nelson JS, Wearden PD. Paracorporeal lung devices: thinking outside the box. Front Pediatr. 2018;6:243.CrossRef Maul TM, Nelson JS, Wearden PD. Paracorporeal lung devices: thinking outside the box. Front Pediatr. 2018;6:243.CrossRef
103.
Zurück zum Zitat Maul TM, Wolff EL, Kuch BA, Rosendorff A, Morell VO, Wearden PD. Activated partial thromboplastin time is a better trending tool in pediatric extracorporeal membrane oxygenation. Pediatr Crit Care Med. 2012;13(6):e363–71.CrossRef Maul TM, Wolff EL, Kuch BA, Rosendorff A, Morell VO, Wearden PD. Activated partial thromboplastin time is a better trending tool in pediatric extracorporeal membrane oxygenation. Pediatr Crit Care Med. 2012;13(6):e363–71.CrossRef
104.
Zurück zum Zitat Atallah S, Liebl M, Fitousis K, Bostan F, Masud F. Evaluation of the activated clotting time and activated partial thromboplastin time for the monitoring of heparin in adult extracorporeal membrane oxygenation patients. Perfusion. 2014;29(5):456–61.CrossRef Atallah S, Liebl M, Fitousis K, Bostan F, Masud F. Evaluation of the activated clotting time and activated partial thromboplastin time for the monitoring of heparin in adult extracorporeal membrane oxygenation patients. Perfusion. 2014;29(5):456–61.CrossRef
105.
Zurück zum Zitat Khaja WA, Bilen O, Lukner RB, Edwards R, Teruya J. Evaluation of heparin assay for coagulation management in newborns undergoing ECMO. Am J Clin Pathol. 2010;134(6):950–4.CrossRef Khaja WA, Bilen O, Lukner RB, Edwards R, Teruya J. Evaluation of heparin assay for coagulation management in newborns undergoing ECMO. Am J Clin Pathol. 2010;134(6):950–4.CrossRef
106.
Zurück zum Zitat Despotis G, Summerfield A, Joist J, Goodnough L, Santoro S, Spitznagel E, et al. Comparison of activated coagulation time and whole blood heparin measurements with laboratory plasma anti-Xa heparin concentration in patients having cardiac operations. J Thorac Cardiovasc Surg. 1994;108(6):1076–82.CrossRef Despotis G, Summerfield A, Joist J, Goodnough L, Santoro S, Spitznagel E, et al. Comparison of activated coagulation time and whole blood heparin measurements with laboratory plasma anti-Xa heparin concentration in patients having cardiac operations. J Thorac Cardiovasc Surg. 1994;108(6):1076–82.CrossRef
107.
Zurück zum Zitat Colby CE, Sheehan A, Benitz W, Van Meurs K, Halamek LP, Moss RL. Maintaining adequate anticoagulation on extracorporeal membrane oxygenation therapy: Hemochron Junior Low Range versus Hemochron 400. J Extracorporeal Technol. 2003;35(1):35–8. Colby CE, Sheehan A, Benitz W, Van Meurs K, Halamek LP, Moss RL. Maintaining adequate anticoagulation on extracorporeal membrane oxygenation therapy: Hemochron Junior Low Range versus Hemochron 400. J Extracorporeal Technol. 2003;35(1):35–8.
108.
Zurück zum Zitat Dornia C, Philipp A, Bauer S, Stroszczynski C, Schreyer AG, Müller T, et al. D-dimers are a predictor of clot volume inside membrane oxygenators during extracorporeal membrane oxygenation. Artif Organs. 2015;39(9):782–7.CrossRef Dornia C, Philipp A, Bauer S, Stroszczynski C, Schreyer AG, Müller T, et al. D-dimers are a predictor of clot volume inside membrane oxygenators during extracorporeal membrane oxygenation. Artif Organs. 2015;39(9):782–7.CrossRef
109.
Zurück zum Zitat Lubnow M, Philipp A, Dornia C, Schroll S, Bein T, Creutzenberg M, et al. D-dimers as an early marker for oxygenator exchange in extracorporeal membrane oxygenation. J Crit Care. 2014;29(3):473.e1-5. Lubnow M, Philipp A, Dornia C, Schroll S, Bein T, Creutzenberg M, et al. D-dimers as an early marker for oxygenator exchange in extracorporeal membrane oxygenation.  J Crit Care. 2014;29(3):473.e1-5.
110.
Zurück zum Zitat Ignjatovic V. Prothrombin time/international normalized ratio. Methods Mol Biol. 2013;992:121–9.CrossRef Ignjatovic V. Prothrombin time/international normalized ratio. Methods Mol Biol. 2013;992:121–9.CrossRef
111.
Zurück zum Zitat Shahangian S, LaBeau KM, Howerton DA. Prothrombin time testing practices: adherence to guidelines and standards. Clin Chem. 2006;52(5):793–4.CrossRef Shahangian S, LaBeau KM, Howerton DA. Prothrombin time testing practices: adherence to guidelines and standards. Clin Chem. 2006;52(5):793–4.CrossRef
112.
Zurück zum Zitat Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Oral anticoagulant therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: american college of chest physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 Suppl):e44S-e88S.CrossRef Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti G. Oral anticoagulant therapy: antithrombotic therapy and prevention of thrombosis, 9th ed: american college of chest physicians evidence-based clinical practice guidelines. Chest. 2012;141(2 Suppl):e44S-e88S.CrossRef
113.
Zurück zum Zitat Warkentin TE, Greinacher A, Craven S, Dewar L, Sheppard JA, Ofosu FA. Differences in the clinically effective molar concentrations of four direct thrombin inhibitors explain their variable prothrombin time prolongation. Thromb Haemost. 2005;94(5):958–64. Warkentin TE, Greinacher A, Craven S, Dewar L, Sheppard JA, Ofosu FA. Differences in the clinically effective molar concentrations of four direct thrombin inhibitors explain their variable prothrombin time prolongation. Thromb Haemost. 2005;94(5):958–64.
114.
Zurück zum Zitat Steinlechner B, Kargl G, Schlömmer C, Holaubek C, Scheriau G, Eichinger S, et al. Can heparin-coated ECMO cannulas induce thrombocytopenia in COVID-19 patients? Case Reports Immunol. 2021;2021:6624682.CrossRef Steinlechner B, Kargl G, Schlömmer C, Holaubek C, Scheriau G, Eichinger S, et al. Can heparin-coated ECMO cannulas induce thrombocytopenia in COVID-19 patients? Case Reports Immunol. 2021;2021:6624682.CrossRef
115.
Zurück zum Zitat Tardy-Poncet B, Nguyen P, Thiranos J-C, Morange P-E, Biron-Andréani C, Gruel Y, et al. Argatroban in the management of heparin-induced thrombocytopenia: a multicenter clinical trial. Crit Care. 2015;19(1):396.CrossRef Tardy-Poncet B, Nguyen P, Thiranos J-C, Morange P-E, Biron-Andréani C, Gruel Y, et al. Argatroban in the management of heparin-induced thrombocytopenia: a multicenter clinical trial. Crit Care. 2015;19(1):396.CrossRef
116.
Zurück zum Zitat Alouidor B, Sweeney RE, Tat T, Wong RK, Yoon JY. Microfluidic point-of-care ecarin-based clotting and chromogenic assays for monitoring direct thrombin inhibitors. J Extra Corpor Technol. 2019;51(1):29–37. Alouidor B, Sweeney RE, Tat T, Wong RK, Yoon JY. Microfluidic point-of-care ecarin-based clotting and chromogenic assays for monitoring direct thrombin inhibitors. J Extra Corpor Technol. 2019;51(1):29–37.
117.
Zurück zum Zitat Hans GA, Besser MW. The place of viscoelastic testing in clinical practice. Br J Haematol. 2016;173(1):37–48.CrossRef Hans GA, Besser MW. The place of viscoelastic testing in clinical practice. Br J Haematol. 2016;173(1):37–48.CrossRef
118.
Zurück zum Zitat Nair P, Hoechter DJ, Buscher H, Venkatesh K, Whittam S, Joseph J, et al. Prospective observational study of hemostatic alterations during adult extracorporeal membrane oxygenation (ECMO) using point-of-care thromboelastometry and platelet aggregometry. J Cardiothorac Vasc Anesth. 2015;29(2):288–96.CrossRef Nair P, Hoechter DJ, Buscher H, Venkatesh K, Whittam S, Joseph J, et al. Prospective observational study of hemostatic alterations during adult extracorporeal membrane oxygenation (ECMO) using point-of-care thromboelastometry and platelet aggregometry. J Cardiothorac Vasc Anesth. 2015;29(2):288–96.CrossRef
119.
Zurück zum Zitat Heubner L, Greiner M, Vicent O, Beyer-Westendorf J, Tiebel O, Scholz U, et al. Predictive ability of viscoelastic testing using ClotPro® for short-term outcome in patients with severe Covid-19 ARDS with or without ECMO therapy: a retrospective study. Thromb J. 2022;20(1):48.CrossRef Heubner L, Greiner M, Vicent O, Beyer-Westendorf J, Tiebel O, Scholz U, et al. Predictive ability of viscoelastic testing using ClotPro® for short-term outcome in patients with severe Covid-19 ARDS with or without ECMO therapy: a retrospective study. Thromb J. 2022;20(1):48.CrossRef
120.
Zurück zum Zitat Whiting P, Al M, Westwood M, Ramos IC, Ryder S, Armstrong N, et al. Viscoelastic point-of-care testing to assist with the diagnosis, management and monitoring of haemostasis: a systematic review and cost-effectiveness analysis. Health Technol Assess. 2015;19(58):1–228, v−vi.CrossRef Whiting P, Al M, Westwood M, Ramos IC, Ryder S, Armstrong N, et al. Viscoelastic point-of-care testing to assist with the diagnosis, management and monitoring of haemostasis: a systematic review and cost-effectiveness analysis. Health Technol Assess. 2015;19(58):1–228, v−vi.CrossRef
121.
Zurück zum Zitat Wagner ML, Johnston M, Jenkins T, Palumbo JS, Rymeski BA. Use of thromboelastography in children on extracorporeal membrane oxygenation. J Pediatr Surg. 2022;57(6):1056–61.CrossRef Wagner ML, Johnston M, Jenkins T, Palumbo JS, Rymeski BA. Use of thromboelastography in children on extracorporeal membrane oxygenation. J Pediatr Surg. 2022;57(6):1056–61.CrossRef
122.
Zurück zum Zitat Herrmann J, Notz Q, Schlesinger T, Stumpner J, Kredel M, Sitter M, et al. Point of care diagnostic of hypercoagulability and platelet function in COVID-19 induced acute respiratory distress syndrome: a retrospective observational study. Thromb J. 2021;19(1):39.CrossRef Herrmann J, Notz Q, Schlesinger T, Stumpner J, Kredel M, Sitter M, et al. Point of care diagnostic of hypercoagulability and platelet function in COVID-19 induced acute respiratory distress syndrome: a retrospective observational study. Thromb J. 2021;19(1):39.CrossRef
123.
Zurück zum Zitat Zaky A. Thromboelastometry versus rotational thromboelastography in cardiac surgery. Semin Cardiothorac Vasc Anesth. 2017;21(3):206–11.CrossRef Zaky A. Thromboelastometry versus rotational thromboelastography in cardiac surgery. Semin Cardiothorac Vasc Anesth. 2017;21(3):206–11.CrossRef
124.
Zurück zum Zitat Neethling C, Calligaro G, Miller M, Opie JJS. The evolution of clot strength in critically-ill COVID-19 patients: a prospective observational thromboelastography study. Thromb J. 2021;19(1):83.CrossRef Neethling C, Calligaro G, Miller M, Opie JJS. The evolution of clot strength in critically-ill COVID-19 patients: a prospective observational thromboelastography study. Thromb J. 2021;19(1):83.CrossRef
125.
Zurück zum Zitat Schmidt DE, Majeed A, Bruzelius M, Odeberg J, Holmström M, Ågren A. A prospective diagnostic accuracy study evaluating rotational thromboelastometry and thromboelastography in 100 patients with von Willebrand disease. Haemophilia. 2017;23(2):309–18.CrossRef Schmidt DE, Majeed A, Bruzelius M, Odeberg J, Holmström M, Ågren A. A prospective diagnostic accuracy study evaluating rotational thromboelastometry and thromboelastography in 100 patients with von Willebrand disease. Haemophilia. 2017;23(2):309–18.CrossRef
126.
Zurück zum Zitat Bembea MM, Annich G, Rycus P, Oldenburg G, Berkowitz I, Pronovost P. Variability in anticoagulation management of patients on extracorporeal membrane oxygenation: an international survey. Pediatr Crit Care Med. 2013;14(2):e77-84.CrossRef Bembea MM, Annich G, Rycus P, Oldenburg G, Berkowitz I, Pronovost P. Variability in anticoagulation management of patients on extracorporeal membrane oxygenation: an international survey. Pediatr Crit Care Med. 2013;14(2):e77-84.CrossRef
127.
Zurück zum Zitat Fuchs G, Berg N, Eriksson A, Prahl WL. Detection of thrombosis in the extracorporeal membrane oxygenation circuit by infrasound: proof of concept. Artif Organs. 2017;41(6):573–9.CrossRef Fuchs G, Berg N, Eriksson A, Prahl WL. Detection of thrombosis in the extracorporeal membrane oxygenation circuit by infrasound: proof of concept. Artif Organs. 2017;41(6):573–9.CrossRef
128.
Zurück zum Zitat Hubbert L, Sundbom P, Loebe M, Peterzén B, Granfeldt H, Ahn H. Acoustic analysis of a mechanical circulatory support. Artif Organs. 2014;38(7):593–8.CrossRef Hubbert L, Sundbom P, Loebe M, Peterzén B, Granfeldt H, Ahn H. Acoustic analysis of a mechanical circulatory support. Artif Organs. 2014;38(7):593–8.CrossRef
129.
Zurück zum Zitat Morita N, Sakota D, Oota-Ishigaki A, Kosaka R, Maruyama O, Nishida M, et al. Real-time, non-invasive thrombus detection in an extracorporeal circuit using micro-optical thrombus sensors. Int J Artif Organs. 2021;44(8):565–73.CrossRef Morita N, Sakota D, Oota-Ishigaki A, Kosaka R, Maruyama O, Nishida M, et al. Real-time, non-invasive thrombus detection in an extracorporeal circuit using micro-optical thrombus sensors. Int J Artif Organs. 2021;44(8):565–73.CrossRef
130.
Zurück zum Zitat Fujiwara T, Sakota D, Ohuchi K, Endo S, Tahara T, Murashige T, et al. Optical dynamic analysis of thrombus inside a centrifugal blood pump during extracorporeal mechanical circulatory support in a porcine model. Artif Organs. 2017;41(10):893–903.CrossRef Fujiwara T, Sakota D, Ohuchi K, Endo S, Tahara T, Murashige T, et al. Optical dynamic analysis of thrombus inside a centrifugal blood pump during extracorporeal mechanical circulatory support in a porcine model. Artif Organs. 2017;41(10):893–903.CrossRef
131.
Zurück zum Zitat Meyer AD, Rishmawi AR, Kamucheka R, Lafleur C, Batchinsky AI, Mackman N, et al. Effect of blood flow on platelets, leukocytes, and extracellular vesicles in thrombosis of simulated neonatal extracorporeal circulation. J Thromb Haemost. 2020;18(2):399–410.CrossRef Meyer AD, Rishmawi AR, Kamucheka R, Lafleur C, Batchinsky AI, Mackman N, et al. Effect of blood flow on platelets, leukocytes, and extracellular vesicles in thrombosis of simulated neonatal extracorporeal circulation. J Thromb Haemost. 2020;18(2):399–410.CrossRef
132.
Zurück zum Zitat Sun W, Wang S, Chen Z, Zhang J, Li T, Arias K, et al. Impact of high mechanical shear stress and oxygenator membrane surface on blood damage relevant to thrombosis and bleeding in a pediatric ECMO circuit. Artif Organs. 2020;44(7):717–26.CrossRef Sun W, Wang S, Chen Z, Zhang J, Li T, Arias K, et al. Impact of high mechanical shear stress and oxygenator membrane surface on blood damage relevant to thrombosis and bleeding in a pediatric ECMO circuit. Artif Organs. 2020;44(7):717–26.CrossRef
133.
Zurück zum Zitat Huu DN, Kikuchi D, Maruyama O, Sapkota A, Takei M. Cole-cole analysis of thrombus formation in an extracorporeal blood flow circulation using electrical measurement. Flow Meas Instrum. 2017;53:172–9.CrossRef Huu DN, Kikuchi D, Maruyama O, Sapkota A, Takei M. Cole-cole analysis of thrombus formation in an extracorporeal blood flow circulation using electrical measurement. Flow Meas Instrum. 2017;53:172–9.CrossRef
134.
Zurück zum Zitat Li J, Sapkota A, Kikuchi D, Sakota D, Maruyama O, Takei M. Red blood cells aggregability measurement of coagulating blood in extracorporeal circulation system with multiple-frequency electrical impedance spectroscopy. Biosens Bioelectron. 2018;112:79–85.CrossRef Li J, Sapkota A, Kikuchi D, Sakota D, Maruyama O, Takei M. Red blood cells aggregability measurement of coagulating blood in extracorporeal circulation system with multiple-frequency electrical impedance spectroscopy. Biosens Bioelectron. 2018;112:79–85.CrossRef
135.
Zurück zum Zitat Yau JW, Stafford AR, Liao P, Fredenburgh JC, Roberts R, Brash JL, et al. Corn trypsin inhibitor coating attenuates the prothrombotic properties of catheters in vitro and in vivo. Acta Biomater. 2012;8(11):4092–100.CrossRef Yau JW, Stafford AR, Liao P, Fredenburgh JC, Roberts R, Brash JL, et al. Corn trypsin inhibitor coating attenuates the prothrombotic properties of catheters in vitro and in vivo. Acta Biomater. 2012;8(11):4092–100.CrossRef
136.
Zurück zum Zitat Larsson M, Rayzman V, Nolte MW, Nickel KF, Björkqvist J, Jämsä A, et al. A factor XIIa inhibitory antibody provides thromboprotection in extracorporeal circulation without increasing bleeding risk. Sci Transl Med. 2014;6(222):222ra17.CrossRef Larsson M, Rayzman V, Nolte MW, Nickel KF, Björkqvist J, Jämsä A, et al. A factor XIIa inhibitory antibody provides thromboprotection in extracorporeal circulation without increasing bleeding risk. Sci Transl Med. 2014;6(222):222ra17.CrossRef
137.
Zurück zum Zitat Weitz JI, Fredenburgh JC. Factors XI and XII as targets for new anticoagulants. Front Med (Lausanne). 2017;4:19. Weitz JI, Fredenburgh JC. Factors XI and XII as targets for new anticoagulants. Front Med (Lausanne). 2017;4:19.
138.
Zurück zum Zitat Wood KL, Ayers B, Gosev I, Kumar N, Melvin AL, Barrus B, et al. Venoarterial-extracorporeal membrane oxygenation without routine systemic anticoagulation decreases adverse events. Ann Thorac Surg. 2020;109(5):1458–66.CrossRef Wood KL, Ayers B, Gosev I, Kumar N, Melvin AL, Barrus B, et al. Venoarterial-extracorporeal membrane oxygenation without routine systemic anticoagulation decreases adverse events. Ann Thorac Surg. 2020;109(5):1458–66.CrossRef
139.
Zurück zum Zitat Raman J, Alimohamed M, Dobrilovic N, Lateef O, Aziz S. A comparison of low and standard anti-coagulation regimens in extracorporeal membrane oxygenation. J Heart Lung Transplant. 2019;38(4):433–9.CrossRef Raman J, Alimohamed M, Dobrilovic N, Lateef O, Aziz S. A comparison of low and standard anti-coagulation regimens in extracorporeal membrane oxygenation. J Heart Lung Transplant. 2019;38(4):433–9.CrossRef
140.
Zurück zum Zitat Carter KT, Kutcher ME, Shake JG, Panos AL, Cochran RP, Creswell LL, et al. Heparin-sparing anticoagulation strategies are viable options for patients on veno-venous ECMO. J Surg Res. 2019;243:399–409.CrossRef Carter KT, Kutcher ME, Shake JG, Panos AL, Cochran RP, Creswell LL, et al. Heparin-sparing anticoagulation strategies are viable options for patients on veno-venous ECMO. J Surg Res. 2019;243:399–409.CrossRef
141.
Zurück zum Zitat Molloy CP, Yao Y, Kammoun H, Bonnard T, Hoefer T, Alt K, et al. Shear-sensitive nanocapsule drug release for site-specific inhibition of occlusive thrombus formation. J Thromb Haemost. 2017;15(5):972–82.CrossRef Molloy CP, Yao Y, Kammoun H, Bonnard T, Hoefer T, Alt K, et al. Shear-sensitive nanocapsule drug release for site-specific inhibition of occlusive thrombus formation. J Thromb Haemost. 2017;15(5):972–82.CrossRef
142.
Zurück zum Zitat Hoefer T, Rana A, Niego B, Jagdale S, Albers HJ, Gardiner EE, et al. Targeting shear gradient activated von Willebrand factor by the novel single-chain antibody A1 reduces occlusive thrombus formation in vitro. Haematologica. 2021;106(11):2874–84.CrossRef Hoefer T, Rana A, Niego B, Jagdale S, Albers HJ, Gardiner EE, et al. Targeting shear gradient activated von Willebrand factor by the novel single-chain antibody A1 reduces occlusive thrombus formation in vitro. Haematologica. 2021;106(11):2874–84.CrossRef
143.
Zurück zum Zitat Zhang M, Pauls JP, Bartnikowski N, Haymet AB, Chan CH, Suen JY, et al. Anti-thrombogenic surface coatings for extracorporeal membrane oxygenation: a narrative review. ACS Biomater Sci Eng. 2021;7(9):4402–19.CrossRef Zhang M, Pauls JP, Bartnikowski N, Haymet AB, Chan CH, Suen JY, et al. Anti-thrombogenic surface coatings for extracorporeal membrane oxygenation: a narrative review. ACS Biomater Sci Eng. 2021;7(9):4402–19.CrossRef
144.
Zurück zum Zitat Ebner M, Birschmann I, Peter A, Spencer C, Härtig F, Kuhn J, et al. Point-of-care testing for emergency assessment of coagulation in patients treated with direct oral anticoagulants. Crit Care. 2017;21(1):32.CrossRef Ebner M, Birschmann I, Peter A, Spencer C, Härtig F, Kuhn J, et al. Point-of-care testing for emergency assessment of coagulation in patients treated with direct oral anticoagulants. Crit Care. 2017;21(1):32.CrossRef
145.
Zurück zum Zitat Galien S, Hultström M, Lipcsey M, Stattin K, Frithiof R, Rosén J, et al. Point of care ultrasound screening for deep vein thrombosis in critically ill COVID-19 patients, an observational study. Thromb J. 2021;19(1):38.CrossRef Galien S, Hultström M, Lipcsey M, Stattin K, Frithiof R, Rosén J, et al. Point of care ultrasound screening for deep vein thrombosis in critically ill COVID-19 patients, an observational study. Thromb J. 2021;19(1):38.CrossRef
146.
Zurück zum Zitat Wool GD. Benefits and pitfalls of point-of-care coagulation testing for anticoagulation management: an ACLPS critical review. Am J Clin Pathol. 2019;151(1):1–17.CrossRef Wool GD. Benefits and pitfalls of point-of-care coagulation testing for anticoagulation management: an ACLPS critical review. Am J Clin Pathol. 2019;151(1):1–17.CrossRef
147.
Zurück zum Zitat Li D, Liu X, Chai Y, Shan J, Xie Y, Liang Y, et al. Point-of-care blood coagulation assay enabled by printed circuit board-based digital microfluidics. Lab on a Chip. 2022;22(4):709–16. Li D, Liu X, Chai Y, Shan J, Xie Y, Liang Y, et al. Point-of-care blood coagulation assay enabled by printed circuit board-based digital microfluidics. Lab on a Chip. 2022;22(4):709–16.
Metadaten
Titel
Current and future strategies to monitor and manage coagulation in ECMO patients
verfasst von
Saeedreza Zeibi Shirejini
Josie Carberry
Zoe K. McQuilten
Aidan J. C. Burrell
Shaun D. Gregory
Christoph E. Hagemeyer
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
Thrombosis Journal / Ausgabe 1/2023
Elektronische ISSN: 1477-9560
DOI
https://doi.org/10.1186/s12959-023-00452-z

Weitere Artikel der Ausgabe 1/2023

Thrombosis Journal 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Bei Herzinsuffizienz muss „Eisenmangel“ neu definiert werden!

16.05.2024 Herzinsuffizienz Nachrichten

Bei chronischer Herzinsuffizienz macht es einem internationalen Expertenteam zufolge wenig Sinn, die Diagnose „Eisenmangel“ am Serumferritin festzumachen. Das Team schlägt vor, sich lieber an die Transferrinsättigung zu halten.

Herzinfarkt mit 85 – trotzdem noch intensive Lipidsenkung?

16.05.2024 Hypercholesterinämie Nachrichten

Profitieren nach einem akuten Myokardinfarkt auch Betroffene über 80 Jahre noch von einer intensiven Lipidsenkung zur Sekundärprävention? Um diese Frage zu beantworten, wurden jetzt Registerdaten aus Frankreich ausgewertet.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Erstmanifestation eines Diabetes-Typ-1 bei Kindern: Ein Notfall!

16.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Manifestiert sich ein Typ-1-Diabetes bei Kindern, ist das ein Notfall – ebenso wie eine diabetische Ketoazidose. Die Grundsäulen der Therapie bestehen aus Rehydratation, Insulin und Kaliumgabe. Insulin ist das Medikament der Wahl zur Behandlung der Ketoazidose.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.