Next Article in Journal
Contemporary Methods for Detection and Intervention of Distal Medium and Small Vessel Occlusions
Previous Article in Journal
Relationship between Prostate Inflammation and Periodontal Disease—A Systematic Review and Meta-Analysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Update on Anticoagulation Strategies in Patients with ECMO—A Narrative Review

1
Department of Anaesthesiology and Intensive Care Medicine, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, 12808 Prague, Czech Republic
2
Emergency Service of Central Bohemia, Vančurova 1544, 27201 Kladno, Czech Republic
3
Department of Anaesthesia, Golden Jubilee University National Hospital, Clydebank G81 4DY, UK
4
Department of Anaesthesiology and Intensive Care Medicine, Institute for Clinical and Experimental Medicine, 14021 Prague, Czech Republic
5
Department of Anaesthesia, Antrim Area Hospital, Antrim BT41 2RL, UK
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2023, 12(18), 6067; https://doi.org/10.3390/jcm12186067
Submission received: 8 August 2023 / Revised: 14 September 2023 / Accepted: 18 September 2023 / Published: 20 September 2023
(This article belongs to the Section Intensive Care)

Abstract

:
The use of extracorporeal membrane oxygenation (ECMO) has recently increased exponentially. ECMO has become the preferred mode of organ support in refractory respiratory or circulatory failure. The fragile balance of haemostasis physiology is massively altered by the patient’s critical condition and specifically the aetiology of the underlying disease. Furthermore, an application of ECMO conveys another disturbance of haemostasis due to blood-circuit interaction and the presence of an oxygenator. The purpose of this review is to summarise current knowledge on the anticoagulation management in patients undergoing ECMO therapy. The unfractionated heparin modality with monitoring of activated partial thromboplastin tests is considered to be a gold standard for anticoagulation in this specific subgroup of intensive care patients. However, alternative modalities with other agents are comprehensively discussed. Furthermore, other ways of monitoring can represent the actual state of coagulation in a more complex fashion, such as thromboelastometric/graphic methods, and might become more frequent. In conclusion, the coagulation system of patients with ECMO is altered by multiple variables, and there is a significant lack of evidence in this area. Therefore, a highly individualised approach is the best solution today.

1. Introduction

The use of extracorporeal membrane oxygenation (ECMO) has increased exponentially in recent times [1], particularly following the H1N1 and COVID-19 pandemics [2]. It represents a modified cardiopulmonary bypass system that facilitates extracorporeal decarboxylation and oxygenation. The application of ECMO encompasses various scenarios involving refractory both heart and lung failure, and these indications have been established for adult and paediatric patients over recent decades, bringing a chance to improve the outcome of these patients [3]. The essential components of the ECMO circuit include a pump, a membrane oxygenator, a heat exchanger, a venous cannula, an arterial or venous infusion cannula, tubing, and connectors. It can be configured either in a venovenous (VV) mode, which is employed for the management of severe respiratory failure, or in a venoarterial (VA) mode, which provides simultaneous support for both respiratory and cardiac functions [4]. VV ECMO can be initiated using peripheral approaches involving two cannulae (femorojugular or femorofemoral) or it can be established using a double-lumen cannula inserted under ultrasound and echocardiographic guidance through the internal jugular vein. Alternatively, the drainage cannula is introduced through a venous access point in VA ECMO, while the return cannula is placed into an arterial access site. VA ECMO classification can be categorised as either central or peripheral, depending on the specific vessels used for cannulation [5]. In the central configuration, the drainage cannula can be directly inserted into the right atrium, with the return cannula placed in the ascending segment of the aorta. In the peripheral configuration, blood can be extracted through the femoral or jugular veins, and it is then returned to the patient through the carotid, axillary, or femoral arteries [5].
As anticipated, ECMO complications are prevalent and linked to a substantial rise in morbidity and mortality [3]. The risks associated with the ECMO procedure can be categorised into two main groups: mechanical and patient-related medical complications. Mechanical complications are represented by issues such as oxygenator or pump failure, heat exchanger malfunction, cannula-related problems, and disruptions in the circuit. On the other hand, patient-related medical complications include bleeding, thromboembolic events, neurological issues, the development of additional organ failures (cardiovascular, renal, or hepatic), metabolic disorders, and infections [3,5]. Thus, it is important to note that despite recent advancements, the mortality rates associated with ECMO use remain high for both modalities [6,7].

2. ECMO Circuit Interactions with Coagulation System

A fragile balance of haemostasis physiology is massively altered in a critical patient, and specifically by the aetiology of the primary disease. Furthermore, the use of ECMO brings another disturbance to haemostasis related to blood interaction with the circuit and an oxygenator [8]. In the extracorporeal circuit, blood is exposed to a non-biologic surface. This inevitably leads to up-regulation of pro-inflammatory and procoagulant pathways. The pathophysiological background of this process is based on the activation of coagulation factor XII and platelet activation with their subsequent dysfunction [9]. These are the reasons why, in patients with ECMO, it is necessary to balance between possible thrombotic and bleeding complications, which are unfortunately still very frequent. According to the ELSO register, thrombotic complications are more common than bleeding, but bleeding carries a significantly higher mortality risk [10]. Furthermore, under specific conditions, for example, severe bleeding or emergency surgery, anticoagulation is usually temporarily suspended. Surprisingly, a recent systematic review analysing anticoagulant-free ECMO support found no increased risk of thrombotic or bleeding complications compared to systemic anticoagulation [8].
Although the updated ELSO guidelines (2021) were published recently, the rapid development in the field and a lack of evidence-based recommendations, as conceded in the guidelines [11], justify a need for a review. According to the current literature, several pharmacological agents can be used as anticoagulants in patients with ECMO. These are: unfractionated heparin (UFH), low-molecular-weight heparin (LMWH), fondaparinux, and direct thrombin inhibitors [11,12]. Nevertheless, more than choosing the right agent, another challenge is finding and deciding on the appropriate target and the most suitable testing method. First, standard coagulation tests (prothrombin time/INR and aPTT) are often altered by the patient’s critical condition (especially shock, sepsis and liver dysfunction). Thus, coagulation times might be spontaneously prolonged. At the same time, testing the patient who shows prolonged coagulation times with viscoelastometric tests (VETs) could demonstrate a hypercoagulation state. Another major issue in patients with ECMO is the occurrence of low platelet count, making the topic even more complex.
In this narrative review, we summarise the knowledge on choosing an anticoagulant agent with an emphasis on the most commonly used UFH. Another aspect is the appropriate method of testing, and the last highlighted issue is the gentle balance between thrombotic and bleeding complications.

3. Considerations for VV versus VA ECMO

The anticoagulation strategy may differ between the VV and VA modalities. The VV ECMO may tend to have more bleeding complications at the insertion [10], likely related to a rapid decrease in pCO2 together with a bolus anticoagulation with cannulation [13]. The VA ECMO may tend more towards thrombosis-related complications, especially with poor LV unloading and intracardiac thrombus formation, and during weaning of the VA modality due to low blood flow (down to 0.5 L/min) in the extracorporeal circuit and oxygenator [10]. Both modalities are fraught with cannulation-related deep venous thrombosis, i.e., related to the drainage cannulas and the return cannula of the VV ECMO [14]. The low-flow segment of the peripheral VA ECMO is between the return arterial cannula and the prograde superficial femoral artery cannula. Here, the arterial thrombus formation indicates a surgical approach with femoral thrombectomy, rather than simple bedside extraction with compression or a closure device.

4. Considerations for Exhaustion of the Coagulation System

Exhaustion of the coagulation cascade typically results from a procoagulant state. A systemic inflammatory syndrome involves major triggers and regulators of the coagulation system. These are damaged endothelium by inflammation, activated intrinsic system with alteration in the protein C pathway, activated platelets, thromboplastin and activation of the extrinsic system, and finally involving the changes in fibrinolysis [15]. A typical example of coagulation activation is a sepsis-related disseminated inflammatory coagulopathy. Here, the mild prolongation of classic coagulation monitors like APTT, PT are accompanied by an increase in acute-phase proteins, including fibrinogen, and a rise in fibrin degradation products (D-dimers) with a D-dimer-related prolongation of the thrombin time [15,16,17]. An activation of platelets may be seen in the aggregometry tests or in the VETs. Specific tests may even show increased activity of certain factors like vWf. With progression of the systemic inflammation and exhaustion of the coagulation cascade, the thrombin activity decreases, accompanied by further extension of the coagulation times (APTT, PT, TT), decreasing fibrinogen, vWf, antithrombin III, and platelet count. The fibrinolytic pathway may require testing with the VETs to reveal hyper- or hypoactivity of plasmin, with potential implications for therapy [18].

5. Special Considerations for COVID-19 and ECMO

Since the outbreak of SARS-CoV-2, an increased risk of thrombosis has been reported in patients with COVID-19 [19]. COVID-19-associated coagulopathy (CAC) manifests as micro- or macrothrombi formation, which can cause damage to multiple organs (lungs, heart, brain, kidneys) [19], resulting in an increase in morbidity and mortality. Compared to other viral infections, patients with COVID-19 have higher rates and severity of clotting events, which is underlined by laboratory findings of elevated plasma levels of D-dimer, C-reactive protein, fibrinogen and P-selectin [19]. Pathophysiological background includes a dysregulation among the inflammatory, immune, coagulation, fibrinolytic, complement and kallikrein–kinin systems. Thus, the term “immune thrombosis” has been established [19].
VV ECMO has been extensively used in the treatment of COVID-19-related ARDS. Less frequently, ECMO in VA configuration has been indicated for combined cardiac-respiratory failure [20]. Multiple studies showed that COVID-19 is connected to an increased risk of endothelial inflammation-mediated thrombosis [21,22]. As a result, the therapeutic anticoagulation instead of a prophylactic modality has been accepted for critically ill COVID-19 patients [16]. However, this approach showed serious limitations for patients requiring ECMO support. Mansour et al. published a nationwide trial involving 620 patients with COVID-19 and ECMO support. The study demonstrated a high prevalence of all bleeding events (49.4%) regardless of their severity.
A different methodology including only life-threatening bleeds, like in the ELSO registry or the International Society on Thrombosis and Haemostasis registry, may not elucidate the difference against the control population, because the bleeding events were independently associated with a higher in-hospital mortality on day 90. In addition, the rate of thrombotic complications was also high (36%), but thrombotic events were not associated with higher mortality rates [21]. These results differ from the large multicenter studies in the population of ECMO patients without COVID-19, which showed a significant increase in mortality due to thrombosis. It is essential to highlight that in the Mansour et al. study, intracranial bleeding (ICH) was reported in an alarming 8% of patients [19]. This finding supports multiple recent publications [20], suggesting a higher risk of intracranial bleeding in COVID-19 patients treated with ECMO [22]. Furthermore, the systematic review and meta-analysis of Jin et al. [23] included 23 studies with 6878 subjects. The results showed that a major bleeding event occurred in 37.4% of cases. ICH was reported in 9.9% of the subjects. The increased risk of ICH was shown in COVID-19 ECMO patients compared to the population without COVID-19 (RR = 2.23 (95% CI: 1.32–3.75)). Unrestricted anticoagulation regimens recommended early in the COVID-19 pandemic [24] may explain the increased incidence of significant bleeding events in patients with ECMO. The key to understanding the change from a hypercoagulable status of severe COVID-19 viral sepsis to a hypocoagulating frequently bleeding COVID-19 patient on ECMO is the extensive endothelial inflammation caused by the coronavirus [15]. COVID-19-caused inflammation of the endothelium initially upregulates the activity of von Willebrand factor (vWf) and factor VIII, leading to inadequate balance by the otherwise often normal levels of its depolymerase (ADAMTS-13). The prothrombotic state associates with high thrombin activity due to contact activation of the coagulation cascade and high levels of the acute-phase inflammatory protein fibrinogen [25]. With an insertion of the ECMO circuit, the situation changes. Acquired vWf disease accompanies almost uniformly current ECMO circuits and is even more relevant to severe COVID-19 patients on ECMO. With the commencement of the ECMO therapy, the upregulated vWf decreases [26]. This, together with an exhaustion of the coagulation system and low thrombin activity and a decrease in the initially upregulated fibrinogen levels, may create a dangerous cumulation of factors leading to life-threatening bleeding [17,27]. Moreover, the alterations in the fibrinolytic system are difficult to predict and should be assessed individually [18]. The endothelial inflammation of the haematoencephalic barrier may potentially relate to the rates of ICH [28]. The potential role of rapid change in pCO2 during ECMO initiation on the incidence of ICH has already been discussed as a potential causative factor for an ICH.
Currently, there is no consensus on an intensified anticoagulation practice for this particular patient population or those on only prophylactic anticoagulation, because the reported numbers of serious thrombotic events also related to ECMO are not negligible [29]. The increased prevalence of ICH highlights the need for a strictly controlled and individualised anticoagulation modality.

6. Unfractionated Heparin as Gold Standard

6.1. Introduction to Heparin

In contrast to most pharmacologic agents used in medicine, heparin is a heterogeneous compound consisting of molecules with variable molecular weights. The molecular weight of majority of the chains varies from 15,000 to 19,000 Daltons. This heterogeneity of heparin also leads to variable biologic effects. Therefore, the dose is usually not given in milligrams but in standardised international units—IU [30,31].
Heparin is used extensively because its effect can be easily titrated, the half-life is short, and its antidote (protamine) is widely accessible [32]. At its effect site, a bond with antithrombin (AT; previously antithrombin III) is created by the pentasaccharide segment of the molecule. The effect of AT is 1000 times multiplied when heparin is bound to its molecule. AT is a potent inhibitor of thrombin (factor II), factor Ixa, and Xa. Subsequently, a thrombin-dependent platelet activation and factors V, VIII, and XI are inhibited [30,33]. In conclusion, the mechanism of action of heparin is complex.
A clinician may face some typical situations when administering a heparin anticoagulation. First, an excessive dose of UFH may be necessary to maintain the desired target of anticoagulation. Second is the occurrence of unexpected and sometime severe low platelet count in the so-called heparin-induced thrombocytopenia (HIT), and lastly in deciding about the right method of monitoring and choosing the right target with a lack of evidence.

6.2. Monitoring and Target

Although UFH is the most frequently used anticoagulant in this specific group of patients, there are no detailed recommendations and a significant lack of data in scientific databases. According to the 2021 ELSO Adult and Paediatric Anticoagulation Guidelines [11], the monitoring of anticoagulation by ACT (in the range of 180–200 s [34]; with a low-concentration cartridge [35]) is considered a gold standard for the guidance of anticoagulation therapy in patients with ECMO. The widely spread standard laboratory-activated partial thromboplastin test (aPTT) is—owing to the guidelines—a gold standard assay for monitoring UFH. However, the laboratory aPTT assay may be influenced by multiple factors compared to the anti-Xa test, and recent data suggest purchasing both aPTT and the anti-Xa tests in parallel, especially in ECMO patients. This approach may allow for a calibration of the aPTT and prevent underdosing or overdosing with the unfractionated heparin modality [36]. Bedside testing for ACT and aPTT is possible, which might differ from standardised laboratory aPTT assays. There is evidence that decisions in anticoagulation management are different for a laboratory-obtained aPTT compared with bedside testing of ACT and aPTT. Furthermore, there is older evidence that standard aPTT from the laboratory showed a stronger correlation with plasma UFH concentration than the mentioned bedside assays [37]. According to current research, other authors prefer not to use ACT [34].
ELSO suggested repeating the test every 1–2 h in case of ACT and every 6–12 h for monitoring the anticoagulation effect with aPTT. Great emphasis is placed on individual protocols of particular ECMO centres, which depend on local experiences. In the case of resistance to heparin, an examination with anti-Xa might be helpful. Routine supplementation with AT does not have scientific evidence.
Under special circumstances, that is, spontaneously prolonged aPTT, VETs might give a reliable image of the actual coagulation status. Thus, hypercoagulability can be uncovered, and anticoagulation can be initiated regardless of spontaneously prolonged aPTT. In patients supported on ECMO, multiple studies evaluated the safety and feasibility of a VET-driven strategy to titrate heparin versus the “conventional” standard of care based on aPTT monitoring [34]. The improved trend of less bleeding complications was observed in a TEG®-guided group (target 16–24 min of the R parameter). Furthermore, required doses of heparin were lower in a TEG® group than aPTT-guided care [38]. Another study investigated optimal values for citrated kaolin TEG R® time. They concluded that TEG® R time > 17.85 min (sensitivity 84%, specificity 68%, PPV 82%, and NPV 59%) is able to minimise the risk of thrombosis in paediatric and neonatal ECMO patients [39]. The essential issue is that according to ELSO [11], there are no relevant data to guide current practice. A whole paradigm about targeting aPTT ratio between 1.5 and 2.5 is based on one prospective non-ECMO study from 1972. The trial concluded that keeping aPTT in this range reduces the prevalence of recurrent venous thromboembolism. Another historic study conducted on patients undergoing continuous renal replacement therapy from 1996 claims that an aPTT ratio of 1.5 can cut the risk of circuit thrombosis by half. On the other hand, the risk of bleeding complications is threefold increased [40]. This lack of crucial data explains why many recommendations exist for an individualised approach.
The most appropriate current approach is a highly individually titrated anticoagulation target. Multiple variables such as platelet count, fibrinogen level, initial results of INR, aPTT, VETs, evidence of prothrombotic/haemorrhagic condition, and clinical signs of thrombosis or bleeding should be considered before making the optimal decision.
The rationale behind this precise anticoagulation titration is to keep the fragile balance between the two extreme situations. While bleeding and thrombosis remain common complications in ECMO patients, haemorrhagic events substantially influence mortality (emphasising fatal intra-cranial bleeding) [10,41]. According to the systematic review of Abruzzo et al., early computed tomography (CT) diagnosis of deep venous thrombosis was obtained in a number of patients as high as 71.4%, and pulmonary embolism was observed in 16.2% of the patients [14]. Therefore, a routine check-up for thrombosis should be considered after ECMO decannulation. Ultrasound examination seems like an ideal non-invasive method without ionising radiation, but unfortunately has a higher rate of false-negative results compared to [14].
Furthermore, not only the target of anticoagulation is what can increase the risk of adverse bleeding. The extensive ELSO data show that rapid changes in PCO2 may also independently increase the risk of neurological complications (seizures, intracranial bleeding) [13]. That is why a gentle approach should be applied to restore blood gas homeostasis without rapid changes.

6.3. Heparin Resistance

This condition is defined as the need for an excessive dose of UFH to achieve the desired laboratory result. For some, this is defined as a dose >35,000 IU/24 h required to achieve a subtherapeutic range. One of the reasons for heparin resistance is considered to be AT deficiency [42]. Furthermore, other risk factors increase the risk of heparin resistance, including smoking, chronic obstructive pulmonary disease, liver dysfunction, nephropathies, hypoalbuminaemia, thrombocytosis, intra-aortic balloon pump, prolonged UFH treatment and presence of an ECMO circuit. AT deficiency might be acquired or (rarely) congenital [33,42,43]. According to some sources, a very high prevalence of heparin resistance was observed in COVID-19 patients (up to 62%) [44]. To demonstrate that this is not a simple one-way pathophysiological pathway, it is necessary to mention that low AT level does not automatically cause heparin resistance. On the other hand, a normal AT level might be present in cases of resistance to heparin. Exogenous AT might look like a simple solution, but it is not widely consensually recommended and might be a reason for adverse bleeding complications.
Because of the heterogeneous nature of UFH molecule size, molecules of lower molecular weight may be more prevalent in some drugs. Hence, in cases of apparent heparin resistance, anti-Xa monitoring might be more appropriate and explain inadequate response in aPTT [30]. In this case, following up with a targeted anti-Xa approach should be considered.

7. Low-Molecular-Weight Heparins and Fondaparinux

Low-molecular-weight heparins (LMWH) are produced by depolymerisation of UFH to approximately one-third of the original size of the molecule (4000–6500 D), thus creating a shorter saccharide chain. Similarly to UFH, there is considerable variability in molecular weight and chemical properties [45].
Fondaparinux is a fully synthetic pentasaccharide, and its units are structurally similar to the cleaved monomeric units of UFH [46]. It was developed during an effort to solve problems associated with naturally sourced UFH (e.g., variability in composition, potential contamination) and eliminate side effects such as HIT [47]. Although LMWH has also been associated with HIT, the risk is considerably lower than with UFH and negligible in the case of fondaparinux [48].
Similarly to UFH, LMWH acts by activating AT. However, due to its relatively short molecular chain, LMWH cannot form the tertiary complex with AT and thrombin, which is essential for the inactivation of thrombin. In contrast, factor Xa is inactivated by AT without the need to form the tertiary complex with thrombin. Thus, in the presence of LMWH, AT exhibits preferential binding to factor Xa [45,49]. Due to its chemical structure, fondaparinux targets the same pathway as LMWH, which means binding to AT with following deactivation of Xa [47].
Both LMWH and fondaparinux are eliminated by the kidneys, have a longer plasma half-life, better bioavailability, and in a standard setting, a more predictable dose response than UFH. However, they cannot be easily reversed by an antidote and their effect is prolonged in patients with renal failure [45,50]. Another disadvantage of standard subcutaneously administered LMWH is its variable bioavailability. This is significantly influenced by peripheral perfusion, often affected by the use of vasopressor medications [51,52]. Therefore, the LMWH administration should always be by the intravenous route.

7.1. Monitoring and Target

7.1.1. LMWH

The primary method of monitoring both LMWH and fondaparinux is by measuring the anti-Xa levels. The effect of fondaparinux is best measured using a specifically calibrated anti-Xa assay because the standard LMWH-calibrated assay overestimates the concentration of fondaparinux by about 20% [39]. Current recommendations for monitoring and anticoagulation targets in ECMO patients are outlined in the 2021 ELSO Adult and Pediatric anticoagulation guidelines. The guidelines suggest a target anti-Xa, albeit only in the context of UFH modality [11].
Concerning LMWH, published articles suggest that most centres opt for prophylactic dosage, especially in V-V ECMO [53,54]. This is in agreement with the current trend towards less or no anticoagulation for the V-V ECMO, supported mainly by retrospective studies and only one prospective pilot study [55]. Therefore, due to the lack of robust prospective data, the prophylactic approach is not included in the 2021 ELSO guidelines.
LMWHs have gradually replaced UFH in many medical indications, and their superiority in prophylactic anticoagulation in critically ill patients has been consistently described [56,57,58]. Additionally, LMWHs have been used in prophylaxis in other types of extracorporeal circuits, for example, continuous renal replacement therapy (CRRT) [59]. Some data originating from a non-intensive care environment even suggest their possible superiority in therapeutic anticoagulation [60,61]. Despite this, there is a paucity of evidence on its use in patients with ECMO. This is also reflected in the 2021 ELSO guidelines, where the emphasis on using UFH/direct thrombin inhibitors is prominent [62].
A single-centre observational study evaluated administering a prophylactic dose of enoxaparin to more than 60 patients with nonsurgical VV ECMO. It showed fewer bleeding events with a higher incidence of thrombotic complications than studies with therapeutic anticoagulation with UFH [53]. Regarding surgical patients, one single-centre retrospective study investigated prophylactic subcutaneous enoxaparin versus intravenous UFH used in perioperative ECMO in 102 lung transplant patients and showed no difference in bleeding risk and fewer thrombotic episodes in the LMWH group [63].
The use of VETs for monitoring of the “LMWH” effect have not been standardised yet, and thus it is not suitable for titration of anticoagulant effect in fragile ECMO patients [64].

7.1.2. Fondaparinux

Similarly to LMWH, the evidence for using fondaparinux in patients with ECMO is scarce, albeit the published articles generally describe positive outcomes. Only several case reports and series on the topic were published, all with one common theme: patients on ECMO who developed HIT. The prophylactic dose once daily appears to be this group’s most prevalent anticoagulation strategy [65,66,67]. In most published case reports, only a prophylactic dose of fondaparinux was used, and anti-Xa was not measured [65,66]. In one case series published by Rychlíčková et al. [67], the therapeutic levels were targeted, the anti-Xa levels were measured, and the dosages were adjusted accordingly. The prophylactic approach using fondaparinux could cause a problem because the low anticoagulation target might not be sufficient for patients with HIT, as suggested by the 2018 American Society of Hematology guideline for management of heparin-induced thrombocytopenia. This guideline recommends achieving therapeutic-intensity anticoagulation [62]. Although this target has not been established for patients with ECMO, it would seem reasonable that higher levels of anticoagulation should be targeted in patients with HIT. Management of anticoagulation with fondaparinux based on VETs is not appropriate. According to minimal data, VETs are only able to detect supratherapeutic doses of fondaparinux [64].
In summary, anticoagulation with fondaparinux and LMWH in patients with ECMO is challenging and guidelines for the optimal target have yet to be established. An individual approach to each case is essential and test results must be interpreted in the context of both the patient and the circuit.

7.2. Heparin-Induced Thrombocytopenia

A decrease in platelet count is a common complication of ECMO therapy (both in VV and VA) and occurs in up to 50% of subjects with ECMO. Post-cardiotomy ECMO patients represent a group with a higher risk of developing significant thrombocytopenia. There are many possibilities for the aetiology of thrombocytopenia in this particular group of patients: the foreign surface of the circuit, platelet activation, nonspecific activation of the inflammatory cascade, sepsis, medications (immunosuppressives, PDE III inhibitors), surgery, and bleeding. Often it is not possible to highlight one particular reason. Thus, aetiology is frequently considered multifactorial [68,69,70].
The prevalence of HIT in patients undergoing ECMO therapy ranges from 0.5% to 5.0% [70,71]. These numbers are probably underestimated, as HIT testing is not routinely performed in all patients. HIT is an antibody-mediated side effect of UFH administration that is characterised by the onset of thrombocytopenia, usually 5–10 days after the beginning of therapy. Paradoxically, the result of HIT is a prothrombotic state with a high risk of thrombosis, especially in cases of rare HIT II. When the thrombocytopenia is severe (<20 × 109/L), then a risk of bleeding arises [68,70,71].
Typically, PF4 is stored in α-platelet granules (and released after adequate activation). The physiological function of PF4 is in binding to heparan sulphate, an endogenous substance very similar to UFH. However, its ability to bind with UFH shows a much higher affinity. Due to the newly created complex heparin–PF4 with the Fc site of the antibodies, the platelets and immune cells are activated. This results in thrombin activation and scavenging of platelets by macrophages [71,72].
The correct HIT diagnostic algorithm should be based on three positive criteria [72]:
(1)
UFH (or LMWH) exposure.
(2)
At least one clinical or laboratory result (low platelet count or new venous/arterial thrombosis).
(3)
Evidence of specific HIT antibodies.
HIT-specific antibodies are examined by ELISA for the detection of the heparin–PF4 complex. This test shows a negative predictive value of 98–99%. This means that the negative result is very reliable for excluding the possibility of HIT. However, the positive predictive value is only 2–15%. Clinical suspicion and mentioned screening methods above predict the result of the confirmation methods well, so together they might often be sufficient. Confirmation tests with higher specificity are based on examination of platelet activation, e.g., SRA (serotonin-release assay) or HIPA (heparin-induced platelet activation assay) [73].
The treatment of HIT is based on discontinuing UFH administration and switching to another anticoagulation modality. Direct thrombin inhibitors, such as argatroban or bivalirudin are recommended [72]. The switch to fondaparinux is also possible and widely accepted as a drug of choice [73].
The risk of developing HIT when using LMWH is lower than UFH, but remains a genuine concern. The pathophysiological mechanisms of HIT may differ between UFH and LMWH, primarily due to the smaller size of LMWH molecules. The size of the heparin molecule plays a crucial role in its capacity to bind with PF4, as confirmed by experimental studies. This suggests that LMWH is less likely to provoke HIT when compared to UFH, as research indicates a higher occurrence of anti-H–PF4 antibody formation in patients treated with UFH compared to those receiving LMWH [48]. Nevertheless, it is essential to acknowledge that LMWH can still potentially induce HIT because specific LMWH preparations may contain larger molecules that can interact with PF4 [48].

8. Direct Thrombin Inhibitors

The direct thrombin inhibitors (DTIs) represent short-acting anticoagulants with a mechanism of action that primarily inhibits thrombin, a key enzyme involved in the coagulation cascade. By targeting both soluble and fibrin-bound thrombin, they prevent the formation of clots. They inhibit fibrinogen to fibrin, thereby limiting clot formation within the ECMO circuit without the need for binding to a cofactor like AT [74]. Only intravenous DTIs, including bivalirudin and argatroban, have been used successfully for anticoagulation in patients with ECMO.

8.1. Bivalirudin

Bivalirudin is a DTI potentially used as an alternative anticoagulant in patients with ECMO. Bivalirudin is approved during percutaneous coronary intervention in patients with or at risk of HIT [75]. Its use in ECMO anticoagulation is considered off-label; however, it has been used as a primary choice for anticoagulant therapy in patients with heparin resistance, HIT, or those requiring surgery [75].
Its mechanism of action is based on reversible binding to the active site of thrombin with the onset of action within 4 min after bolus administration [74]. Bivalirudin has a relatively small volume of distribution, indicating that it remains primarily within the bloodstream. It does not bind extensively to plasma proteins and is predominantly metabolised by proteolytic cleavage via plasma carboxypeptidases, forming various metabolites with reduced anticoagulant activity [76]. Bivalirudin and its metabolites are primarily eliminated renally. The half-life of bivalirudin ranges from 25 to 80 min in patients with normal renal function [77]. In patients with impaired renal function, including those with end-stage renal disease that requires hemodialysis, dosage adjustments are necessary to prevent drug accumulation and potential bleeding complications. In patients with ECMO, the reported dose of bivalirudin as a continuous infusion is in the range of 0.02–0.05 µg/kg/min [78].

8.2. Monitoring and Targets

The aPTT test is widely used to adjust and monitor the dosage of bivalirudin [77]. The same aPTT targets are commonly used for bivalirudin as for UFH, with the aim of a range of 1.5 to 2.5 times the patient’s baseline aPTT [78]. However, the correlation between the dosage and the response of aPTT is not flawless and has been described as weak to moderate [79]. Alternative tests may correlate strongly with bivalirudin doses, such as the ecarin chromogenic assay, the dilute thrombin time (dTT) and the chromogenic anti-IIa assay dTT, which involves plasma-diluted thrombin time and exhibits a better correlation with bivalirudin dosage compared to aPTT [80]. The bivalirudin dosing regimen and its monitoring targets are summarised in Table 1. The primary safety issue of bivalirudin lies in the absence of a specific antidote. A cessation of bivalirudin infusion can manage cases of bleeding, as its effect vanishes quickly due to its short half-life. In cases of severe bleeding, the elimination of bivalirudin can be facilitated by plasmapheresis or hemofiltration [81]; the administration of activated factor VII can reverse its effect [82]. Another issue concerns the lower effectiveness of bivalirudin under low-flow conditions with increased degradation of bivalirudin in the region of a stasis. Specifically, several studies reported increased thrombosis in reperfusion cannulas and insufficiently unloaded left ventricle in patients on ECMO anticoagulated with bivalirudin [83,84].
The limited scientific evidence supporting the use of bivalirudin in the context of ECMO anticoagulation is derived primarily from retrospective cohort studies and case series. All these studies compared patients who received bivalirudin with a control group who received UFH [85,86,87]. These reports did not describe any significant increase in bleeding or thrombotic complications associated with bivalirudin compared to UFH. Furthermore, a recent meta-analysis of nine retrospective studies showed a reduction in hospital mortality (OR = 0.65, 95% CI (0.44, 0.95), p = 0.03) and thrombotic events (OR = 0.55, 95% CI (0.37, 0.83), p = 0.004) in bivalirudin-anticoagulated patients of ECMO compared to the UFH group. However, major bleeding incidents, the duration of ECMO, and critical circuit events did not differ between the groups [88]. Based on the above reports, bivalirudin represents safe and reliable anticoagulation in patients with ECMO. However, further studies must fully elucidate its role in the ECMO anticoagulation strategy.

8.3. Argatroban

Argatroban represents another DTI as an alternative anticoagulant to UFH in ECMO patients. Argatroban is metabolised in the liver through several pathways, including the cytochrome P450 enzyme [89], resulting in a final half-life of 45 min [90]. There are four metabolites, one partially active [91]. Therefore, in patients with liver failure, the half-life can be prolonged up to two- to threefold, requiring adequate dose reduction [90]. Several studies report liver failure in critically ill patients, including patients with ECMO, with the need for a significant reduction in argatroban infusion rates [92,93,94,95,96]. Renal failure, with or without the need for CRRT, does not influence argatroban metabolism or clearance [95]. Therefore, dose adjustments are not necessary [97]. Argatroban is administered as a continuous intravenous infusion with reported infusion rates of 0.1–0.3 µg/kg/min in patients with ECMO [98].

8.4. Monitoring and Target

Similarly to bivalirudin, aPTT represents the most common monitoring method for argatroban anticoagulation, with a target of 50–70 s (Table 1) reported in the literature [98]. ACT has also been used as a single parameter or combined with aPTT to guide argatroban anticoagulation with a wide range of 150 to 230 s [98]. Furthermore, the ecarin chromogenic assay (ECA), with its high specificity to monitor DTI or VETs, did not correlate with aPPT levels [97]. Interestingly, a significant proportion of patients who reached therapeutic levels of aPTT had normal values of ECA and VETs [99]. Such findings allude to the fact that the currently used argatroban aPTT targets may not provide adequate anticoagulation in patients with ECMO. On the other hand, none of the studies reported a higher incidence of thrombotic or bleeding complications in ECMO patients anticoagulated with argatroban compared to UFH [98]. Monitoring of argatroban effect with VETs is nowadays not standardised. One study investigated VETs to guide anticoagulation in child ECMO patients with bivalirudin. The authors discovered a moderate correlation between aPTT and VET results [64].
There is no specific antidote to argatroban. In severe bleeding, activated factor VII was successfully used to reverse its effect [100]. Despite its higher cost, argatroban anticoagulation may represent an alternative strategy compared to UFH due to the reported lower administration of blood products [101].

9. Conclusions

The balance of hemostatic mechanisms in ECMO patients is seriously disturbed from many points of view. First, the critical condition itself is a serious factor. Second, the unique influence of a particular underlying disease must be considered, i.e., COVID-19. Third, the disturbance caused by artificial materials of the extracorporeal circuit, oxygenator, and changes in blood homeostasis also play a significant role. Last but not least, the physician’s efforts to manage possible thrombotic or bleeding events, the choice of anticoagulant agent, and the targeting of the right numbers of the monitoring modality are all critical factors that should lead to better clinical outcomes.

Author Contributions

Conceptualisation, J.Š. and M.P.; methodology, J.Š., M.P. and P.M.; writing—original draft preparation, J.Š., M.S., H.Ř., M.L., M.B. and M.P.; writing—review and editing: P.M., M.B. and M.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Czech Ministry of Health, grant MZCZ-DRO-VFN64165.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ratnani, I.; Tuazon, D.; Zainab, A.; Uddin, F. The Role and Impact of Extracorporeal Membrane Oxygenation in Critical Care. Methodist DeBakey Cardiovasc. J. 2018, 14, 110–119. [Google Scholar] [CrossRef] [PubMed]
  2. Aljishi, R.S.; Alkuaibi, A.H.; Al Zayer, F.A.; Al Matouq, A.H. Extracorporeal Membrane Oxygenation for COVID-19: A Systematic Review. Cureus 2022, 14, e27522. [Google Scholar] [CrossRef] [PubMed]
  3. Makdisi, G.; Wang, I.-W. Extra Corporeal Membrane Oxygenation (ECMO) review of a lifesaving technology. J. Thorac. Dis. 2015, 7, E166–E176. [Google Scholar] [CrossRef] [PubMed]
  4. Organisation ELS. ELSO Guidelines. 2020. Available online: https://wwwelsoorg/ecmo-resources/elso-ecmoguidelinesaspx (accessed on 25 May 2023).
  5. Squiers, J.J.; Lima, B.; DiMaio, J.M. Contemporary extracorporeal membrane oxygenation therapy in adults: Fundamental principles and systematic review of the evidence. J. Thorac. Cardiovasc. Surg. 2016, 152, 20–32. [Google Scholar] [CrossRef] [PubMed]
  6. Sanivarapu, R.R.; Osman, U.; Kumar, A.L. A Systematic Review of Mortality Rates Among Adult Acute Respiratory Distress Syndrome Patients Undergoing Extracorporeal Membrane Oxygenation Therapy. Cureus 2023, 15, e43590. [Google Scholar] [CrossRef]
  7. El Sibai, R.; Bachir, R.; El Sayed, M. ECMO use and mortality in adult patients with cardiogenic shock: A retrospective observational study in U.S. hospitals. BMC Emerg. Med. 2018, 18, 20. [Google Scholar] [CrossRef]
  8. Olson, S.R.; Murphree, C.R.; Zonies, D.; Meyer, A.D.; Mccarty, O.J.T.; Deloughery, T.G.; Shatzel, J.J. Thrombosis and Bleeding in Extracorporeal Membrane Oxygenation (ECMO) Without Anticoagulation: A Systematic Review. ASAIO J. 2020, 67, 290–296. [Google Scholar] [CrossRef]
  9. Mulder, M.M.G.; Fawzy, I.; Lancé, M.D. ECMO and anticoagulation: A comprehensive review. Neth. J. Crit. Care 2018, 26, 6–13. [Google Scholar]
  10. Nunez, J.I.; Gosling, A.F.; O’gara, B.; Kennedy, K.F.; Rycus, P.; Abrams, D.; Brodie, D.; Shaefi, S.; Garan, A.R.; Grandin, E.W. Bleeding and thrombotic events in adults supported with venovenous extracorporeal membrane oxygenation: An ELSO registry analysis. Intensive Care Med. 2021, 48, 213–224. [Google Scholar] [CrossRef]
  11. McMichael, A.B.; Ryerson, L.M.; Ratano, D.; Fan, E.; Faraoni, D.; Annich, G.M. 2021 ELSO Adult and Pediatric Anticoagulation Guidelines. ASAIO J. 2022, 68, 303–310. [Google Scholar] [CrossRef]
  12. Burša, F.; Sklienka, P.; Frelich, M.; Jor, O.; Ekrtová, T.; Máca, J. Anticoagulation Management during Extracorporeal Membrane Oxygenation—A Mini-Review. Medicina 2022, 58, 1783. [Google Scholar] [CrossRef] [PubMed]
  13. Karagiannidis, C.; Windisch, W.; Bein, T. Rapid Changes in Arterial Carbon Dioxide Levels Caused by Extracorporeal Membrane Oxygenation. The Temptation of a Fascinating Technology. Am. J. Respir. Crit. Care Med. 2020, 201, 1466–1468. [Google Scholar] [CrossRef]
  14. Abruzzo, A.; Gorantla, V.; Thomas, S.E. Venous thromboembolic events in the setting of extracorporeal membrane oxy-genation support in adults: A systematic review. Thromb. Res. 2022, 212, 58–71. [Google Scholar] [CrossRef]
  15. Yusuff, H.; Zochios, V.; Brodie, D. Thrombosis and coagulopathy in COVID-19 patients rceiving ECMO: A narrative review of current literature. J. Cardiothorac. Vasc. Anesth. 2022, 36, 3312–3317. [Google Scholar] [CrossRef] [PubMed]
  16. Bonaventura, A.; Vecchié, A.; Dagna, L.; Martinod, K.; Dixon, D.L.; Van Tassell, B.W.; Dentali, F.; Montecucco, F.; Massberg, S.; Levi, M.; et al. Endothelial dysfunction and immunothrombosis as key pathogenic mechanisms in COVID-19. Nat. Rev. Immunol. 2021, 21, 319–329. [Google Scholar] [CrossRef] [PubMed]
  17. Aliter, K.F.; Al-Horani, R.A. Thrombin Inhibition by Argatroban: Potential Therapeutic Benefits in COVID-19. Cardiovasc. Drugs Ther. 2020, 35, 195–203. [Google Scholar] [CrossRef]
  18. Zuo, Y.; Warnock, M.; Harbaugh, A.; Yalavarthi, S.; Gockman, K.; Zuo, M.; Madison, J.A.; Knight, J.S.; Kanthi, Y.; Lawrence, D.A. Plasma tissue plasminogen activator and plasminogen activator inhibitor-1 in hospitalized COVID-19 patients. Sci. Rep. 2021, 11, 1580. [Google Scholar] [CrossRef]
  19. Conway, E.M.; Mackman, N.; Warren, R.Q.; Wolberg, A.S.; Mosnier, L.O.; Campbell, R.A.; Gralinski, L.E.; Rondina, M.T.; van de Veerdonk, F.L.; Hoffmeister, K.M.; et al. Understanding COVID-19-associated coagulopathy. Nat. Rev. Immunol. 2022, 22, 639–649. [Google Scholar] [CrossRef]
  20. Mansour, A.; Flecher, E.; Schmidt, M.; Rozec, B.; Gouin-Thibault, I.; Esvan, M.; Fougerou, C.; Levy, B.; Porto, A.; Ross, J.T.; et al. Bleeding and thrombotic events in patients with severe COVID-19 supported with extracorporeal membrane oxygenation: A nationwide cohort study. Intensive Care Med. 2022, 48, 1039–1052. [Google Scholar] [CrossRef]
  21. Fanning, J.P.; Weaver, N.; Fanning, R.B.; Griffee, M.J.; Cho, S.-M.; Panigada, M.; Obonyo, N.G.; Zaaqoq, A.M.; Rando, H.; Chia, Y.W.; et al. Hemorrhage, Disseminated Intravascular Coagulopathy, and Thrombosis Complications Among Critically Ill Patients with COVID-19: An International COVID-19 Critical Care Consortium Study. Crit. Care Med. 2023, 51, 619–631. [Google Scholar] [CrossRef]
  22. Lannon, M.; Duda, T.; Greer, A.; Hewitt, M.; Sharma, A.; Martyniuk, A.; Owen, J.; Amin, F.; Sharma, S. Intracranial hemorrhage in patients treated for SARS-CoV-2 with extracorporeal membrane oxygenation: A systematic review and meta-analysis. J. Crit. Care 2023, 77, 154319. [Google Scholar] [CrossRef] [PubMed]
  23. Jin, Y.; Zhang, Y.; Liu, J.; Zhou, Z. Thrombosis and bleeding in patients with COVID-19 requiring extracorporeal membrane oxygenation: A systematic review and meta-analysis. Res. Pract. Thromb. Haemost. 2023, 7, 100103. [Google Scholar] [CrossRef] [PubMed]
  24. Shekar, K.; Badulak, J.; Peek, G.; Boeken, U.; Dalton, H.J.; Arora, L.; Zakhary, B.; Ramanathan, K.; Starr, J.; Akkanti, B.; et al. Extracorporeal life support organization COVID-19 interim Guidelines. Asaio J. Am. Soc. Artif. Intern. Organs 2000, 66, 707–721. [Google Scholar] [CrossRef]
  25. Mancini, I.; Baronciani, L.; Artoni, A.; Colpani, P.; Biganzoli, M.; Cozzi, G.; Novembrino, C.; Anzoletti, M.B.; De Zan, V.; Pagliari, M.T.; et al. The ADAMTS13-von Willebrand factor axis in COVID-19 patients. J. Thromb. Haemost. 2020, 19, 513–521. [Google Scholar] [CrossRef]
  26. Kalbhenn, J.; Glonnegger, H.; Wilke, M.; Bansbach, J.; Zieger, B. Hypercoagulopathy, acquired coagulation disorders and anticoagulation before, during and after extracorporeal membrane oxygenation in COVID-19: A case series. Perfusion 2021, 36, 592–602. [Google Scholar] [CrossRef] [PubMed]
  27. Boira, I.; Esteban, V.; Vañes, S.; Castelló, C.; Celis, C.; Chiner, E. Major Bleeding Complications in COVID-19 Patients. Cureus 2021, 13, e16816. [Google Scholar] [CrossRef] [PubMed]
  28. McAlpine, L.S.; Zubair, A.S.; Maran, I.; Chojecka, P.; Lleva, P.; Jasne, A.S.; Navaratnam, D.; Matouk, C.; Schindler, J.; Sheth, K.N.; et al. Ischemic Stroke, Inflammation, and Endotheliopathy in COVID-19 Patients. Stroke 2021, 52, e233–e238. [Google Scholar] [CrossRef]
  29. Bemtgen, X.; Zotzmann, V.; Benk, C.; Rilinger, J.; Steiner, K.; Asmussen, A.; Bode, C.; Wengenmayer, T.; Maier, S.; Staudacher, D.L. Thrombotic circuit complications during veno-venous extracorporeal membrane oxygenation in COVID-19. J. Thromb. Thrombolysis 2021, 51, 301–307. [Google Scholar] [CrossRef]
  30. Spiess, B.D.; Armour, S.; Horrow, J.; Kaplan, J.A.; Koch, C.G.; Karkouti, K.; Body, S.C. Chapter 27—Transfusion Medicine and Coagulation Disorders. In Kaplan’s Essentials of Cardiac Anesthesia, 2nd ed.; Kaplan, J.A., Ed.; Elsevier: Amsterdam, The Netherlands, 2018; pp. 685–714. ISBN 9780323497985. [Google Scholar]
  31. Devlin, A.; Mycroft-West, C.; Procter, P.; Cooper, L.; Guimond, S.; Lima, M.; Yates, E.; Skidmore, M. Tools for the Quality Control of Pharmaceutical Heparin. Medicina 2019, 55, 636. [Google Scholar] [CrossRef]
  32. Esper, S.A.; Welsby, I.J.; Subramaniam, K.; Wallisch, W.J.; Levy, J.H.; Waters, J.H.; Triulzi, D.J.; Hayanga, J.W.A.; Schears, G.J. Adult extracorporeal membrane oxygenation: An international survey of transfusion and anticoagulation techniques. Vox Sang. 2017, 112, 443–452. [Google Scholar] [CrossRef]
  33. Bojar, R.M. Manual of Perioperative Care in Adult Cardiac Surgery; John Wiley & Sons: Hoboken, NJ, USA, 2020. [Google Scholar] [CrossRef]
  34. Chlebowski, M.M.; Baltagi, S.; Carlson, M.; Levy, J.H.; Spinella, P.C. Clinical controversies in anticoagulation monitoring and antithrombin supplementation for ECMO. Crit. Care 2020, 24, 19. [Google Scholar] [CrossRef] [PubMed]
  35. Dalton, H.; Martin, M.; Garcia-Filion, P.; Shavelle, D.; Huang, P.-H.; Clark, J.; Beinart, S.; Israel, A.; Korpi-Steiner, N. Activated clotting time in inpatient diagnostic and interventional settings. J. Thromb. Thrombolysis 2022, 54, 660–668. [Google Scholar] [CrossRef] [PubMed]
  36. Toulon, P.; Smahi, M.; De Pooter, N. APTT therapeutic range for monitoring unfractionated heparin therapy. Significant impact of the anti-Xa reagent used for correlation. J. Thromb. Haemost. 2021, 19, 2002–2006. [Google Scholar] [CrossRef] [PubMed]
  37. Smythe, M.A.; Koerber, J.M.; Nowak, S.N.; Mattson, J.C.; Begle, R.L.; Westley, S.J.; Balasubramaniam, M. Correlation between Activated Clotting Time and Activated Partial Thromboplastin Times. Ann. Pharmacother. 2002, 36, 7–11. [Google Scholar] [CrossRef] [PubMed]
  38. Panigada, M.; EIapichino, G.; Brioni, M.; Panarello, G.; Protti, A.; Grasselli, G.; Occhipinti, G.; Novembrino, C.; Consonni, D.; Arcadipane, A.; et al. Thromboelastography-based anticoagulation management during extracorporeal membrane oxygenation: A safety and feasibility pilot study. Ann. Intensive Care 2018, 8, 7. [Google Scholar] [CrossRef]
  39. Henderson, N.; Sullivan, J.E.; Myers, J.; Wells, T.; Calhoun, A.; Berkenbosch, J.; Tzanetos, D.T. Use of thromboelastography to predict thrombotic complications in pediatric and neonatal extracorporeal membranous oxygenation. J. Extra-Corpor. Technol. 2018, 50, 149–154. [Google Scholar] [CrossRef]
  40. van de Wetering, J.; Westendorp, R.G.; van der Hoeven, J.G.; Stolk, B.; Feuth, J.D.; Chang, P.C. Heparin use in continuous renal replacement procedures. J. Am. Soc. Nephrol. 1996, 7, 145–150. [Google Scholar] [CrossRef]
  41. Nguyen, T.P.; Phan, X.T.; Nguyen, T.H.; Huynh, D.Q.; Tran, L.T.; Pham, H.M.; Nguyen, T.N.; Kieu, H.T.; Pham, T.T.N. Major Bleeding in Adults Undergoing Peripheral Extracorporeal Membrane Oxygenation (ECMO): Prognosis and Predictors. Crit. Care Res. Pract. 2022, 2022, 5348835. [Google Scholar] [CrossRef]
  42. Durrani, J.; Malik, F.; Ali, N.; Jafri, S.I.M. To be or not to be a case of heparin resistance. J. Community Hosp. Intern. Med. Perspect. 2018, 8, 145–148. [Google Scholar] [CrossRef]
  43. Chen, Y.; Phoon, P.H.Y.; Hwang, N.C. Heparin Resistance During Cardiopulmonary Bypass in Adult Cardiac Surgery. J. Cardiothorac. Vasc. Anesth. 2022, 36, 4150–4160. [Google Scholar] [CrossRef]
  44. Novelli, C.; Borotto, E.; Beverina, I.; Punzi, V.; Radrizzani, D.; Brando, B. Heparin dosage, level, and resistance in SARS-CoV2 infected patients in intensive care unit. Int. J. Lab. Hematol. 2021, 43, 1284–1290. [Google Scholar] [CrossRef]
  45. Kleinschmidt, K.; Charles, R. Pharmacology of low molecular weight harparins. Emerg. Med. Clin. North Am. 2001, 19, 1025–1049. [Google Scholar] [CrossRef] [PubMed]
  46. PubChem [Internet]. Bethesda (MD): National Library of Medicine (US), National Center for Biotechnology Information. 2004-. PubChem Compound Summary for CID 5282448, Fondaparinux. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Fondaparinux (accessed on 13 May 2023).
  47. Toschi, V.; Lettino, M. Fondaparinux: Pharmacology and clinical experience in cardiovascular medicine. Mini-Rev. Med. Chem. 2007, 7, 383–387. [Google Scholar] [CrossRef] [PubMed]
  48. Martel, N.; Lee, J.; Wells, P.S. Risk for heparin-induced thrombocytopenia with unfractionated and low-molecular-weight heparin thromboprophylaxis: A meta-analysis. Blood 2005, 106, 2710–2715. [Google Scholar] [CrossRef]
  49. Solari, F.; Varacallo, M. Low Molecular Weight Heparin (LMWH). In StatPearl; StatPearls: Treasure Island, FL, USA, 2023. [Google Scholar]
  50. Hirsh, J.; Warkentin, T.E.; Shaughnessy, S.G.; Anand, S.S.; Halperin, J.L.; Raschke, R.; Granger, C.; Ohman, E.M.; Dalen, J.E. Heparin and Low-Molecular-Weight Heparin Mechanisms of Action, Pharmacokinetics, Dosing, Monitoring, Efficacy, and Safety. Chest 2001, 119, 64S–94S. [Google Scholar] [CrossRef]
  51. Cihlar, R.; Sramek, V.; Papiez, A.; Penka, M.; Suk, P. Pharmacokinetic Comparison of Subcutaneous and Intravenous Nadroparin Administration for Thromboprophylaxis in Critically Ill Patients on Vasopressors. Pharmacology 2019, 105, 73–78. [Google Scholar] [CrossRef]
  52. Dörffler-Melly, J.; de Jonge, E.; de Pont, A.-C.; Meijers, J.; Vroom, M.B.; Büller, H.R.; Levi, M. Bioavailability of subcutaneous low-molecular-weight heparin to patients on vasopressors. Lancet 2002, 359, 849–850. [Google Scholar] [CrossRef]
  53. Krueger, K.; Schmutz, A.; Zieger, B.; Kalbhenn, J. Venovenous Extracorporeal Membrane Oxygenation with Prophylactic Subcutaneous Anticoagulation Only: An Observational Study in More Than 60 Patients. Artif. Organs 2016, 41, 186–192. [Google Scholar] [CrossRef]
  54. Kurihara, C.; Walter, J.M.; Karim, A.; Thakkar, S.; Saine, M.; Odell, D.D.; Kim, S.; Tomic, R.; Wunderink, R.G.; Budinger, G.R.S.; et al. Feasibility of Venovenous Extracorporeal Membrane Oxygenation Without Systemic Anticoagulation. Ann. Thorac. Surg. 2020, 110, 1209–1215. [Google Scholar] [CrossRef] [PubMed]
  55. Aubron, C.; McQuilten, Z.; Bailey, M.; Board, J.; Buhr, H.; Cartwright, B.; Dennis, M.; Hodgson, C.; Forrest, P.; McIlroy, D.; et al. Low-Dose Versus Therapeutic Anticoagulation in Patients on Extracorporeal Membrane Oxygenation: A Pilot Randomized Trial. Crit. Care Med. 2019, 47, e563–e571. [Google Scholar] [CrossRef] [PubMed]
  56. Beitland, S.; Sandven, I.; Kjærvik, L.K.; Sandset, P.M.; Sunde, K.; Eken, T. Thromboprophylaxis with low molecular weight heparin versus unfractioned heparin in intensive care patients: A systematic review with meta-analysis and trial sequential analysis. Intensive Care Med. 2015, 41, 1209–1219. [Google Scholar] [CrossRef] [PubMed]
  57. PROTECT Investigators for the Canadian Critical Care Trials Group. The Australian and New Zealand Intensive Care Society Clinical Trials Group Dalteparin versus unfractionated heparin in critically ill patients. N. Engl. J. Med. 2011, 364, 1305–1314. [Google Scholar] [CrossRef] [PubMed]
  58. De, A.; Roy, P.; Garg, V.K.; Pandey, N.K. Low-molecular-weight heparin and unfractioned heparin in prophylaxis against deep vein thrombosis in critically ill patients undergoing major surgery. Blood Coagul. Fibrinolysis 2010, 21, 57–61. [Google Scholar] [CrossRef] [PubMed]
  59. Pon, T.K.; Dager, W.E.; Roberts, A.J.; White, R.H. Subcutaneous Enoxaparin for Therapeutic Anticoagulation in Hemodialysis Patients. Thromb. Res. 2014, 133, 1023–1028. [Google Scholar] [CrossRef]
  60. Ucar, E.Y.; Akgun, M.; Araz, O.; Tas, H.; Kerget, B.; Meral, M.; Kaynar, H.; Saglam, L. Comparison of LMWH Versus UFH for Hemorrhage and Hospital Mortality in the Treatment of Acute Massive Pulmonary Thromboembolism after Thrombolytic Treatment. Lung 2014, 193, 121–127. [Google Scholar] [CrossRef]
  61. Merli, G.; Spiro, T.E.; Olsson, C.G.; Abildgaard, U.; Davidson, B.L.; Eldor, A.; Elias, D.; Grigg, A.; Musset, D.; Rodgers, G.M.; et al. Subcutaneous enoxaparin once or twice daily compared with intravenous unfractioned heparin for treatment of venous thromboembolic disease. Ann. Intern. Med. 2001, 134, 191–202. [Google Scholar] [CrossRef]
  62. Johnson, P.N.; Benefield, E.C.; Bui, P.-Y.N.; Gausman, J.N.; Marlar, R.A.; Gessouroun, M.R. Fondaparinux Monitoring: Need for a Local Fondaparinux-Calibrated Anti–F actor Xa Assay. J. Pediatr. Pharmacol. Ther. 2013, 18, 318–319. [Google Scholar] [CrossRef]
  63. Gratz, J.; Pausch, A.; Schaden, E.; Baierl, A.; Jaksch, P.; Erhart, F.; Hoetzenecker, K.; Wiegele, M. Low molecular weight heparin versus unfractioned heparin for anticoagulation during perioperative extracorporeal membrane oxygenation: A single center experience in 102 lung transplant patients. Artif. Organs 2020, 44, 638–646. [Google Scholar] [CrossRef]
  64. Pavoni, V.; Gianesello, L.; Conti, D.; Ballo, P.; Dattolo, P.; Prisco, D.; Görlinger, K. “In Less than No Time”: Feasibility of Rotational Thromboelastometry to Detect Anticoagulant Drugs Activity and to Guide Reversal Therapy. J. Clin. Med. 2022, 11, 1407. [Google Scholar] [CrossRef]
  65. Parlar, A.I.; Sayar, U.; Cevirme, D.; Yuruk, M.A.; 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, 344–347. [Google Scholar] [CrossRef]
  66. Degani, A.; Milanesi, E.; Rossini, B.; Ricccardi, M.C.; Pellegrini, C.; Doronzo, F.; Piscione, V.; Pelenghi, S.; Belliato, M. Case report: Use of subcutaneous Fondaparinux in a COVID 19 patient with heparin-induced thrombocytopenia during venous-venous ECMO. Perfusion 2021, 36 (Suppl. 1), 46–47. [Google Scholar]
  67. Rychlíčková, J.; Šrámek, V.; Suk, P. Use of fondaparinux in patients with heparin-induced thrombocytopenia on veno-venous extracorporeal membrane oxygenation: A three-patient case series report. Front. Med. 2023, 10, 1112770. [Google Scholar] [CrossRef] [PubMed]
  68. Jiritano, F.; Serraino, G.F.; Ten Cate, H.; Fina, D.; Matteucci, M.; Mastroroberto, P.; Lorusso, R. Platelets and extra-corporeal membrane oxygenation in adult patients: A systematic review and meta-analysis. Intensive Care Med. 2020, 46, 1154–1169. [Google Scholar] [CrossRef]
  69. Moreno-Duarte, I.; Ghadimi, K. Heparin induced thrombocytopenia for the perioperative and critical care clinician. Curr. Anesthesiol. Rep. 2020, 10, 501–511. [Google Scholar] [CrossRef] [PubMed]
  70. Althaus, K.; Straub, A.; Häberle, H.; Rosenberger, P.; Hidiatov, O.; Hammer, S.; Nowak-Harnau, S.; Enkel, S.; Riessen, R.; Bakchoul, T. Heparin-induced thrombocytopenia: Diagnostic challenges in intensive care patients especially with extracorporeal circulation. Thromb. Res. 2020, 188, 52–60. [Google Scholar] [CrossRef]
  71. Hogan, M.; Berger, J.S. Heparin-induced thrombocytopenia (HIT): Review of incidence, diagnosis, and management. Vasc. Med. 2020, 25, 160–173. [Google Scholar] [CrossRef]
  72. Páramo, J.A.; Lozano, M.L.; González-Porras, J.R.; Mateo, J. Estado actual del diagnóstico y tratamiento de la trombocitopenia inducida por heparina (TIH). Med. Clin. 2021, 158, 82–89. [Google Scholar] [CrossRef]
  73. Linkins, L.; Hu, G.; Warkentin, T.E. Systematic review of fondaparinux for heparin-induced thrombocytopenia: When there are no randomized controlled trials. Res. Pract. Thromb. Haemost. 2018, 2, 678–683. [Google Scholar] [CrossRef]
  74. Di Nisio, M.; Middeldorp, S.; Büller, H.R. Direct Thrombin Inhibitors. N. Engl. J. Med. 2005, 353, 1028–1040. [Google Scholar] [CrossRef]
  75. Stone, G.W.; Witzenbichler, B.; Guagliumi, G.; Peruga, J.Z.; Brodie, B.R.; Dudek, D.; Kornowski, R.; Hartmann, F.; Gersh, B.J.; Pocock, S.J.; et al. Bivalirudin during Primary PCI in Acute Myocardial Infarction. N. Engl. J. Med. 2008, 358, 2218–2230. [Google Scholar] [CrossRef]
  76. Van De Car, D.A.; Rao, S.V.; Ohman, E.M. Bivalirudin: A review of the pharmacology and clinical application. Expert Rev. Cardiovasc. Ther. 2010, 8, 1673–1681. [Google Scholar] [CrossRef] [PubMed]
  77. Van Cott, E.M.; Roberts, A.J.; Dager, W.E. Laboratory monitoring of parenteral direct thrombin inhibitors. Semin. Thromb. Hemost. 2017, 43, 270–276. [Google Scholar] [CrossRef] [PubMed]
  78. Helms, J.; Frere, C.; Thiele, T.; Tanaka, K.A.; Neal, M.D.; Steiner, M.E.; Connors, J.M.; Levy, J.H. Anticoagulation in adult patients supported with extracorporeal membrane oxygenation: Guidance from the Scientific and Standardization Committees on Perioperative and Critical Care Haemostasis and Thrombosis of the International Society on Thrombosis and Haemostasis. J. Thromb. Haemost. 2022, 21, 373–396. [Google Scholar] [CrossRef] [PubMed]
  79. Snyder, C.W.; Goldenberg, N.A.; Nguyen, A.T.H.; Smithers, C.J.; Kays, D.W. A perioperative bivalirudin anticoagulation protocol for neonates with congenital diaphragmatic hernia on extracorporeal membrane oxygenation. Thromb. Res. 2020, 193, 198–203. [Google Scholar] [CrossRef]
  80. Beyer, J.T.; Lind, S.E.; Fisher, S.; Trujillo, T.C.; Wempe, M.F.; Kiser, T.H. Evaluation of intravenous direct thrombin inhibitor monitoring tests: Correlation with plasma concentrations and clinical outcomes in hospitalized patients. J. Thromb. Thrombolysis 2019, 49, 259–267. [Google Scholar] [CrossRef]
  81. Preston, T.J.; Dalton, H.J.; Nicol, K.K.; Ferrall, B.R.; Miller, J.C.; Hayes, D. Plasma Exchange on Venovenous Extracorporeal Membrane Oxygenation with Bivalirudin Anticoagulation. World J. Pediatr. Congenit. Heart Surg. 2014, 6, 119–122. [Google Scholar] [CrossRef]
  82. Young, G.; Yonekawa, K.E.; Nakagawa, P.A.; Blain, R.C.; Lovejoy, A.E.; Nugent, D.J. Recombinant activated factor VII effectively reverses the anticoagulant effects of heparin, enoxaparin, fondaparinux, argatroban, and bivalirudin ex vivo as measured using thromboelastography. Blood Coagul. Fibrinolysis 2007, 18, 547–553. [Google Scholar] [CrossRef]
  83. Ranucci, M. Bivalirudin and post-cardiotomy ECMO: A word of caution. Crit. Care 2012, 16, 427. [Google Scholar] [CrossRef]
  84. Pieri, M.; Agracheva, N.; Bonaveglio, E.; Greco, T.; De Bonis, M.; Covello, R.D.; Zangrillo, A.; Pappalardo, F. Bivalirudin Versus Heparin as an Anticoagulant During Extracorporeal Membrane Oxygenation: A Case-Control Study. J. Cardiothorac. Vasc. Anesth. 2013, 27, 30–34. [Google Scholar] [CrossRef]
  85. Kaseer, H.; Soto-Arenall, M.; Sanghavi, D.; Moss, J.; Ratzlaff, R.; Pham, S.; Guru, P. Heparin vs bivalirudin anticoagulation for extracorporeal membrane oxygenation. J. Card. Surg. 2020, 35, 779–786. [Google Scholar] [CrossRef]
  86. Macielak, S.; Burcham, P.; Whitson, B.; Abdel-Rasoul, M.; Rozycki, A. Impact of anticoagulation strategy and agents on extracorporeal membrane oxygenation therapy. Perfusion 2019, 34, 671–678. [Google Scholar] [CrossRef] [PubMed]
  87. Rivosecchi, R.M.; Arakelians, A.R.; Ryan, J.; Murray, H.; Ramanan, R.; Gomez, H.; Phillips, D.D.; Sciortino, C.; Arlia, P.M.; Freeman, D.C.; et al. Comparison of Anticoagulation Strategies in Patients Requiring Venovenous Extracorporeal Membrane Oxygenation: Heparin Versus Bivalirudin. Crit. Care Med. 2021, 49, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
  88. Li, D.-H.; Sun, M.-W.; Zhang, J.-C.; Zhang, C.; Deng, L.; Jiang, H. Is bivalirudin an alternative anticoagulant for extracorporeal membrane oxygenation (ECMO) patients? A systematic review and meta-analysis. Thromb. Res. 2021, 210, 53–62. [Google Scholar] [CrossRef] [PubMed]
  89. Ahmad, S.; Ahsan, A.; George, M.; Iqbal, O.; Jeske, W.P.; McKenna, R.; Lewis, B.E.; Walenga, J.M.; Fareed, J. Simultaneous monitoring of argatroban and its major metabolite using an HPLC method: Potential clinical applications. Clin. Appl. Thromb./Hemost. 1999, 5, 252–258. [Google Scholar] [CrossRef] [PubMed]
  90. Swan, S.K.; Hursting, M.J. The pharmacokinetics and pharmacodynamics of argatroban: Effects of age, ender, and hepatic or renal dysfunction. Pharmacotherapy 2000, 20, 318–329. [Google Scholar] [CrossRef]
  91. McKeage, K.; Plosker, G.L. Argatroban. Drugs 2001, 61, 515–522; discussion 523–514. [Google Scholar] [CrossRef]
  92. Beiderlinden, M.; Treschan, T.; Gorlinger, K.; Peters, J. Argatroban in extracorporeal membrane oxygenation. Artif. Organs 2007, 31, 461–465. [Google Scholar] [CrossRef]
  93. Dolch, M.E.; Frey, L.; Hatz, R.; Uberfuhr, P.A.; Beiras-Fernandez, A.; Behr, J.; Irlbeck, M. Lung Transplant Group TM: Extracorporeal membrane oxygenation bridging to lung transplant complicated by heparin-induced thrombocytopenia. Exp. Clin. Transplant. Off. J. Middle East Soc. Organ Transplant. 2010, 8, 329–332. [Google Scholar]
  94. Saugel, B.; Phillip, V.; Moessmer, G.; Schmid, R.M.; Huber, W. Argatroban therapy for heparin-induced thrombocytopenia in ICU patients with multiple organ dysfunction syndrome: A retrospective study. Crit. Care 2010, 14, R90. [Google Scholar] [CrossRef]
  95. Guzzi, L.M.; McCollum, D.A.; Hursting, M.J. Effect of renal function on argatroban therapy in heparin-induced thrombocytopenia. J. Thromb. Thrombolysis 2006, 22, 169–176. [Google Scholar] [CrossRef]
  96. Dingman, J.S.; Smith, Z.R.; Coba, V.E.; Peters, M.A.; To, L. Argatroban dosing requirements in extracorporeal life support and other critically ill populations. Thromb. Res. 2020, 189, 69–76. [Google Scholar] [CrossRef] [PubMed]
  97. Geli, J.; Capoccia, M.; Maybauer, D.M.; Maybauer, M.O. Argatroban Anticoagulation for Adult Extracorporeal Membrane Oxygenation: A Systematic Review. J. Intensive Care Med. 2021, 37, 459–471. [Google Scholar] [CrossRef] [PubMed]
  98. Seidel, H.; Kolde, H.J. Monitoring of Argatroban and Lepirudin: What is the Input of Laboratory Values in “Real Life”? Clin. Appl. Thromb./Hemost. 2018, 24, 287–294. [Google Scholar] [CrossRef] [PubMed]
  99. Smythe, M.A.; Forsyth, L.L.; Warkentin, T.E.; Smith, M.D.; Sheppard, J.-A.I.; Shannon, F. Progressive, Fatal Thrombosis Associated with Heparin-Induced Thrombocytopenia After Cardiac Surgery Despite “Therapeutic” Anticoagulation With Argatroban: Potential Role for PTT and ACT Confounding. J. Cardiothorac. Vasc. Anesth. 2015, 29, 1319–1321. [Google Scholar] [CrossRef]
  100. Rieg, A.D.; Grottke, O.; Schälte, G.; Spillner, J.; Autschbach, R.; Rossaint, R.; Hein, M. Recombinant factor VIIa as a reversal agent of the direct thrombin inhibitor argatroban. Chirurgia 2011, 24, 283–286. [Google Scholar]
  101. Cho, A.E.; Jerguson, K.; Peterson, J.; Patel, D.V.; Saberi, A.A. Cost-effectiveness of Argatroban Versus Heparin Anticoagulation in Adult Extracorporeal Membrane Oxygenation Patients. Hosp. Pharm. 2019, 54, 276–281. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of anticoagulant agents, dosing, and targets of the anticoagulation.
Table 1. Summary of anticoagulant agents, dosing, and targets of the anticoagulation.
Anticoagulant AgentMechanism of ActionDosingTarget
Unfractionated heparinInhibits mainly thrombin and Xa by activating antithrombinVariable (start 10 IU/kg/h, titrated to reach the target) cont. inf.ACT 180–200 s/APTT ratio 1.6–1.8/anti-Xa 0.3–0.7
Low-molecular-weight heparinInhibits mainly Xa by activating antithrombinNo recommendation (mainly standard prophylactic s.c. dose)Unknown
FondaparinuxInhibits only Xa by activating antithrombinNo recommendation (mainly 2.5–7.5 mg/day s.c.)Unknown
BivalirudinReversibly binds to thrombin0.02–0.05 µg/kg/min (cont. inf.)APTT ratio 1.5–2.5
ArgatrobanReversibly binds to thrombin0.1–0.3 µg/kg/min (cont. inf.)APTT 50–70 s/ACT 150–230 s
cont. inf. = continuous infusion.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Šoltés, J.; Skribuckij, M.; Říha, H.; Lipš, M.; Michálek, P.; Balík, M.; Pořízka, M. Update on Anticoagulation Strategies in Patients with ECMO—A Narrative Review. J. Clin. Med. 2023, 12, 6067. https://doi.org/10.3390/jcm12186067

AMA Style

Šoltés J, Skribuckij M, Říha H, Lipš M, Michálek P, Balík M, Pořízka M. Update on Anticoagulation Strategies in Patients with ECMO—A Narrative Review. Journal of Clinical Medicine. 2023; 12(18):6067. https://doi.org/10.3390/jcm12186067

Chicago/Turabian Style

Šoltés, Ján, Michal Skribuckij, Hynek Říha, Michal Lipš, Pavel Michálek, Martin Balík, and Michal Pořízka. 2023. "Update on Anticoagulation Strategies in Patients with ECMO—A Narrative Review" Journal of Clinical Medicine 12, no. 18: 6067. https://doi.org/10.3390/jcm12186067

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop