Review Open Access
Copyright ©The Author(s) 2019. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Crit Care Med. Oct 16, 2019; 8(6): 87-98
Published online Oct 16, 2019. doi: 10.5492/wjccm.v8.i6.87
Anticoagulation with direct thrombin inhibitors during extracorporeal membrane oxygenation
Barry Burstein, Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN 55905, United States
Patrick M Wieruszewski, Yan-Jun Zhao, Department of Pharmacy, Mayo Clinic, Rochester, MN 55905, United States
Nathan Smischney, Department of Anesthesia, Mayo Clinic, Rochester, MN 55905, United States
ORCID number: Barry Burstein (0000-0003-0203-9294); Patrick M Wieruszewski (0000-0002-5871-5186); Yan-Jun Zhao (0000-0003-1938-9848); Nathan Smischney (0000-0003-1051-098X).
Author contributions: All authors equally contributed to this paper with conception and design of the study, literature review and analysis, drafting and critical revision and editing, and final approval of the final version.
Conflict-of-interest statement: No potential conflicts of interest to declare.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Nathan Smischney, MD, MSc, Assistant Professor, Department of Anesthesia, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, United States. smischney.nathan@mayo.edu
Telephone: +1-507-2554305 Fax: +1-507-2554267
Received: April 28, 2019
Peer-review started: May 9, 2019
First decision: August 2, 2019
Revised: August 13, 2019
Accepted: September 5, 2019
Article in press: September 5, 2019
Published online: October 16, 2019

Abstract

Use of extracorporeal membrane oxygenation to support patients with critical cardiorespiratory illness is increasing. Systemic anticoagulation is an essential element in the care of extracorporeal membrane oxygenation patients. While unfractionated heparin is the most commonly used agent, unfractionated heparin is associated with several unique complications that can be catastrophic in critically ill patients, including heparin-induced thrombocytopenia and acquired antithrombin deficiency. These complications can result in thrombotic events and subtherapeutic anticoagulation. Direct thrombin inhibitors (DTIs) are emerging as alternative anticoagulants in patients supported by extracorporeal membrane oxygenation. Increasing evidence supports DTIs use as safe and effective in extracorporeal membrane oxygenation patients with and without heparin-induced thrombocytopenia. This review outlines the pharmacology, dosing strategies and available protocols, monitoring parameters, and special use considerations for all available DTIs in extracorporeal membrane oxygenation patients. The advantages and disadvantages of DTIs in extracorporeal membrane oxygenation relative to unfractionated heparin will be described.

Key Words: Extracorporeal membrane oxygenation, Anticoagulants, Antithrombins, Bivalirudin, Argatroban, Heparin

Core tip: In contrast to unfractionated heparin, direct thrombin inhibitors are not associated with heparin-induced thrombocytopenia or acquired antithrombin deficiency. Direct thrombin inhibitors, specifically bivalirudin and argatroban, are equally safe and possibly more efficacious than unfractionated heparin. Dosage and monitoring parameters are easily manageable and more predictable than unfractionated heparin. As extracorporeal membrane oxygenation increases in use, direct thrombin inhibitors may potentially be considered as a primary anticoagulant in patients with or without complications of unfractionated heparin.



INTRODUCTION

Extracorporeal membrane oxygenation (ECMO) is increasingly used in patients with refractory respiratory or cardiogenic shock[1]. Patients can be supported in either a veno-venous (VV) or veno-arterial (VA) configuration. Systemic anticoagulation is necessary and crucial due to continuous contact between the patient’s blood and the foreign surfaces of all components of the ECMO circuit. This interaction triggers the coagulation cascade and can lead to pump or oxygenator thrombi, or fibrin stranding within the inflow cannula resulting in potentially devastating thromboembolic events[2]. The ideal anticoagulant has rapid onset and offset, is easily titratable based on available monitoring parameters, is reversible, and is not affected by organ dysfunction commonly seen in ECMO patients[3]. Intravenous unfractionated heparin (UFH) is the standard anticoagulant in most centers due to its availability, rapid onset of action, reversibility, cost profile, and familiarity among practitioners[4].

Complications related to UFH are common: bleeding, non-immune and immune heparin-induced thrombocytopenia (HIT), and heparin-resistance have all been described[5,6]. HIT is of particular concern, with mortality as high as 20%-30%[7]. The incidence of HIT in patients treated with UFH is approximately 2.6%, although this does not reflect the incidence in ECMO patients[8]. HIT is often suspected in ECMO patients; however available data are inconsistent, suggesting that the incidence ranges from less than 0.36% to 17%[9-12]. HIT reverses the anticoagulant effect of heparin and leads to massive platelet activation and thrombosis, which can be catastrophic[13]. All forms of heparin must be immediately discontinued once the diagnosis of HIT is suspected. In ECMO patients, the heparin-coated elements of the circuit must also be exchanged. UFH can also lead to acquired antithrombin deficiency which may result in heparin resistance and suboptimal anticoagulation[2]. This is of particular concern in patients receiving ECMO secondary to the inability to liberate from cardiopulmonary bypass due to high intraoperative heparin loads. Alternative anticoagulation strategies have been proposed. In non-ECMO patients with suspected or confirmed HIT, direct thrombin inhibitors (DTIs) are the primary alternative[13]. This article will review DTI anticoagulation strategies in adult ECMO patients, as well as available safety and outcome data. Heparin alternatives in pediatric ECMO patients have previously been reviewed[14].

PHARMACOLOGY OF DIRECT THROMBIN INHIBITORS

Thrombin is a serine protease that plays a crucial role in the coagulation cascade and the generation and stabilization of clot. Upon activation, thrombin facilitates the formation of insoluble fibrin from soluble fibrinogen[15,16]. Thrombin contains three binding sites that are essential to its coagulant-and thereby anticoagulant-effects, including the catalytic site, exosite-1, and exosite-2.

The DTIs obtain their name from their direct binding to thrombin to exert anticoagulant effects. This is in contrast to UFH and low molecular weight heparins, which are indirect thrombin inhibitors. These indirect inhibitors bind to antithrombin, a hepatically-synthesized glycoprotein, forming a heparin-antithrombin complex which subsequently binds to exosite-2 on thrombin and blocks the catalytic site[16,17]. Because of this binding, indirect thrombin inhibitors only exert effects on circulating thrombin, as the catalytic site of fibrin-bound thrombin is occupied in preformed clot[17]. DTIs, on the other hand, bind directly to either the catalytic site or both the catalytic site and exosite-1 on thrombin, depending on the valency of the agent, in the absence of antithrombin[15,17]. Because of this, a major pharmacologic advantage over indirect thrombin inhibitors is the ability of DTIs to bind both circulating and fibrin-bound thrombin. DTIs for clinical use vary based on their valency, modality of binding (reversible vs. irreversible), and pharmacokinetic profile (Table 1).

Table 1 Pharmacology of direct thrombin inhibitors.
ValenceThrombin bindingOnset of actionHalf-lifeProtein bindingMetabolismSpecial considerations
BivalirudinBivalentReversible2-4 min25 minNoneSerum proteasesAvoid during low-flow states Removed by non-diffusive dialysis modalities Dose reductions necessary in renal dysfunction
ArgatrobanUnivalentReversible30 min45 min20% albumin 34% alpha-acid glycoproteinHepatic (hydroxylation, aromatization)Dose reductions necessary in hepatic dysfunction
ADVANTAGES OF DIRECT THROMBIN INHIBITORS

Anticoagulation is an essential element of ECMO support. While UFH remains the standard at most ECMO centers, DTIs are emerging as a reasonable and safe alternative due to several advantages when compared to UFH. The advantages of DTIs over UFH are: (1) Direct binding of both circulating and clot-bound thrombin, which results in increased efficacy relative to UFH[17]; (2) Anticoagulant effect that is independent of antithrombin, allowing for more consistent and predictable effect without concern for antithrombin depletion[15,17]; (3) Avoidance of HIT, as thrombocytopenia is common among ECMO patients and the diagnosis of HIT in this setting is challenged by the multitude of factors that can precipitate a drop in platelet count. Healthcare teams must maintain a low threshold of concern when HIT is suspected. DTIs appear to be at least as safe as UFH with no evidence of increased bleeding or thrombosis, and evidence suggests that patients who receive DTIs are more often in therapeutic targeted range of anticoagulation. While DTIs are an appropriate choice in the setting of suspected or confirmed HIT, they may be a reasonable option as first-line anticoagulation for better efficacy in maintaining ECMO circuit patency and to avoid concerns for HIT altogether. Although DTIs do not have target-specific antidote, their half-life is very short and the anticoagulant effect tapers off rapidly despite the widely prevalent end organ dysfunction in ECMO patients. ECMO patients rarely require complete reversal of anticoagulation given the high risk of thrombosis.

AVAILABLE AGENTS
Bivalirudin

Bivalirudin is a synthetic, bivalent DTI, which binds directly to both the catalytic site and exosite-1 on thrombin in a reversible fashion[16] (Table 1). Bivalirudin dissociates from the catalytic site following proteolytic cleavage, reconstituting thrombin’s ability to facilitate fibrin formation[18]. This may be problematic during states of blood stasis as will be discussed. It is administered by intravenous infusion, has an onset of action within minutes, and has a half-life of 25 min in patients with normal renal function[18]. Bivalirudin has a low volume of distribution and is therefore widely distributed in plasma and has negligible protein binding.

Bivalirudin is available as an adjunct to anticoagulation therapy during percutaneous coronary interventions, as well as the primary anticoagulant during coronary artery bypass grafting in patients with HIT[19]. Use in ECMO, with or without HIT, is off-label. The available literature for use in adults supported by ECMO is derived from retrospective studies, a single case control trial, and multiple case reports[20-26] (Table 2). Two studies compared bivalirudin to a matched control group who received UFH[20,25]. Four studies described patients with suspected or confirmed HIT due to previous UFH use[21,23,24,26]; the remaining studies used bivalirudin as the initial anticoagulation strategy[20,22,24,25]. Both VV and VA configurations are described. There is one published protocol describing initial bivalirudin dosing and subsequent dose adjustments[22].

Table 2 Summary of studies reporting on argatroban in adult patients supported with extracorporeal membrane oxygenation.
First author, YearStudy typePopulationCircuit (VA/VV)CRRTBolus dose (mg/kg)Initial infusion (mg/kg/min)MonitoringDurationMajor bleedingThrombosisOther adverse eventsOutcome
Jyoti et al[21], 2013Case report54M ARDS, HITVVNoNA0.6ACT 200-220 s aPTT 60-80 s552 h (23 d)NANANA
Pappalardo et al[23], 2009Case report71F post-cardiotomy, HITVANo0.50.5ACT 180-220 s6 dNAVentricular fibrillation due to LA thrombus, suspected to be due to heparin in tubing with residual HIT. BIV dosing increasedNADecannulated and discharged in stable condition
Pieri et al[24], 2013Case controln = 10 (4 HIT)VV (n = 5) VA (n = 5)n = 7 (70%)N/A0.025aPTT 45-60 s8 d (range 6-23)n = 3 (30%)n = 1 (10%)No difference in bleeding or thrombosis compared to UFH patients Less dose corrections than UFH Less supra-therapeutic aPTTs than UFHn = 4 (40%) died
Berei et al[20], 2018Retrospectiven = 44 CS (n = 37) Sepsis (n = 11) Respiratory (n = 3) Mixed (n = 4)VA (n = 26) VV (n = 2)n = 17 (39%)UFH 80 units/kg at cannulation No BIV bolus0.04aPTT 45-65 s (low intensity) or 60-80 s (high intensity)156.9 h (mean)n = 20 (45.5%)n = 10 (22.7%)Increased flow rates during first 96 h High intensity BIV had more TTR with no difference in outcomesNo difference in death at 30 d between BIV and UFH (36% vs 32%)
Netley et al[22], 2017Retrospectiven = 11 ARDS (n = 8) ECLS (n = 3)VA (n = 4) VV (n = 7)n = 4 (36%)NA2.5aPTT 40-60 s, 50-70 s, or 60-80 sMean 9.9 d (range 4-22)n = 8 (72.7%)n = 2 (18.2%), both after hospital dischargeNAn = 5 (45%) died after withdrawal of care n = 6 (55%) discharged from hospital
Ranucci et al[25], 2011Retrospectiven = 8, post-cardiotomyVANANA0.03-0.05 ½ dose if reduced CrClACT 160-180 s or aPTT 50-80 s or TEG r 12-30 min39-262 hNANoneBleeding not reported, but less average blood loss (mL/kg/d) in BIV patientsn = 2 (25%) survived n = 2 (25%) dead on ECMO n = 4 (50%) weaned but died
Walker et al[26], 2019Retrospectiven = 14 ARDS (n = 12) Post-cardiotomy (n =2) HIT (n = 11/13)VV (n = 11) VA (n = 3)n = 6 (43%)0.2 (n = 1, others NA)0.04-0.26aPTT 1.5-2.5 × baselineMedian 5.2 d (range 0.9-28.4 d)n = 4 (29%)Circuit clotting (n = 5, 36%)Infusion held during major bleeding episodes with no need for correction Higher infusion rates noted with CRRTn = 9 (64%) decannulated n = 7 (50%) survived to discharge

In most reported studies of bivalirudin, initial bolus doses were administered followed by a weight-based infusion. The described dosing is heterogeneous; bolus dosing ranges from 0.04 mg/kg to 2.5 mg/kg. In reports without bolus dosing there was no sign of increased thromboembolic risk during the time until therapeutic anticoagulation was achieved[21,24-26]. The maintenance infusion was adjusted based on monitoring parameters, and doses ranged from 0.025 mg/kg/h to 2.5 mg/kg/h. In studies where average infusion rate is described, the dose ranges from 0.05 mg/kg/h to 0.26 mg/kg/h to maintain therapeutic targets[21,23,26]. When compared to patients receiving UFH infusions, patients treated with bivalirudin are more often in therapeutic range[20,24].

Renal dysfunction: Bivalirudin is metabolized in the plasma by proteolytic cleavage and has a prolonged half-life in the setting of renal dysfunction[27]. Renal dysfunction is therefore an important consideration when initiating and adjusting bivalirudin infusions. Initial dosing, as well as adjustments in the maintenance infusion rate, may vary, and over-anticoagulation is an important concern in these patients. Limited studies with established protocols describe lower initial bolus doses in patients with renal dysfunction[25], while others use the same bolus dose as patients without renal dysfunction while adjusting subsequent maintenance infusion rates[22]. Ranucci et al[25] administered half-dose initially in patients with renal dysfunction, followed by the regular dose adjustment protocol. The protocol by Netley et al[22] stratified patients based on creatinine clearance above 30 mL/min, between 10-29 mL/min, and less than 10 mL/min or requiring intermittent hemodialysis. The initial bolus dose was standardized for all patients regardless of creatinine clearance, and subsequent dose adjustments were limited as the severity of renal dysfunction progressed.

Continuous renal replacement therapy (CRRT) utilizing convective (hemofiltration) or combination convective/diffusive (hemodiafiltration) modes has been found to modestly remove bivalirudin, but purely diffusive modalities are not expected to be major determinants of clearance due to bivalirudin’s larger molecular size (1980 Da)[28]. There are multiple reports of successful anticoagulation with bivalirudin while supporting patients with CRRT[20,22,24]. Walker et al[26] described moderately reduced dose requirements in patients on CRRT, although the doses were higher than those with renal dysfunction who were not receiving CRRT. While it would be expected that patients with renal dysfunction require less bivalirudin to maintain a similar anticoagulation profile, retrospective data suggest that these patients may require higher doses[24]. The reasons for this finding are unclear. Given the tendency to reduce initial and/or maintenance bivalirudin doses in patients with renal dysfunction, careful attention should be paid to avoid under-anticoagulation which may result in thrombotic events. The currently available reports do not signal an increased risk of thrombosis in patients with renal dysfunction, although the number of studied patients is limited.

Hepatic dysfunction: There is no bivalirudin dose adjustment necessary in patients with hepatic dysfunction, although there are no specific reports of outcomes in patients with hepatic disease.

Monitoring: Bivalirudin prolongs the activated clotting time (ACT), activated partial thromboplastic time (aPTT), thrombin time, prothrombin time, and international normalized ratio[19]. Ecarin clotting time and chromogenic anti-factor IIa assay are the most reliable methods for DTI monitoring; however, they are not readily available in clinical laboratories. The aPTT testing is the most commonly used and well validated assay to monitor DTIs. The manufacturer recommends monitoring with ACT in certain conditions, namely HIT, with an ACT target greater than 225 s. No specific recommendation from the manufacturer is available in ECMO patients as this is an off-label indication. Most reviewed studies, including available protocols, used aPTT as a monitoring parameter. Netley et al[22] used aPTT alone with adjustable target based on physician preference; 40-60 s, 50-70 s, or 60-80 s were all described, with adjustments in bivalirudin doses based on the difference between the measured aPTT and the target aPTT. The aPTT was measured 2 h after the initial dose and then every 4 h afterwards. Ranucci et al[25] adjusted bivalirudin dosing based on ACT primarily (target 160-180 s), followed by aPTT (target 50-80 s) and then kaolin-activated thromboelastography (TEG, TEG 5000; Haemonetics Corp, Braintree, MA, United States) r time (target 12-30 min). Parameters were checked every 4, 12, and 8 h, respectively. In the retrospective study by Berei et al[20], aPTT targets between 45-65 s (low intensity) and 60-80 s (high intensity) were chosen based on physician preference. Given the wide variability of monitoring modalities used and goal ranges reported in the literature, and the possibility of DTI resistance and unreliable aPTT monitoring at very high DTI doses, it is of utmost importance to use clinical indicators including ECMO circuit patency and potential thrombotic and bleeding complications as ultimate guidance for anticoagulation titration rather than solely relying on numbers of certain lab values.

Safety and outcomes: Bivalirudin appears to be a safe anticoagulation strategy for patients supported by ECMO, with no overall evidence of increased bleeding or thrombotic complications relative to UFH[20,24,25]. One study demonstrated an increased incidence of bleeding events in patients treated with bivalirudin compared to those receiving UFH, without meeting statistical significance[20]. Walker et al[26] described 4 of 14 patients who required reduction in aPTT targets or complete suspension of bivalirudin due to bleeding, although this rate is similar to reported bleeding rates in UFH patients. Some reports suggest a lower risk of thrombosis and vascular complications, as well as decreased transfusion requirements relative to UFH[20,25], although cases requiring circuit exchange due to thrombosis have been reported[26]. Reports of patients who required ECMO support up to 23 d suggest that outcomes with prolonged bivalirudin use are similar to UFH[21,24].

Argatroban

Argatroban is a synthetic, univalent DTI, and thereby binds directly to the catalytic site on thrombin in a reversible fashion[16]. Compared to the other DTIs, it has a relatively small molecular size (527 Da). It is administered by intravenous infusion, has an onset of action within 30 minutes, and has a half-life of 45 min in patients with normal hepatic function (Table 1). Of the DTIs, argatroban has the greatest serum protein binding, with 20% to albumin and 34% to alpha1-acid glycoprotein. Lidocaine-an antiarrhythmic used on occasion in cardiothoracic surgical patients-may decrease argatroban concentrations up to 20% due to its high affinity for alpha1-acid glycoprotein[29].

Argatroban is available as therapeutic anticoagulation for confirmed or suspected HIT in patients who develop thrombocytopenia while on ECMO support. There are limited data available regarding argatroban use in ECMO; most are case reports and case series (Table 3)[30-37]. There is one retrospective study of 39 adult patients treated with argatroban, which also included a small proportion of patients supported with pumpless extracorporeal lung assist[38]. Patients supported with both VV and VA systems are described, and all are limited to patient population with suspected or confirmed HIT. There are no reported cases with argatroban as a first-line anticoagulant in ECMO patients.

Table 3 Summary of studies reporting on argatroban in adult patients supported with extracorporeal membrane oxygenation.
First author, YearStudy typePopulationCircuit (VA/VV)CRRTBolus doseInitial infusionMonitoringDurationMajorbleedingThrombosisOther adverse eventsOutcome
Sin et al[37], 2017Case report27M ARDS, HITVVYesNA0.2 µg/kg/minaPTT 50-60 s60 dHemothorax developed while on heparin, resolved on ARGA day 27NoneTransient elevations in liver enzymes, no clinical consequencePatient transferred for lung transplantation
Ratzlaff et al[35], 2016Case report58M ARDS, HITVVNoNA0.1-0.3 µg/kg/minaPTT 60-90 s11 dNoneNoneNAWithdrawal of care after 28 d of ECMO support
Johnston et al[34], 2002Case report32M CS, HITVANo10 mg2 µg/kg/hACT 200-400 s aPTT 80-90 s7 dNoneNANADecannulated on ECMO day 10
Dolch et al[32], 2010Case report40M ARDS, HITVVNoNA0.35 µg/kg/minaPTT 45-60 s108 dMajor bleeding after lung transplant (ECMO day 114) – ARGA heldNAHepatic failure post-transplant Infusion reduced to 0.02 µg/kg/minPatient underwent lung transplant on ECMO day 114, complicated by graft failure Died on post-operative day 17 (multi-organ failure)
Fernandes et al[33], 2019Case report44M CS, HITVAYesNA1.5 mg/haPTT 60-70 s20 dMediastinal bleeding due to pulmonary edema Massive intraoperative hemorrhage during LVAD insertion, DICLV and RV thrombus during intraoperative DICNASurvived to discharge
Cornell et al[31], 2007Case seriesn = 4 with HIT ARDS (n = 3) CS (n = 1)VA (n = 2) VV (n = 2)NoNA0.2-2.0 µg/kg/minACT 210-230 s88-184 hMajor bleeding (n = 2)NANASurvival to discharge (n = 2, 50%) Death (n = 2, 50%)
Beiderlinden et al[30], 2007Case seriesn = 9 with ARDS, HITVVn = 8NA2.0 µg/kg/min (n = 1) 0.2 µg/kg/min (n = 8)aPTT 50-60 s4 ± 1 d (mean)Major bleeding (n = 1) in patient who received higher initial infusion doseNoneNASurvived (n = 6) Died (n = 3)
Rougé et al[36], 2017Case series49M CS, HIT 69M ARDS, HITVAn = 1NA0.2 µg/kg/min 1 µg/kg/minaPTT 1.5-3.0 × baseline10 d 8 dNACircuit clotting (n = 1)ALF requiring dose reductionSurvived (n = 1) Decannulated, but died prior to discharge (n = 1)
Menk et al[38], 2017Retrospectiven = 34 ARDS, HIT or heparin resistanceVV (n = 24) pECLA (n = 9)NANA0.3 µg/kg/minaPTT 50-75 s265 h (131-460)n = 11—no differences compared to matched UFH cohortn = 6—no differences compared to matched UFH cohortNAn = 21 (54%) died

Dosing: Only one patient in the reviewed literature received the manufacturer-recommended initial dose of 2 µg/kg/min. The patient subsequently suffered major bleeding complications and required a rapid dose reduction[30]. In all other reported cases, patients subsequently received doses at a rate approximately 10% the manufacturer recommended dose, with most initial doses ranging from 0.1-0.3 µg/kg/min. This is consistent with literature from other critically ill populations which suggests that an initial dose of 0.2 µg/kg/min results in adequate dosing based on aPTT measurement, without excessive bleeding or thrombosis[39].

Renal dysfunction: No dose adjustment is required for patients with renal dysfunction, with or without renal replacement therapy, which is a major advantage relative to other agents. There are several reports of successful clinical outcomes with argatroban in patients who require CRRT while supported by ECMO[30,33,37].

Hepatic dysfunction: Argatroban is metabolized hepatically by hydroxylation and aromatization, and therefore has a prolonged half-life of up to four times normal in the setting of liver dysfunction[40]. While CYP3A4 and CYP3A5 provide a minor metabolic pathway, co-administration of inhibitors or inducers of these enzymes do not result in significant changes in argatroban concentrations[41].

Argatroban is not contraindicated in patients with hepatic dysfunction; however significant dose reductions may be required. There are few described cases of argatroban use in patients with liver dysfunction. One reported case of a patient with acute respiratory distress syndrome who underwent lung transplantation complicated by postoperative hepatic dysfunction describes a maintenance dose of 0.02 µg/kg/min in order to achieve the target aPTT[32]. The patient did not suffer from any adverse bleeding or thrombotic events related to the relatively low dose requirement.

Monitoring: The majority of reported cases and the single retrospective study used aPTT as the target for dose adjustment[30,32,33,35,37,38]. Some cases reported use of ACT, either alone or in conjunction with aPTT, as the therapeutic monitoring parameter[31,34]. In cases where aPTT was used, there was no standardized target; most reported a goal of 50-70 s. Of note, Menk et al[38] found that bleeding events occurred when the maximum aPTT was above 50-60 s, and two thirds of bleeding events occurred when the maximum aPTT was above 75 s. Conversely, transient aPTT values below 50 s did not signal an increase in thromboembolic events. The authors of that study therefore recommended strict aPTT monitoring with a target of approximately 50 s. This is consistent with published guidelines which suggest an aPTT goal range of 1.5-2.5 times baseline[38]. ACT monitoring, although less frequently used, varies substantially with reported targets between 200 and 400 s. As discussed previously, clinical end points remain the ultimate guidance for adequacy of anticoagulation in these critically ill and complex ECMO patient populations.

Safety and outcomes: There are many reported cases of successful clinical outcomes using argatroban in ECMO patients with suspected or confirmed HIT. In cases in which patients did not survive, none were reported to be directly associated with argatroban use. The reported duration of support is as long as 95 d. The rate of bleeding or thrombosis is low. In the retrospective study of 39 ECMO patients treated with argatroban, the rate of major bleeding or thrombosis was comparable to patients who received heparin[2,38]. There are few reported major bleeding events with argatroban, many of which were related to surgical interventions. Bleeding at cannulation sites is reported, and blood transfusions were frequent, however the reported rate is similar to that seen with heparin[38].

Other agents

Desirudin and lepirudin are recombinant-DNA forms of the naturally occurring peptide, hirudin, present in the salivary glands of leeches. These recombinant hirudins are bivalent DTIs and thereby bind directly to both the catalytic and exosite-1 of thrombin in an irreversible fashion[42]. Because they are composed of non-human proteins, anti-hirudin antibodies may be formed leading to potential immunologic reactions including anaphylaxis[42]. The clinical use of recombinant hirudins during ECMO is limited by these factors and the more favorable pharmacokinetic profiles of the newer synthetic DTIs. Only lepirudin has been reported in adult patients requiring ECMO support. The single case report noted a successful outcome[43]. Lepirudin was withdrawn from market in 2012 and is no longer in production.

Dabigatran etexilate is an oral, synthetic, peptide-like DTI[44]. The onset of action is approximately 1 h and the half-life is 12 to 17 h. Renal excretion is the primary determinant of dabigatran removal, with up to 80% in the urine, and therefore the half-life is prolonged in the setting of renal dysfunction. Dabigatran is a substrate of p-glycoprotein and is therefore prone to a large number of clinically relevant drug interactions with p-glycoprotein inhibitors or inducers. Because of all of these reasons, there is no role for dabigatran in anticoagulation during ECMO.

LIMITATIONS AND SPECIAL USE CONSIDERATIONS
Bleeding

Bleeding is a common cause of morbidity and mortality among ECMO patients, regardless of anticoagulation strategy[2]. There are no formal recommendations for anticoagulation management when bleeding is encountered while on ECMO. For patients receiving DTI therapy, holding DTIs temporarily or short term can be considered. As previously mentioned, there is no specific antidote for DTIs; however, the short half-life of available agents results in rapid offset of anticoagulant effect when the infusion is held or decreased. Once surgical bleeding has been excluded, treating teams can consider performing TEG to further assess the cause of bleeding and transfuse blood products as needed. In patients with hyperfibrinolysis, successful TEG-guided use of tranexamic acid has been described in patients on ECMO and DTIs[45]. Once hemostasis has been achieved, DTI infusions can be restarted at a lower infusion rate, targeting a lower anticoagulation goal based on aPTT or ACT. In the protocol published by Netley et al[22], significant bleeding triggered the reduction of the bivalirudin infusion rate, targeting the lower limit of the therapeutic range (i.e., aPTT 40 s). Prothrombin complex concentrates may be used in life-threatening situations[2]. As a general rule, ECMO flow should be increased whenever anticoagulation is reduced or suspended in order to minimize the risk of thrombus formation.

Prothrombin time interference

DTIs commonly prolong the prothrombin time and international normalized ratio in a dose-dependent manner, which may confound monitoring of warfarin in clinical practice. The derangement in these values may depend on the specific assay used[46]. However, it typically does not interfere with ECMO anticoagulation as we primarily use aPTT/ACT. Factor Xa testing may be considered during DTI transitions to warfarin.

Low circuit flow states

While argatroban undergoes mostly hepatic metabolism, bivalirudin is primary metabolized by proteolytic enzymes which rapidly cleave the molecule. This results in the short half-life that has been described. However, instances where blood is stagnant may induce thrombosis due to rapid local cleavage of bivalirudin[47]. Although ECMO is a continuous circuit without any “low-flow” chambers, in cases of cardiac dysfunction the cardiac chambers may act as a source of stagnation. This may particularly be realized in patients who are undergoing active ECMO “wean” trials where circuit flows are down-titrated to assess ability to decannulate. In addition, any setting wherein the native myocardium experiences low pulsatility and allows blood to pool in the ventricles is of very high concern. This may result in localized thrombosis despite adequate bivalirudin dosing. This can be particularly problematic in post-cardiotomy ECMO patients with prosthetic heart valves as valve thrombosis may lead to significant morbidity and mortality. It has been suggested that these circumstances may be avoided by minimizing intracardiac blood flow in cases of cardiogenic shock or avoid low-pulsatility states, and by using UFH as alternative anticoagulation in cases where intracardiac spontaneous echo contrast, or “smoke effect,” is noted, or low-flow states are suspected[47]. During EMCO weaning and trial-off period, especially in setting of VA ECMO wean and if the duration of the trial-off is prolonged, preemptive heparin administration is essential to minimize thrombus formation.

Direct thrombin inhibitor resistance and high-dose response

Resistance to UFH has been reported both in ECMO and non-ECMO cases, with multiple potential explanations[48,49]. Less is known regarding DTI resistance, although cases have been reported using both bivalirudin and argatroban[50-53]. Very few described cases included patients requiring extracorporeal support. In cases of DTI resistance, increasing doses are required to achieve the target aPTT. This has been reported with initial dosing of DTI, although there are also delayed presentations with therapeutic levels initially followed by progressive unexplained dose increases to maintain therapeutic aPTT levels[26,50,53]. The mechanism of DTI resistance is unclear but may be associated with elevated factor VIII and fibrinogen, which has been reported in some cases[52,53]. Notably, these patients ultimately require very high doses of DTI to achieve aPTT targets, often well above the maximum recommended dose. It has been suggested that increases in DTI dosage may result in less aPTT prolongation at high doses than at low doses, which may explain why patients on high doses of DTI require greater than expected dose increases[51]. Conversely, the increase in the international normalized ratio may be more pronounced at high DTI doses[51,53]. Early recognition and rapid titration are essential, as there are multiple reports of clinical thrombosis due to subtherapeutic anticoagulation during delayed titration to target aPTT levels[53]. This point highlights the importance of applying clinical end points (circuit patency, bleeding vs. thrombosis) as ultimate guidance for ECMO anticoagulation management. In cases of DTI resistance, alternative monitoring parameters have been proposed as aPTT may be unreliable. These include ACT, thrombin time, or direct drug level measurement[53,54]. The therapeutic targets for these parameters, however, are variably defined.

CONCLUSION

Systemic anticoagulation with DTIs in ECMO patients is a feasible and safe alternative, with several advantages over UFH. The primary indication for DTIs is in cases of suspected or confirmed HIT, however reports suggest that DTIs may be effective as an initial anticoagulation strategy for all ECMO patients. Multiple dosing and monitoring protocols have been proposed for both bivalirudin and argatroban, and further prospective trials should determine the optimal pathway to safe, effective anticoagulation in this critically ill population.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Critical Care Medicine

Country of origin: United States

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Drabek T, Qi XS, Willms DC S-Editor: Yan JP L-Editor: A E-Editor: Liu MY

References
1.  Bartlett RH, Gattinoni L. Current status of extracorporeal life support (ECMO) for cardiopulmonary failure. Minerva Anestesiol. 2010;76:534-540.  [PubMed]  [DOI]  [Cited in This Article: ]
2.  The Extracorporeal Life Support Organization (ELSO). ELSO Anticoagulation Guideline 2014 [cited April 11, 2019].  Available from: https://www.elso.org/Portals/0/Files/elsoanticoagulationguideline8-2014-table-contents.pdf.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Askenazi DJ, Selewski DT, Paden ML, Cooper DS, Bridges BC, Zappitelli M, Fleming GM. Renal replacement therapy in critically ill patients receiving extracorporeal membrane oxygenation. Clin J Am Soc Nephrol. 2012;7:1328-1336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 162]  [Article Influence: 13.5]  [Reference Citation Analysis (0)]
4.  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:e77-e84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 266]  [Cited by in F6Publishing: 269]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
5.  Rastan AJ, Dege A, Mohr M, Doll N, Falk V, Walther T, Mohr FW. Early and late outcomes of 517 consecutive adult patients treated with extracorporeal membrane oxygenation for refractory postcardiotomy cardiogenic shock. J Thorac Cardiovasc Surg. 2010;139:302-311, 311.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 386]  [Cited by in F6Publishing: 420]  [Article Influence: 30.0]  [Reference Citation Analysis (0)]
6.  Rastan AJ, Lachmann N, Walther T, Doll N, Gradistanac T, Gommert JF, Lehmann S, Wittekind C, Mohr FW. Autopsy findings in patients on postcardiotomy extracorporeal membrane oxygenation (ECMO). Int J Artif Organs. 2006;29:1121-1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 127]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
7.  Salter BS, Weiner MM, Trinh MA, Heller J, Evans AS, Adams DH, Fischer GW. Heparin-Induced Thrombocytopenia: A Comprehensive Clinical Review. J Am Coll Cardiol. 2016;67:2519-2532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 114]  [Article Influence: 16.3]  [Reference Citation Analysis (0)]
8.  Martel N, Lee J, Wells PS. Risk for heparin-induced thrombocytopenia with unfractionated and low-molecular-weight heparin thromboprophylaxis: A meta-analysis. Blood. 2005;106:2710-2715.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 567]  [Cited by in F6Publishing: 516]  [Article Influence: 27.2]  [Reference Citation Analysis (0)]
9.  Kimmoun A, Oulehri W, Sonneville R, Grisot PH, Zogheib E, Amour J, Aissaoui N, Megarbane B, Mongardon N, Renou A, Schmidt M, Besnier E, Delmas C, Dessertaine G, Guidon C, Nesseler N, Labro G, Rozec B, Pierrot M, Helms J, Bougon D, Chardonnal L, Medard A, Ouattara A, Girerd N, Lamiral Z, Borie M, Ajzenberg N, Levy B. Prevalence and outcome of heparin-induced thrombocytopenia diagnosed under veno-arterial extracorporeal membrane oxygenation: A retrospective nationwide study. Intensive Care Med. 2018;44:1460-1469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
10.  Choi JH, Luc JGY, Weber MP, Reddy HG, Maynes EJ, Deb AK, Samuels LE, Morris RJ, Massey HT, Loforte A, Tchantchaleishvili V. Heparin-induced thrombocytopenia during extracorporeal life support: Incidence, management and outcomes. Ann Cardiothorac Surg. 2019;8:19-31.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 28]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
11.  Glick D, Dzierba AL, Abrams D, Muir J, Eisenberger A, Diuguid D, Abel E, Agerstrand C, Bacchetta M, Brodie D. Clinically suspected heparin-induced thrombocytopenia during extracorporeal membrane oxygenation. J Crit Care. 2015;30:1190-1194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 48]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
12.  Warkentin TE, Greinacher A, Koster A. Heparin-induced thrombocytopenia in patients with ventricular assist devices: Are new prevention strategies required? Ann Thorac Surg. 2009;87:1633-1640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 58]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
13.  Arepally GM. Heparin-induced thrombocytopenia. Blood. 2017;129:2864-2872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 240]  [Cited by in F6Publishing: 261]  [Article Influence: 37.3]  [Reference Citation Analysis (0)]
14.  Pollak U. Heparin-induced thrombocytopenia complicating extracorporeal membrane oxygenation support in pediatric patients: Review of the literature and alternative anticoagulants. Perfusion. 2018;33:7-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
15.  Gurm HS, Bhatt DL. Thrombin, an ideal target for pharmacological inhibition: A review of direct thrombin inhibitors. Am Heart J. 2005;149:S43-S53.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
16.  White CM. Thrombin-directed inhibitors: Pharmacology and clinical use. Am Heart J. 2005;149:S54-S60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 29]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
17.  Di Nisio M, Middeldorp S, Büller HR. Direct thrombin inhibitors. N Engl J Med. 2005;353:1028-1040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 471]  [Cited by in F6Publishing: 404]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
18.  Weitz JI, Bates ER. Direct thrombin inhibitors in cardiac disease. Cardiovasc Toxicol. 2003;3:13-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 22]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
19.  Hospira Inc. Product Information: Bivalirudin intravenous injection solution, bivalirudin intravenous injection solution. Lake Forest, IL, United Sates, 2015. .  [PubMed]  [DOI]  [Cited in This Article: ]
20.  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;64:623-629.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 69]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
21.  Jyoti A, Maheshwari A, Daniel E, Motihar A, Bhathiwal RS, Sharma D. Bivalirudin in venovenous extracorporeal membrane oxygenation. J Extra Corpor Technol. 2014;46:94-97.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Netley J, Roy J, Greenlee J, Hart S, Todt M, Statz B. Bivalirudin Anticoagulation Dosing Protocol for Extracorporeal Membrane Oxygenation: A Retrospective Review. J Extra Corpor Technol. 2018;50:161-166.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  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;24:135-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 50]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
24.  Pieri M, Agracheva N, Bonaveglio E, Greco T, De Bonis M, Covello RD, 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 118]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
25.  Ranucci M, Ballotta A, Kandil H, Isgrò G, Carlucci C, Baryshnikova E, Pistuddi V; Surgical and Clinical Outcome Research Group. Bivalirudin-based versus conventional heparin anticoagulation for postcardiotomy extracorporeal membrane oxygenation. Crit Care. 2011;15:R275.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 166]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
26.  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;65:134-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
27.  Robson R, White H, Aylward P, Frampton C. Bivalirudin pharmacokinetics and pharmacodynamics: Effect of renal function, dose, and gender. Clin Pharmacol Ther. 2002;71:433-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 130]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
28.  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;45:1185-1192.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 40]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
29.  Kondo LM, Wittkowsky AK, Wiggins BS. Argatroban for prevention and treatment of thromboembolism in heparin-induced thrombocytopenia. Ann Pharmacother. 2001;35:440-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
30.  Beiderlinden M, Treschan T, Görlinger K, Peters J. Argatroban in extracorporeal membrane oxygenation. Artif Organs. 2007;31:461-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 87]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
31.  Cornell T, Wyrick P, Fleming G, Pasko D, Han Y, Custer J, Haft J, Annich G. A case series describing the use of argatroban in patients on extracorporeal circulation. ASAIO J. 2007;53:460-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
32.  Dolch ME, Frey L, Hatz R, Uberfuhr PA, 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. 2010;8:329-332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.1]  [Reference Citation Analysis (0)]
33.  Fernandes P, O'Neil M, Del Valle S, Cave A, Nagpal D. A 24-hour perioperative case study on argatroban use for left ventricle assist device insertion during cardiopulmonary bypass and veno-arterial extracorporeal membrane oxygenation. Perfusion. 2019;34:337-344.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Johnston N, Wait M, Huber L. Argatroban in adult extracorporeal membrane oxygenation. J Extra Corpor Technol. 2002;34:281-284.  [PubMed]  [DOI]  [Cited in This Article: ]
35.  Ratzlaff RA, Ripoll JG, Kassab LL, Diaz-Gomez JL. Acute oxygenator failure: A new presentation of heparin-induced thrombocytopenia in a patient undergoing venovenous extracorporeal membrane oxygenation support. BMJ Case Rep. 2016;2016:pii: bcr2016218179.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
36.  Rougé A, Pelen F, Durand M, Schwebel C. Argatroban for an alternative anticoagulant in HIT during ECMO. J Intensive Care. 2017;5:39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
37.  Sin JH, Lopez ND. Argatroban for Heparin-Induced Thrombocytopenia during Venovenous Extracorporeal Membrane Oxygenation with Continuous Venovenous Hemofiltration. J Extra Corpor Technol. 2017;49:115-120.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Menk M, Briem P, Weiss B, Gassner M, Schwaiberger D, Goldmann A, Pille C, Weber-Carstens S. Efficacy and safety of argatroban in patients with acute respiratory distress syndrome and extracorporeal lung support. Ann Intensive Care. 2017;7:82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
39.  Beiderlinden M, Treschan TA, Görlinger K, Peters J. Argatroban anticoagulation in critically ill patients. Ann Pharmacother. 2007;41:749-754.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 83]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
40.  Swan SK, Hursting MJ. The pharmacokinetics and pharmacodynamics of argatroban: Effects of age, gender, and hepatic or renal dysfunction. Pharmacotherapy. 2000;20:318-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 267]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
41.  Tran JQ, Di Cicco RA, Sheth SB, Tucci M, Peng L, Jorkasky DK, Hursting MJ, Benincosa LJ. Assessment of the potential pharmacokinetic and pharmacodynamic interactions between erythromycin and argatroban. J Clin Pharmacol. 1999;39:513-519.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Schaden E, Kozek-Langenecker SA. Direct thrombin inhibitors: Pharmacology and application in intensive care medicine. Intensive Care Med. 2010;36:1127-1137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 23]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
43.  Balasubramanian SK, Tiruvoipati R, Chatterjee S, Sosnowski A, Firmin RK. Extracorporeal membrane oxygenation with lepirudin anticoagulation for Wegener's granulomatosis with heparin-induced thrombocytopenia. ASAIO J. 2005;51:477-479.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
44.  Lee CJ, Ansell JE. Direct thrombin inhibitors. Br J Clin Pharmacol. 2011;72:581-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 163]  [Article Influence: 13.6]  [Reference Citation Analysis (0)]
45.  Seelhammer TG, Mangla J, Demirci O. The Use of Thromboelastography to Titrate Tranexamic Acid Therapy for Abatement of Lysis-Induced Hemorrhagic Complications During Venoarterial Extracorporeal Membrane Oxygenation. J Cardiothorac Vasc Anesth. 2019;33:1059-1062.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
46.  Gosselin RC, Dager WE, King JH, Janatpour K, Mahackian K, Larkin EC, Owings JT. Effect of direct thrombin inhibitors, bivalirudin, lepirudin, and argatroban, on prothrombin time and INR values. Am J Clin Pathol. 2004;121:593-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
47.  Ranucci M. Bivalirudin and post-cardiotomy ECMO: A word of caution. Crit Care. 2012;16:427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 30]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
48.  Hage A, Louzada M, Kiaii B. Sepsis-induced heparin resistance during extracorporeal membrane oxygenation. CMAJ. 2019;191:E283-E285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
49.  Finley A, Greenberg C. Review article: Heparin sensitivity and resistance: Management during cardiopulmonary bypass. Anesth Analg. 2013;116:1210-1222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 126]  [Article Influence: 11.5]  [Reference Citation Analysis (0)]
50.  Bostwick A. Direct Thrombin Inhibitor Resistance. Abstract published at Hospital Medicine; 2015 Mar 29-Apr 1, National Harbor, USA.. J Hosp Med. 2015;10:Suppl 2: Abstract 461.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Hellwig TR, Peitz GJ, Gulseth MP. High-dose argatroban for treatment of heparin-induced thrombocytopenia with thrombosis: A case report and review of laboratory considerations. Am J Health Syst Pharm. 2012;69:490-495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
52.  Kennedy DM, Alaniz C. Apparent argatroban resistance in a patient with elevated factor VIII levels. Ann Pharmacother. 2013;47:e29.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
53.  Cardinale M, Ha M, Liu MH, Reardon DP. Direct Thrombin Inhibitor Resistance and Possible Mechanisms. Hosp Pharm. 2016;51:922-927.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 12]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
54.  Seelhammer TG, Bohman JK, Aganga DO, Maltais S, Zhao Y. Bivalirudin and ECLS: Commentary and Considerations. ASAIO J. 2019;65:e52-e53.  [PubMed]  [DOI]  [Cited in This Article: ]