Next Article in Journal
What Is New in Metabolic Dysfunction-Associated Steatotic Liver Disease in Lean Individuals: From Bench to Bedside
Previous Article in Journal
Correlations between Resting Electrocardiogram Findings and Disease Profiles: Insights from the Qatar Biobank Cohort
Previous Article in Special Issue
Reperfusion Injury: How Can We Reduce It by Pre-, Per-, and Postconditioning
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Optimal Antithrombotic Strategies in Cardiogenic Shock

by
Michal Droppa
and
Tobias Geisler
*
Department of Cardiology and Angiology, University Hospital of Tuebingen, 72076 Tuebingen, Germany
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2024, 13(1), 277; https://doi.org/10.3390/jcm13010277
Submission received: 30 November 2023 / Revised: 22 December 2023 / Accepted: 24 December 2023 / Published: 3 January 2024

Abstract

:
Cardiogenic shock (CS) represents a critical condition with a high mortality rate. The most common cause of CS is coronary artery disease, and patients typically present with myocardial infarction, necessitating immediate treatment through percutaneous coronary intervention (PCI) and often requiring mechanical circulatory support. CS is associated with a prothrombotic situation, while on the other hand, there is often a significant risk of bleeding. This dual challenge complicates the selection of an optimal antithrombotic strategy. The choice of antithrombotic agents must be personalized, taking into consideration all relevant conditions. Repeated risk assessment, therapeutic monitoring, and adjusting antithrombotic therapy are mandatory in these patients. This review article aims to provide an overview of the current evidence and practical guidance on antithrombotic strategies in the context of CS.

Graphical Abstract

1. Introduction

Cardiogenic shock (CS) is a critical condition with high mortality. It most commonly occurs in the setting of acute coronary syndrome (ACS) and requires immediate treatment with percutaneous coronary intervention (PCI). Despite optimal interventional therapy, CS is associated with high morbidity and mortality. It is often accompanied by a prothrombotic situation, making effective adjunctive antithrombotic therapy a cornerstone of therapy in this high-risk condition. However, challenges exist with regard to administration, pharmacodynamics, and the combination of anticoagulation and antiplatelet therapy. During CS, there is an imbalance between required and available organ perfusion. As a result, drug absorption and metabolism may be altered, leading to reduced efficacy and the risk of thrombotic complications. In addition, CS patients are threatened by major bleeding events, in particular those undergoing mechanical support, making a dynamic, repeated risk assessment and adjustment of antithrombotic therapy mandatory. Therefore, the choice of an appropriate antithrombotic strategy must be individualized, taking all conditions into account. This review article will give an overview of current evidence and practical guidance on antithrombotic strategies in CS.

2. Definition and Pathophysiology

The current definition and classification of the CS are based on the SCAI Shock Expert Consensus [1]. It emphasizes the dynamic process of the CS and includes five stages (Figure 1).
Stages A and B are patients “at risk” and patients with beginning hemodynamic instability without signs of systemic hypoperfusion. The “classic” cardiogenic shock includes patients from stage C; the patients have evidence of systemic and organ hypoperfusion. The hemodynamics of patients with CS are characterized by hypotension (systolic blood pressure < 90 mmHg, or a mean arterial pressure < 60 mmHg, or a drop from baseline of >30 mmHg). Manifest CS requires pharmacological or mechanical intervention beyond volume resuscitation. Other hemodynamic features include a low cardiac index (<2.2 L/min/m2) and high pulmonary capillary wedge pressure (>15 mmHg). Verification of organ hypoperfusion relies on clinical examination and laboratory findings (elevated serum lactate, BNP, renal, and hepatic markers). Various conditions, such as myocarditis, cardiomyopathies, valvular disease, pericardial tamponade, pulmonary embolism, or arrhythmias, can lead to cardiogenic shock. However, the most common cause of CS remains coronary artery disease, which usually manifests as ACS. Immediate revascularization is the standard therapy for CS in patients with STEMI and NSTEMI and can significantly improve the prognosis. PCI with coronary stent implantation is usually performed to improve the critical coronary perfusion, so the antithrombic strategy is of utmost importance and will be discussed in the following review.
There are several important pathophysiologic changes during CS that may influence the pharmacokinetics of the antithrombotic agents used. Organ hypoperfusion may delay drug absorption from the gastrointestinal tract. Underlying pathophysiologic mechanisms include decreased gastrointestinal motility, delayed gastric emptying, and diminished absorption. Furthermore, since CS patients are often under mechanical ventilation, oral medications must be administered via a nasogastric tube, further prolonging drug administration. There are also pharmacokinetic changes in drug metabolism. Hepatic dysfunction in CS may reduce the activation of prodrugs to active metabolites, diminishing drug efficacy. This becomes particularly relevant for oral P2Y12 receptor inhibitors that are activated by liver enzymes. On the other hand, hepatic elimination of drugs may be reduced, which may lead to the accumulation of substances such as oral anticoagulants (OAC), thus increasing the risk of bleeding. Similar to liver dysfunction, acute kidney injury often accompanies CS. This can lead to the accumulation of renally excreted drugs. Renal replacement therapy may also affect drug elimination, depending on the method used. Therefore, more frequent drug monitoring may be required in CS. Mild therapeutic hypothermia may also affect the pharmacokinetics of antithrombotic drugs, most likely due to impaired prodrug activation. Mechanical circulatory support with extracorporeal membrane oxygenation (ECMO) may induce sequestration of some drugs and decrease drug clearance. Drug–drug interactions may also be relevant for patients with CS. Mechanically ventilated patients require analgesia/sedation with morphine derivatives, which reduce the effect of oral antiplatelet drugs mainly due to prolonged absorption.
Similar to other disease conditions, there is a relevant overlap of bleeding and thrombotic risk factors. Risk factors can be generally divided into patient-dependent and treatment-dependent risk factors (graphical abstract). Rates for major and fatal bleeding in CS have been reported at rates between 10–20% and 3–5%, respectively, depending on bleeding classification and location (i.e., access versus non-access site bleedings) [2,3]. In the CULPPRIT-SHOCK trial, bleeding events complicated the course of CS and were associated with increased mortality. Sepsis, peripheral ischemic complications, new atrial fibrillation, and treatment with active mechanical support by ECMO or Impella were risk factors for bleeding complications [4].

3. Characteristics of Antithrombotic Substances

The pharmacological properties of different antithrombotic substances and special aspects of CS are summarized in Table 1.

3.1. Aspirin

Aspirin (acetylsalicylic acid) is an irreversible inhibitor of cyclooxygenase-1. Through irreversible acetylation of this enzyme, the formation of thromboxane-A2 in platelets is blocked. The effect persists for the life-time of platelets in the circulation (7–10 days). Aspirin can be administered per os or intravenously. The usual loading dose in aspirin-naïve patients is 150–300 mg. Aspirin is given in doses of 75–100 mg/day.
Aspirin is the standard first-line therapy for patients with ACS. According to ESC guidelines, a loading dose of 150–300 mg orally or 75–250 mg intravenously should be given as soon as possible [5]. There are no large randomized controlled trials comparing intravenous and oral aspirin. However, pharmacological data show worse outcomes with aspirin undertreatment [6] and better platelet inhibition with intravenous administration [7]. Given the unfavorable prognosis of CS and the importance of a rapid antithrombotic effect, it seems reasonable to administer aspirin intravenously.

3.2. P2Y12 Receptor Inhibitors

3.2.1. Clopidogrel

Clopidogrel is a thienopyridine prodrug that irreversibly inhibits the platelet P2Y12-receptor after biotransformation in the liver. It prevents ADP-mediated platelet activation. Hepatic activation of clopidogrel requires two steps of activation by cytochrome P450. Therefore, the therapeutic effect of clopidogrel occurs with a delay of 30–60 min after oral administration of the loading dose (usually 600 mg). A significant limitation of clopidogrel is also the inter-individual variability of its metabolism, resulting in a low clopidogrel response to therapy in a relevant number of patients [26].

3.2.2. Prasugrel

Prasugrel is an antiplatelet agent that acts similarly to clopidogrel by irreversibly inhibiting the P2Y12 receptor and thus the ADP-dependent pathway of platelet aggregation. This thienopyridine derivative is also a prodrug, but has a shorter one-step biotransformation and therefore a faster onset of antiplatelet activity than clopidogrel [27].

3.2.3. Ticagrelor

Ticagrelor is a nucleoside analogue that reversibly blocks the P2Y12 receptor at a different binding site than clopidogrel and prasugrel. Unlike thienopyridines, it does not require bioactivation. It has a rapid onset of action of about 30 min.
There are no large randomized trials comparing the use of different P2Y12 receptor inhibitors in the population of patients with CS. A retrospective analysis of the IABP-Shock II trial showed no difference in mortality between clopidogrel, prasugrel, and ticagrelor, with ticagrelor being associated with the lowest bleeding risk [8]. Two small retrospective studies with clinical endpoints (stent thrombosis, mortality) comparing clopidogrel and prasugrel/ticagrelor in the setting of cardiac arrest and mild therapeutic hypothermia have been published with conflicting results [9,10]. In three pharmacological studies, prasugrel and ticagrelor showed significantly better platelet inhibition than clopidogrel in patients after cardiac resuscitation with mild hypothermia [11,12,13]. In a meta-analysis of 1100 patients with CS or cardiac arrest, newer P2Y12 receptor inhibitors were associated with lower mortality rates without differences in bleeding risk [14].
Because of the aforementioned limitations of delayed onset of action and interindividual variability, clopidogrel has been largely replaced by the newer P2Y12 receptor inhibitors prasugrel and ticagrelor in the setting of ACS. Results from retrospective analyses and pharmacological data also support the use of the newer P2Y12 receptor inhibitors in the setting of CS. Thus, the use of clopidogrel in patients with CS, where an immediate onset of action is required, appears to be limited. However, clopidogrel has the lowest bleeding risk of all the P2Y12 receptor inhibitors and may therefore play an important role in the later phase of treatment in patients at high risk of bleeding or in combination with oral anticoagulants. All oral P2Y12 receptor inhibitors can be administered by nasogastric tube after crushing. Crushing may also be useful in patients without a nasogastric tube if a faster onset of action is desired, as shown in pharmacological studies [15,16]. A delayed onset of platelet inhibition was observed for all oral P2Y12 inhibitors in ACS/CS patients who were treated with opioids such as morphine and fentanyl, potentially increasing the risk of ischemic events [17,18,19]. These effects have been explained by impaired drug metabolism leading to reduced active drug concentrations due to reduced gastric emptying and delayed intestinal absorption rather than drug interactions on a pharmacological level [20,21,22]. Use of alternative analgesics (e.g., IV acetaminophen or peripheral opioid receptor antagonist) [23,24], coadministration of metoclopramide [25], crushed administration of oral P2Y12 receptor inhibitors, or use of intravenous platelet inhibitors might overcome attenuated platelet inhibition.

3.2.4. Cangrelor

Cangrelor is a direct, reversible P2Y12 receptor inhibitor that blocks ADP-dependent platelet aggregation. It is administered intravenously as a bolus, followed by a continuous infusion. It inhibits platelets immediately after application. It is characterized by a very short plasmatic half-time of 3–6 min, the platelet function recovers completely within 30 min. The seamless transition from cangrelor to oral P2Y12 is of utmost importance to avoid gaps in platelet inhibition leading to thrombotic events/stent thrombosis. According to the drug label, a loading dose of clopidogrel, ticagrelor, or prasugrel should be administered immediately after cessation of the cangrelor infusion. Alternatively, a loading dose of ticagrelor or prasugrel, but not clopidogrel, can be administered 30 min before the end of the infusion [28]. The current ESC guidelines recommend that, in cases of concomitant ticagrelor treatment, ticagrelor (LD 180 mg) shouldbe administered at the time of PCI to minimize the potential gap in platelet inhibition during the transition phase [5].
Cangrelor was approved on the basis of large randomized trials comparing its effect with that of clopidogrel in patients with elective PCI and ACS [29,30,56,57]. Cangrelor was superior to clopidogrel for the combined endpoints of death, myocardial infarction, ischemia-driven revascularization, or stent thrombosis at 48 h after PCI. However, there are no large randomized trials comparing cangrelor with newer P2Y12 receptor inhibitors, and patients with CS were not included in the mentioned trials. Pharmacological studies show superior platelet inhibition with cangrelor even when newer oral P2Y12 receptor inhibitors are used in the setting of ACS [31,32,33]. In contrast to clinical trials, PCI in CS is one of the most common indications in real-world registries. In one retrospective analysis, cangrelor was associated with a better post-procedural TIMI flow grade; another retrospective analysis showed no difference in stent thrombosis in STEMI patients with cardiac arrest [34,35]. Data from a small RCT and a national registry suggest that cangrelor prevents ischemic complications in the early hours after PCI in CS patients and is more effective than oral P2Y12 inhibitors with an acceptable safety profile [36,37]. The limitation of cangrelor is a possible treatment gap during the transition to oral platelet inhibitors due to the very short half-time. However, pharmacologic studies showed constant platelet inhibition using early loading with both ticagrelor and prasugrel during the transition phase [39,40,41]. A larger randomized trial (DAPT-SHOCK, ClinicalTrials.gov Identifier: NCT03551964) is currently evaluating the effects of cangrelor versus crushed ticagrelor in CS patients undergoing PCI with the primary combined endpoint of death, myocardial infarction, and stroke.
Cangrelor was further investigated as bridging therapy (lower dose at 0.75 μg/kg per minute) in patients with prior ACS awaiting CABG Surgery (BRIDGE-trial) showing superior platelet inhibition with tolerable bleeding rates [38].

3.3. Glycoprotein IIb/IIIa Receptor Inhibitors (GPI)

GPIs inhibit platelets via the glycoprotein IIb/IIIa receptor, which mediates the binding of activated platelets to fibrinogen. There are two substances currently available: non-peptide antagonist tirofiban and the synthetic peptide eptifibatide. The monoclonal antibody abciximab has been withdrawn from the market. GPIs are administered as a bolus during coronary intervention, followed by a continuous infusion over 12–24 h. The dose is adjusted according to weight and renal function. They have an immediate onset of antiplatelet action and a plasmatic half-life of approximately 2–3 h. Stronger antiplatelet effects of tirofiban compared to chewed prasugrel and even cangrelor have been shown in the FABOLUS-FASTER trial, a pharmacodynamic study in STEMI patients [42].
According to current guidelines, GPIs are recommended as a bailout therapy during coronary intervention for ACS with thrombotic complications (e.g., high coronary thrombus burden, slow-flow/no-flow after stent implantation, stent thrombosis), as they are associated with increased bleeding events in combination with anticoagulants and oral antiplatelet therapy. In STEMI patients, routine use was associated with reduced mortality, but at the expense of bleeding [43]. Trial evidence supporting the use of GPIs in CS is still sparse. The PRAGUE-7 trial did not find a benefit of routine upstream use of abciximab compared to standard therapy; however, the small sample size limits definite conclusions. There are conflicting associations in registries, including CS patients treated with more potent oral antiplatelet compounds, reporting a mortality benefit under treatment with GPIs in some studies [44,45] and no benefit in others [46,47]. Potential drawbacks of GPIs are bleeding and the development of immune thrombocytopenia (ITP), which is a threat for long-term use in ICU patients, e.g., under ECMO. ITP with severe thrombocytopenia has been reported in clinical trials between 0.2–0.5% for tirofiban and eptifibatide [48,49,50].

3.4. Unfractionated Heparin (UFH)

UFH consists of a mixture of glycosaminoglycans of different lengths (15–150 hexose units). The anticoagulant effect is achieved by activating antithrombin III, which inhibits the coagulation factors Xa and thrombin. UFH is administered intravenously as a bolus or as a continuous infusion. It has a short half-life of about 1–2 h. Due to the inter-individual differences in metabolism and the narrow therapeutic window, anticoagulation is controlled by activated partial thromboplastin time (aPTT) or at higher doses by activated clotting time (ACT). In the event of bleeding, heparin can be antagonized with protamine. The occurrence of heparin-induced thrombocytopenia (HIT) should be mentioned as a possible side effect. UFH is administered as standard for anticoagulation during PCI (target ACT 250–350 s). It prevents the progression of an existing coronary thrombus and catheter-associated thromboembolic complications.

3.5. Low-Molecular-Weight Heparins (LMWHs) and Fondaparinux

LMWHs inhibit factor Xa in complex with antithrombin III and have a minimal direct effect on thrombin. They exhibit more predictable pharmacokinetics compared to unfractionated heparin (UFH) and are typically administered subcutaneously to patients requiring anticoagulation. Fondaparinux is a synthetic pentasaccharide that selectively inhibits Factor Xa. Like LMWH, it can be administered subcutaneously to patients with ACS. According to the current guidelines, it can be considered in patients with NSTEMI as a delayed invasive strategy, but is not recommended for STEMI patients [5,58]. Enoxaparin is the only LMWH approved for intravenous use during PCI in ACS patients. Due to the extended plasmatic half-lives of LMWH and fondaparinux, they are usually avoided in PCI for critically ill patients, and UFH is preferred.
There are no data from interventional trials comparing different anticoagulants and different dosing regimens guided by monitoring the anticoagulant effects of LMWH and UFH specifically in CS.

3.6. Bivalirudin

Bivalirudin is a synthetic hirudin derivative that inhibits thrombin selectively and independently of antithrombin III. It is administered periinterventionally and intravenously as a bolus and as a continuous infusion for up to 4 h in a weight-adapted manner in patients with ACS. Compared to heparin, bivalirudin is characterized by more predictable pharmacokinetics with less inter-individual variability. In randomized studies, patients taking bivalirudin showed a lower risk of bleeding than those taking heparin in combination with GPI. However, with selective use of GPI, bivalirudin showed no clear advantage and an increased risk of thrombotic complications (e.g., stent thrombosis) compared to heparin. Bivalirudin is therefore only offered as an alternative to heparin (e.g., in cases of heparin-induced thrombocytopenia or significant risk of bleeding) during percutaneous interventions. CS Patients requiring VA ECMO showed fewer thrombotic and bleeding events under bivalirudin compared to UFH treatment in one recent retrospective study [53].

3.7. Argatroban

Argatroban is a direct, reversible thrombin inhibitor. Similar to bivalirudin, it is administered intravenously as a bolus and continuous infusion. Its effect can be monitored by aPTT or ACT. Argatroban is a safe alternative in cases of heparin-induced thrombocytopenia (HIT) in patients requiring anticoagulation and during PCI. Some retrospective studies favor the use of argatroban, even in patients without HIT, during therapy with ECMO [54,55].

4. Antithrombotic Therapy in Special Situations (Figure 2)

4.1. Mechanical Circulatory Support (MCS)

4.1.1. Extracorporeal Membrane Oxygenation (ECMO)

Patients on ECMO are jeopardized by thrombotic as well as bleeding complications that can occur simultaneously, making effective and safe antithrombotic therapy challenging. Blood is exposed to a large foreign surface area, including the membrane oxygenator, tubes, and pump. Adsorption of proteins involved in the coagulation cascade, exaggerated thrombin generation, and consumption of coagulation factors can foster procoagulatory and/or hyperfibrinolytic processes. UFH is most frequently used during ECMO to prevent clotting of the circuit. ACT of 180 to 200 s should be targeted. Of note, the ACT can be prolonged under hypothermia and be affected by coagulation factor levels. In addition, the correlation with other coagulation tests is inconsistent in a substantial number of cases [51]. Thus, for the management of anticoagulation, other laboratory parameters, including platelet count, hemoglobin, antithrombin levels, anti-Xa/heparin levels, and aPTT, should always be taken into consideration [52]. Viscoelastic assays (TEG-6S, ROTEM) using specific activators (e.g., tissue factor, ellagic acid, kaolin) provide complementary information to evaluate whole blood thrombogenicity and are often used for bleeding management. However, these assays have limitations in assessing the contribution of platelet reactivity in patients on dual antiplatelet therapy [59], and larger studies are warranted to establish trigger values for therapeutic decision making [60].
Figure 2. Cardiogenic shock—Aspects of Antithrombotic Therapy in Special Situations. # Calculate total UFH dose, including UFH purge concentration. * Contraindicated in liver failure and lactate acidosis. AF—atrial fibrillation; aPTT—activated partial thromboplastin time; CS—cardiogenic shock; DAPT—dual antiplatelet therapy; ECMO—extracorporeal membrane oxygenation; GPI—glycoprotein IIb/IIIa inhibitors; HBR—high bleeding risk; HIT—heparin-induced thrombocytopenia; MCS—mechanical circulatory support; NG—nasogastric tube; PCI—percutaneous coronary intervention; OAC—oral anticoagulation; RCA—regional citrate anticoagulation; TTM—targeted temperature management; UHF—unfractionated heparin.
Figure 2. Cardiogenic shock—Aspects of Antithrombotic Therapy in Special Situations. # Calculate total UFH dose, including UFH purge concentration. * Contraindicated in liver failure and lactate acidosis. AF—atrial fibrillation; aPTT—activated partial thromboplastin time; CS—cardiogenic shock; DAPT—dual antiplatelet therapy; ECMO—extracorporeal membrane oxygenation; GPI—glycoprotein IIb/IIIa inhibitors; HBR—high bleeding risk; HIT—heparin-induced thrombocytopenia; MCS—mechanical circulatory support; NG—nasogastric tube; PCI—percutaneous coronary intervention; OAC—oral anticoagulation; RCA—regional citrate anticoagulation; TTM—targeted temperature management; UHF—unfractionated heparin.
Jcm 13 00277 g002
Besides exaggerated anticoagulation, coagulation disorders can increase the bleeding risk under ECMO and should be carefully monitored. High shear stress and continuous flow of ECMO (as well as the Impella device) can induce proteolysis of von Willebrand factor, leading to acquired von Willebrand syndrome (avWS). Removal of the ECMO is currently the only solution for severe avWS, while preliminary data suggest the benefits of targeting ADAMTS13 [61]. Thrombocytopenia under ECMO is a relevant and challenging problem. Its causes and thus therapeutic considerations are manifold, including hemolysis, ineffective anticoagulation resulting in thrombosis in the vasculature and pump thrombosis, disseminated intravascular coagulation (DIC), HIT ideally detected by functional tests, sepsis, and/or bleeding. Factor XI inhibition targeting the contact activation pathway may be an attractive approach to reduce bleeding in ECMO patients in the future [62]. There is a lack of evidence on how to overcome thrombocytopenia. Treatable causes should be identified and addressed (e.g., switching from UFH to direct thrombin inhibitors such as argatroban or bivalirudin, anti-infectious therapy in the case of sepsis). Small studies suggest a benefit of cangrelor in cases of thrombocytopenia to counteract exaggerated immune complex-mediated platelet activation and further platelet consumption under ECMO [63,64]. Antiplatelet therapy in CS patients with previous PCI while on heparin anticoagulation represents a major challenge due to dynamic changes in the bleeding risk and potential vascular access site complications, making antiplatelet compounds with a long half-life undesirable. Nevertheless, in the experimental arm of the ECLS shock study, aspirin was given to 86.1%, clopidogrel to 27.2%, prasugrel to 49%, and ticagrelor to 22.3% of the patients. Moderate or severe bleeding occurred in 23.4% of ECLS patients [3]. There are only a few studies evaluating the use of short-acting IV antiplatelet drugs. Eptifibatide and cangrelor appear to prevent in-stent thrombosis but may increase bleeding in patients on ECMO in case reports [65,66]. Low-dose cangrelor (0.125 µg/kg/min starting dose continued for 5 days) combined with standard-intensity anticoagulation with bivalirudin was a feasible anti-thrombotic strategy in patients undergoing PCI during VA-ECMO support for ACS-related CS/CA. Still, major bleeding occurred in 21% of cases [67].

4.1.2. Impella

Treatment with the microaxial flow pump (Impella, Abiomed, Danvers, MA, USA) requires anticoagulation via the purge solution containing up to 25,000 U/500 mL UFH and systemic anticoagulation with a starting dose of 11–12 U/kg bodyweight. The total UFH rate should not exceed 1800 U/h. As an example, a 95 kg patient with a purge rate of 10 mL/h and UFH purge concentration of 50 U/mL would require a total UFH infusion rate of 1140 U/h, resulting in a 500 U/h purge rate and the remaining 640 U/h by IV heparinization. UFH effects should be monitored with a target ACT of 160–180 s. Guiding according to factor Xa levels (window between 0.1 and 0.3 U/mL) has been associated with a low rate of thrombotic and bleeding complications [68]. In patients with HIT, alternative systemic anticoagulation with argatroban or bivalirudin and either an anticoagulant-free or alternative anticoagulant-containing purge solution is required. The dextrose concentration (either 5% or 20% dextrose) has an impact on the anticoagulatory effects of the purge solution. Thus, 5% dextrose runs through the Impella system more quickly, delivering more anticoagulant over time. This has to be taken into account for dosing of UFH and monitoring of anticoagulation effects [69].

4.2. Hemofiltration

Acute renal failure is common in CS and is associated with increased bleeding and thrombotic risk. Continuous venovenous hemofiltration (CVVH) with early initiation and longer duration was associated with improved outcomes in patients with CS and acute renal failure after cardiac surgery [70]. Regional citrate anticoagulation seems to be superior to UFH with regard to circuit life and bleeding reduction [71,72,73]. In a small randomized study, the addition of a small-molecule GPI tirofiban to UHF was studied in patients with CS requiring hemofitration. In comparison to anticoagulation with UHF alone, tirofiban significantly reduced platelet loss during continuous renal replacement therapy [74]. However, this small study was not powered for clinical endpoints such as mortality or bleeding.

4.3. Hypothermia/Targeted Temperature Management (TTM)

Hypothermia/targeted temperature management (32–34 °C) with or without VA-ECMO may attenuate the detrimental effects of ischemia reperfusion injury in cardiogenic shock, and may be associated with a mortality reduction and improved neurological outcome; however, the controversial results exist and the certainty of the evidence is relatively low [75,76,77,78,79]. Recent guidelines recommend continuous monitoring of core temperature and actively preventing fever for at least 72 h rather than enforcing hypothermia [78]. Hypothermia has a major impact on coagulation, fibrinolysis, platelet, and endothelial cell function [80,81]. Although it has a moderate effect on spontaneous platelet activity, it substantially influences agonist-induced platelet activation [82,83]. The higher risk for stent thrombosis reported in smaller studies [84,85] could not be confirmed in a large analysis containing 49,109 cardiac arrest patients, of whom 1155 underwent hypothermia [86]. The effects of P2Y12 inhibitors are attenuated under hypothermia [13], but prasugrel and ticagrelor still achieve sufficient platelet inhibition in contrast to clopidogrel [87,88]. Mild hypothermia (34 to 37 °C) augmented eptifibatide- and tirofiban-induced inhibition of platelet aggregation [89]. Cangrelor infusion prevented platelet activation and a subsequent increase in platelet count under hypothermia in an animal model as well as an in-vitro model of extracorporeal circulation (Chandler-loop) [60]. UFH dose adjustment is required in hypothermia with titration under tight control of aPTT and/or ACT.

5. Atrial Fibrillation

AF is more frequent in patients with ACS and CS (~20%) [90]. The presence of AF increases the risk of CS and contributes to the deterioration of hemodynamics. AF already present on admission was associated with increased mortality compared to new-onset AF, and the latter was not associated with increased mortality compared to non-AF CS patients [71,90]. Antithrombotic therapy for CS patients with concomitant AF is challenging. Patients with CS already on oral anticoagulation should receive an UFH bolus according to the ACS guidelines, i.e., if the patient is on a NOAC or if the INR is <2.5 in VKA-treated patients [5]. There is a lack of evidence regarding anticoagulation/antiplatelet combination therapy in CS patients with AF undergoing PCI. The risk for stent thrombosis is usually high, and there is consensus that effective antiplatelet therapy (i.e., DAPT) is essential during the first month [5,91,92,93]. However, there are situations of high bleeding risk, such as bleeding complications under ECMO, when individual treatment decisions are required. Thus, modifications of anticoagulant type and dose (dose reduction, switching from UFH to bivalirudin/argatroban) may be necessary in these patients.

6. Management of Antithrombotic Therapy in CS Patients with Bleeding

In cases of severe bleeding, heparin can be reversed by protamine sulfate. Management of bleeding should be tailored to every patient with the reversal of anticoagulation, transfusion (when the indication exists), and re-evaluation of the anticoagulation strategy. In patients undergoing MCS with bleeding complications, coagulation disorders (e.g., Factor II, V, VIII, X deficiency, HIT, DIC, hyperfibrinolysis, avWS) should be identified and treated accordingly under coagulation monitoring (e.g., using viscoelastic assays). The timing and resumption of antiplatelet therapy need to be judged on an individual, case-based, interdisciplinary decision [94].

7. Bridging to Destination Therapy/Surgery

There are few case series using short-half-life antiplatelet agents during bridging to surgery or ventricular assist device (VAD) in patients with ACS complicated by cardiogenic shock. In a series of patients under VA-ECMO treated with cangrelor (dose 0.75 µg/kg/min), hemorrhagic complications occurred frequently, with no recurrent coronary events observed [64]. Lowering the dose (i.e., 0.5 µg/kg/min) guided by platelet function monitoring was associated with lower bleeding rates without incidences of stent thrombosis in another case series of patients bridged to surgical procedures [95]. No specific guidelines exist regarding oral antiplatelet interruption/bridging protocol in CS patients undergoing coronary bypass surgery and/or LVAD therapy. Recommendations for preoperative antiplatelet management are to continue aspirin until the day of cardiac surgery and discontinue ticagrelor, clopidogrel, or prasugrel 3, 5, or 7 days before surgery, respectively, if the urgency of the indication allows [96].

8. Conclusions and Gaps in Evidence

Management of antithrombotic therapy remains a major challenge due to the dynamical change in thrombotic and bleeding risk and the altered pharmacodynamics of antithrombotic drugs in CS and organ failure. There is a lack of evidence from studies comparing different antithrombotic medication protocols in patients with CS, in particular those undergoing mechanical cardiac support. Also, larger randomized studies to evaluate the efficacy and safety of IV versus oral antiplatelet compounds in CS patients are warranted. An ongoing randomized trial is investigating the effects of cangrelor versus crushed ticagrelor in CS patients undergoing PCI (DAPT-SHOCK, ClinicalTrials.gov Identifier: NCT03551964). There is a lack of evidence regarding anticoagulation/antiplatelet combination therapy in CS patients with AF undergoing PCI. Systematic studies should further investigate anticoagulation protocols and give guidance on how to standardize hematological and hemostatic management in critically ill patients.

Author Contributions

M.D. and T.G. writing—original draft preparation, review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

Tobias Geisler received speaker honoraria/research grants from AstraZeneca, Bayer, Bristol Myers Squibb/Pfizer, Ferrer/Chiesi, Medtronic and Edwards Lifesciences. Michal Droppa received speaker honoraria from AstraZeneca. None of them was related to this study.

References

  1. Naidu, S.S.; Baran, D.A.; Jentzer, J.C.; Hollenberg, S.M.; van Diepen, S.; Basir, M.B.; Grines, C.L.; Diercks, D.B.; Hall, S.; Kapur, N.K.; et al. SCAI SHOCK Stage Classification Expert Consensus Update: A Review and Incorporation of Validation Studies: This Statement Was Endorsed by the American College of Cardiology (ACC), American College of Emergency Physicians (ACEP), American Heart Association (AHA), European Society of Cardiology (ESC) Association for Acute Cardiovascular Care (ACVC), International Society for Heart and Lung Transplantation (ISHLT), Society of Critical Care Medicine (SCCM), and Society of Thoracic Surgeons (STS) in December 2021. J. Am. Coll. Cardiol. 2022, 79, 933–946. [Google Scholar] [CrossRef] [PubMed]
  2. Thiele, H.; Akin, I.; Sandri, M.; Fuernau, G.; de Waha, S.; Meyer-Saraei, R.; Nordbeck, P.; Geisler, T.; Landmesser, U.; Skurk, C.; et al. PCI Strategies in Patients with Acute Myocardial Infarction and Cardiogenic Shock. N. Engl. J. Med. 2017, 377, 2419–2432. [Google Scholar] [CrossRef] [PubMed]
  3. Thiele, H.; Zeymer, U.; Akin, I.; Behnes, M.; Rassaf, T.; Mahabadi, A.A.; Lehmann, R.; Eitel, I.; Graf, T.; Seidler, T.; et al. Extracorporeal Life Support in Infarct-Related Cardiogenic Shock. N. Engl. J. Med. 2023, 389, 1286–1297. [Google Scholar] [CrossRef] [PubMed]
  4. Freund, A.; Jobs, A.; Lurz, P.; Feistritzer, H.-J.; De Waha-Thiele, S.; Meyer-Saraei, R.; Montalescot, G.; Huber, K.; Noc, M.; Windecker, S.; et al. Frequency and Impact of Bleeding on Outcome in Patients with Cardiogenic Shock. JACC Cardiovasc. Interv. 2020, 13, 1182–1193. [Google Scholar] [CrossRef] [PubMed]
  5. Byrne, R.A.; Rossello, X.; Coughlan, J.J.; Barbato, E.; Berry, C.; Chieffo, A.; Claeys, M.J.; Dan, G.-A.; Dweck, M.R.; Galbraith, M.; et al. 2023 ESC Guidelines for the Management of Acute Coronary Syndromes. Eur. Heart J. 2023, 44, 3720–3826. [Google Scholar] [CrossRef] [PubMed]
  6. Tilemann, L.; Mohr, S.K.; Preusch, M.; Chorianopoulos, E.; Giannitsis, E.; Katus, H.A.; Müller, O.J. Platelet Function Monitoring for Stent Thrombosis in Critically III Patients with an Acute Coronary Syndrome. J. Intervent. Cardiol. 2018, 31, 277–283. [Google Scholar] [CrossRef] [PubMed]
  7. Zeymer, U.; Hohlfeld, T.; Vom Dahl, J.; Erbel, R.; Münzel, T.; Zahn, R.; Roitenberg, A.; Breitenstein, S.; Pap, Á.F.; Trenk, D. Prospective, Randomised Trial of the Time Dependent Antiplatelet Effects of 500 Mg and 250 Mg Acetylsalicylic Acid i. v. and 300 Mg p. o. in ACS (ACUTE). Thromb. Haemost. 2017, 117, 625–635. [Google Scholar] [CrossRef] [PubMed]
  8. Orban, M.; Kleeberger, J.; Ouarrak, T.; Freund, A.; Feistritzer, H.-J.; Fuernau, G.; Geisler, T.; Huber, K.; Dudek, D.; Noc, M.; et al. Clopidogrel vs. Prasugrel vs. Ticagrelor in Patients with Acute Myocardial Infarction Complicated by Cardiogenic Shock: A Pooled IABP-SHOCK II and CULPRIT-SHOCK Trial Sub-Analysis. Clin. Res. Cardiol. Off. J. Ger. Card. Soc. 2021, 110, 1493–1503. [Google Scholar] [CrossRef]
  9. Gouffran, G.; Rosencher, J.; Bougouin, W.; Jakamy, R.; Joffre, J.; Lamhaut, L.; Dumas, F.; Cariou, A.; Varenne, O. Stent Thrombosis after Primary Percutaneous Coronary Intervention in Comatose Survivors of Out-of-Hospital Cardiac Arrest: Are the New P2Y12 Inhibitors Really More Effective than Clopidogrel? Resuscitation 2016, 98, 73–78. [Google Scholar] [CrossRef]
  10. Jiménez-Brítez, G.; Freixa, X.; Flores-Umanzor, E.; San Antonio, R.; Caixal, G.; Garcia, J.; Hernandez-Enriquez, M.; Andrea, R.; Regueiro, A.; Masotti, M.; et al. Out-of-Hospital Cardiac Arrest and Stent Thrombosis: Ticagrelor versus Clopidogrel in Patients with Primary Percutaneous Coronary Intervention under Mild Therapeutic Hypothermia. Resuscitation 2017, 114, 141–145. [Google Scholar] [CrossRef]
  11. Steblovnik, K.; Blinc, A.; Mijovski, M.B.; Fister, M.; Mikuz, U.; Noc, M. Ticagrelor Versus Clopidogrel in Comatose Survivors of Out-of-Hospital Cardiac Arrest Undergoing Percutaneous Coronary Intervention and Hypothermia: A Randomized Study. Circulation 2016, 134, 2128–2130. [Google Scholar] [CrossRef] [PubMed]
  12. Moudgil, R.; Al-Turbak, H.; Osborne, C.; Hibbert, B.; So, D.Y.F.; Le May, M.R. Superiority of Ticagrelor Over Clopidogrel in Patients After Cardiac Arrest Undergoing Therapeutic Hypothermia. Can. J. Cardiol. 2014, 30, 1396–1399. [Google Scholar] [CrossRef] [PubMed]
  13. Ibrahim, K.; Christoph, M.; Schmeinck, S.; Schmieder, K.; Steiding, K.; Schoener, L.; Pfluecke, C.; Quick, S.; Mues, C.; Jellinghaus, S.; et al. High Rates of Prasugrel and Ticagrelor Non-Responder in Patients Treated with Therapeutic Hypothermia after Cardiac Arrest. Resuscitation 2014, 85, 649–656. [Google Scholar] [CrossRef] [PubMed]
  14. Patlolla, S.H.; Kandlakunta, H.; Kuchkuntla, A.R.; West, C.P.; Murad, M.H.; Wang, Z.; Kochar, A.; Rab, S.T.; Gersh, B.J.; Holmes, D.R.; et al. Newer P2Y12 Inhibitors vs Clopidogrel in Acute Myocardial Infarction With Cardiac Arrest or Cardiogenic Shock: A Systematic Review and Meta-Analysis. Mayo Clin. Proc. 2022, 97, 1074–1085. [Google Scholar] [CrossRef] [PubMed]
  15. Parodi, G.; Xanthopoulou, I.; Bellandi, B.; Gkizas, V.; Valenti, R.; Karanikas, S.; Migliorini, A.; Angelidis, C.; Abbate, R.; Patsilinakos, S.; et al. Ticagrelor Crushed Tablets Administration in STEMI Patients: The MOJITO Study. J. Am. Coll. Cardiol. 2015, 65, 511–512. [Google Scholar] [CrossRef] [PubMed]
  16. Rollini, F.; Franchi, F.; Hu, J.; Kureti, M.; Aggarwal, N.; Durairaj, A.; Park, Y.; Seawell, M.; Cox-Alomar, P.; Zenni, M.M.; et al. Crushed Prasugrel Tablets in Patients with STEMI Undergoing Primary Percutaneous Coronary Intervention: The CRUSH Study. J. Am. Coll. Cardiol. 2016, 67, 1994–2004. [Google Scholar] [CrossRef]
  17. Meine, T.J.; Roe, M.T.; Chen, A.Y.; Patel, M.R.; Washam, J.B.; Ohman, E.M.; Peacock, W.F.; Pollack, C.V.; Gibler, W.B.; Peterson, E.D.; et al. Association of Intravenous Morphine Use and Outcomes in Acute Coronary Syndromes: Results from the CRUSADE Quality Improvement Initiative. Am. Heart J. 2005, 149, 1043–1049. [Google Scholar] [CrossRef]
  18. Montalescot, G.; van ’t Hof, A.W.; Lapostolle, F.; Silvain, J.; Lassen, J.F.; Bolognese, L.; Cantor, W.J.; Cequier, A.; Chettibi, M.; Goodman, S.G.; et al. Prehospital Ticagrelor in ST-Segment Elevation Myocardial Infarction. N. Engl. J. Med. 2014, 371, 1016–1027. [Google Scholar] [CrossRef]
  19. Farag, M.; Spinthakis, N.; Srinivasan, M.; Sullivan, K.; Wellsted, D.; Gorog, D.A. Morphine Analgesia Pre-PPCI Is Associated with Prothrombotic State, Reduced Spontaneous Reperfusion and Greater Infarct Size. Thromb. Haemost. 2018, 118, 601–612. [Google Scholar] [CrossRef]
  20. Hobl, E.-L.; Stimpfl, T.; Ebner, J.; Schoergenhofer, C.; Derhaschnig, U.; Sunder-Plassmann, R.; Jilma-Stohlawetz, P.; Mannhalter, C.; Posch, M.; Jilma, B. Morphine Decreases Clopidogrel Concentrations and Effects: A Randomized, Double-Blind, Placebo-Controlled Trial. J. Am. Coll. Cardiol. 2014, 63, 630–635. [Google Scholar] [CrossRef]
  21. Hobl, E.-L.; Reiter, B.; Schoergenhofer, C.; Schwameis, M.; Derhaschnig, U.; Kubica, J.; Stimpfl, T.; Jilma, B. Morphine Decreases Ticagrelor Concentrations but Not Its Antiplatelet Effects: A Randomized Trial in Healthy Volunteers. Eur. J. Clin. Invest. 2016, 46, 7–14. [Google Scholar] [CrossRef]
  22. Kubica, J.; Adamski, P.; Ostrowska, M.; Sikora, J.; Kubica, J.M.; Sroka, W.D.; Stankowska, K.; Buszko, K.; Navarese, E.P.; Jilma, B.; et al. Morphine Delays and Attenuates Ticagrelor Exposure and Action in Patients with Myocardial Infarction: The Randomized, Double-Blind, Placebo-Controlled IMPRESSION Trial. Eur. Heart J. 2016, 37, 245–252. [Google Scholar] [CrossRef] [PubMed]
  23. Tavenier, A.H.; Hermanides, R.S.; Ottervanger, J.P.; Tolsma, R.; van Beurden, A.; Slingerland, R.J.; Ter Horst, P.G.J.; Gosselink, A.T.M.; Dambrink, J.-H.E.; van Leeuwen, M.A.H.; et al. Impact of Opioids on P2Y12 Receptor Inhibition in Patients with ST-Elevation Myocardial Infarction Who Are Pre-Treated with Crushed Ticagrelor: Opioids aNd Crushed Ticagrelor In Myocardial Infarction Evaluation (ON-TIME 3) Trial. Eur. Heart J. Cardiovasc. Pharmacother. 2022, 8, 4–12. [Google Scholar] [CrossRef] [PubMed]
  24. Franchi, F.; Rollini, F.; Park, Y.; Hu, J.; Kureti, M.; Rivas Rios, J.; Faz, G.; Yaranov, D.; Been, L.; Pineda, A.M.; et al. Effects of Methylnaltrexone on Ticagrelor-Induced Antiplatelet Effects in Coronary Artery Disease Patients Treated With Morphine. JACC Cardiovasc. Interv. 2019, 12, 1538–1549. [Google Scholar] [CrossRef] [PubMed]
  25. Sikora, J.; Niezgoda, P.; Barańska, M.; Buszko, K.; Skibińska, N.; Sroka, W.; Pstrągowski, K.; Siller-Matula, J.; Bernd, J.; Gorog, D.; et al. METoclopramide Administration as a Strategy to Overcome MORPHine-ticagrelOr Interaction in PatientS with Unstable Angina PectorIS-The METAMORPHOSIS Trial. Thromb. Haemost. 2018, 118, 2126–2133. [Google Scholar] [CrossRef] [PubMed]
  26. Geisler, T.; Langer, H.; Wydymus, M.; Göhring, K.; Zürn, C.; Bigalke, B.; Stellos, K.; May, A.E.; Gawaz, M. Low Response to Clopidogrel Is Associated with Cardiovascular Outcome after Coronary Stent Implantation. Eur. Heart J. 2006, 27, 2420–2425. [Google Scholar] [CrossRef] [PubMed]
  27. Wallentin, L.; Varenhorst, C.; James, S.; Erlinge, D.; Braun, O.O.; Jakubowski, J.A.; Sugidachi, A.; Winters, K.J.; Siegbahn, A. Prasugrel Achieves Greater and Faster P2Y12receptor-Mediated Platelet Inhibition than Clopidogrel Due to More Efficient Generation of Its Active Metabolite in Aspirin-Treated Patients with Coronary Artery Disease. Eur. Heart J. 2008, 29, 21–30. [Google Scholar] [CrossRef]
  28. Available online: https://www.Ema.Europa.Eu/En/Documents/Product-Information/Kengrexal-Epar-Product-Information_de.Pdf (accessed on 28 November 2023).
  29. Bhatt, D.L.; Stone, G.W.; Mahaffey, K.W.; Gibson, C.M.; Steg, P.G.; Hamm, C.W.; Price, M.J.; Leonardi, S.; Gallup, D.; Bramucci, E.; et al. Effect of Platelet Inhibition with Cangrelor during PCI on Ischemic Events. N. Engl. J. Med. 2013, 368, 1303–1313. [Google Scholar] [CrossRef]
  30. Steg, P.G.; Bhatt, D.L.; Hamm, C.W.; Stone, G.W.; Gibson, C.M.; Mahaffey, K.W.; Leonardi, S.; Liu, T.; Skerjanec, S.; Day, J.R.; et al. Effect of Cangrelor on Periprocedural Outcomes in Percutaneous Coronary Interventions: A Pooled Analysis of Patient-Level Data. Lancet 2013, 382, 1981–1992. [Google Scholar] [CrossRef]
  31. Droppa, M.; Spahn, P.; Takhgiriev, K.; Müller, K.A.L.; Alboji, A.; Straub, A.; Rath, D.; Jeong, Y.-H.; Gawaz, M.; Geisler, T. Periprocedural Platelet Inhibition with Cangrelor in P2Y12-Inhibitor Naïve Patients with Acute Coronary Syndromes—A Matched-Control Pharmacodynamic Comparison in Real-World Patients. Int. J. Cardiol. 2016, 223, 848–851. [Google Scholar] [CrossRef]
  32. Westman, P.C.; Lipinski, M.J.; Torguson, R.; Waksman, R. A Comparison of Cangrelor, Prasugrel, Ticagrelor, and Clopidogrel in Patients Undergoing Percutaneous Coronary Intervention: A Network Meta-Analysis. Cardiovasc. Revascularization Med. Mol. Interv. 2017, 18, 79–85. [Google Scholar] [CrossRef] [PubMed]
  33. Franchi, F.; Rollini, F.; Rivas, A.; Wali, M.; Briceno, M.; Agarwal, M.; Shaikh, Z.; Nawaz, A.; Silva, G.; Been, L.; et al. Platelet Inhibition with Cangrelor and Crushed Ticagrelor in Patients With ST-Segment-Elevation Myocardial Infarction Undergoing Primary Percutaneous Coronary Intervention. Circulation 2019, 139, 1661–1670. [Google Scholar] [CrossRef]
  34. Droppa, M.; Vaduganathan, M.; Venkateswaran, R.V.; Singh, A.; Szumita, P.M.; Roberts, R.J.; Qamar, A.; Hack, L.; Rath, D.; Gawaz, M.; et al. Cangrelor in Cardiogenic Shock and after Cardiopulmonary Resuscitation: A Global, Multicenter, Matched Pair Analysis with Oral P2Y12 Inhibition from the IABP-SHOCK II Trial. Resuscitation 2019, 137, 205–212. [Google Scholar] [CrossRef] [PubMed]
  35. Grimfjärd, P.; Lagerqvist, B.; Erlinge, D.; Varenhorst, C.; James, S. Clinical Use of Cangrelor: Nationwide Experience from the Swedish Coronary Angiography and Angioplasty Registry (SCAAR). Eur. Heart J. Cardiovasc. Pharmacother. 2019, 5, 151–157. [Google Scholar] [CrossRef] [PubMed]
  36. Kordis, P.; Bozic Mijovski, M.; Berden, J.; Steblovnik, K.; Blinc, A.; Noc, M. Cangrelor for Comatose Survivors of Out-of-Hospital Cardiac Arrest Undergoing Percutaneous Coronary Intervention: The CANGRELOR-OHCA Study. EuroIntervention J. Eur. Collab. Work. Group Interv. Cardiol. Eur. Soc. Cardiol. 2023, 18, 1269–1271. [Google Scholar]
  37. Zeymer, U.; Lober, C.; Richter, S.; Olivier, C.B.; Huber, K.; Haring, B.; Schwimmbeck, P.; Andrassy, M.; Akin, I.; Cuneo, A.; et al. Cangrelor in Patients with Percutaneous Coronary Intervention for Acute Myocardial Infarction after Cardiac Arrest and/or with Cardiogenic Shock. Eur. Heart J. Acute Cardiovasc. Care 2023, 12, 462–463. [Google Scholar] [CrossRef] [PubMed]
  38. Angiolillo, D.J.; Firstenberg, M.S.; Price, M.J.; Tummala, P.E.; Hutyra, M.; Welsby, I.J.; Voeltz, M.D.; Chandna, H.; Ramaiah, C.; Brtko, M.; et al. Bridging Antiplatelet Therapy with Cangrelor in Patients Undergoing Cardiac Surgery: A Randomized Controlled Trial. JAMA 2012, 307, 265–274. [Google Scholar] [CrossRef]
  39. Hochholzer, W.; Kleiner, P.; Younas, I.; Valina, C.M.; Löffelhardt, N.; Amann, M.; Bömicke, T.; Ferenc, M.; Hauschke, D.; Trenk, D.; et al. Randomized Comparison of Oral P2Y12-Receptor Inhibitor Loading Strategies for Transitioning from Cangrelor: The ExcelsiorLOAD2 Trial. JACC Cardiovasc. Interv. 2017, 10, 121–129. [Google Scholar] [CrossRef]
  40. Schneider, D.J.; Agarwal, Z.; Seecheran, N.; Keating, F.K.; Gogo, P. Pharmacodynamic Effects during the Transition between Cangrelor and Ticagrelor. JACC Cardiovasc. Interv. 2014, 7, 435–442. [Google Scholar] [CrossRef]
  41. Schneider, D.J.; Seecheran, N.; Raza, S.S.; Keating, F.K.; Gogo, P. Pharmacodynamic Effects during the Transition between Cangrelor and Prasugrel. Coron. Artery Dis. 2015, 26, 42–48. [Google Scholar] [CrossRef]
  42. Gargiulo, G.; Esposito, G.; Avvedimento, M.; Nagler, M.; Minuz, P.; Campo, G.; Gragnano, F.; Manavifar, N.; Piccolo, R.; Tebaldi, M.; et al. Cangrelor, Tirofiban, and Chewed or Standard Prasugrel Regimens in Patients With ST-Segment-Elevation Myocardial Infarction: Primary Results of the FABOLUS-FASTER Trial. Circulation 2020, 142, 441–454. [Google Scholar] [CrossRef] [PubMed]
  43. Karathanos, A.; Lin, Y.; Dannenberg, L.; Parco, C.; Schulze, V.; Brockmeyer, M.; Jung, C.; Heinen, Y.; Perings, S.; Zeymer, U.; et al. Routine Glycoprotein IIb/IIIa Inhibitor Therapy in ST-Segment Elevation Myocardial Infarction: A Meta-Analysis. Can. J. Cardiol. 2019, 35, 1576–1588. [Google Scholar] [CrossRef] [PubMed]
  44. Kanic, V.; Vollrath, M.; Penko, M.; Markota, A.; Kompara, G.; Kanic, Z. GPIIb-IIIa Receptor Inhibitors in Acute Coronary Syndrome Patients Presenting with Cardiogenic Shock and/or After Cardiopulmonary Resuscitation. Heart Lung Circ. 2018, 27, 73–78. [Google Scholar] [CrossRef] [PubMed]
  45. Bernat, I.; Abdelaal, E.; Plourde, G.; Bataille, Y.; Cech, J.; Pesek, J.; Koza, J.; Jirous, S.; Machaalany, J.; Déry, J.-P.; et al. Early and Late Outcomes after Primary Percutaneous Coronary Intervention by Radial or Femoral Approach in Patients Presenting in Acute ST-Elevation Myocardial Infarction and Cardiogenic Shock. Am. Heart J. 2013, 165, 338–343. [Google Scholar] [CrossRef] [PubMed]
  46. Saleiro, C.; de Campos, D.; Ribeiro, J.M.; Lopes, J.; Puga, L.; Sousa, J.P.; Gomes, A.R.M.; Siserman, A.; Lourenço, C.; Gonçalves, L.; et al. Glycoprotein IIb/IIIa Inhibitor Use in Cardiogenic Shock Complicating Myocardial Infarction: The Portuguese Registry of Acute Coronary Syndromes. Rev. Port. Cardiol. 2023, 42, 113–120. [Google Scholar] [CrossRef] [PubMed]
  47. De Felice, F.; Tomassini, F.; Fiorilli, R.; Gagnor, A.; Parma, A.; Cerrato, E.; Musto, C.; Nazzaro, M.S.; Varbella, F.; Violini, R. Effect of Abciximab Therapy in Patients Undergoing Coronary Angioplasty for Acute ST-Elevation Myocardial Infarction Complicated by Cardiogenic Shock. Circ. J. 2015, 79, 1568–1574. [Google Scholar] [CrossRef] [PubMed]
  48. Merlini, P.A.; Rossi, M.; Menozzi, A.; Buratti, S.; Brennan, D.M.; Moliterno, D.J.; Topol, E.J.; Ardissino, D. Thrombocytopenia Caused by Abciximab or Tirofiban and Its Association with Clinical Outcome in Patients Undergoing Coronary Stenting. Circulation 2004, 109, 2203–2206. [Google Scholar] [CrossRef]
  49. ESPRIT Investigators. Enhanced Suppression of the Platelet IIb/IIIa Receptor with Integrilin Therapy Novel Dosing Regimen of Eptifibatide in Planned Coronary Stent Implantation (ESPRIT): A Randomised, Placebo-Controlled Trial. Lancet 2000, 356, 2037–2044. [Google Scholar] [CrossRef]
  50. PURSUIT Trial Investigators. Inhibition of Platelet Glycoprotein IIb/IIIa with Eptifibatide in Patients with Acute Coronary Syndromes. N. Engl. J. Med. 1998, 339, 436–443. [Google Scholar] [CrossRef]
  51. Hohlfelder, B.; Kelly, D.; Hoang, M.; Anger, K.E.; Sylvester, K.W.; Kaufman, R.M.; Connors, J.M. Activated Clotting Times Demonstrate Weak Correlation With Heparin Dosing in Adult Extracorporeal Membrane Oxygenation. Am. J. Ther. 2022, 29, e385–e393. [Google Scholar] [CrossRef]
  52. Levy, J.H.; Staudinger, T.; Steiner, M.E. How to Manage Anticoagulation during Extracorporeal Membrane Oxygenation. Intensive Care Med. 2022, 48, 1076–1079. [Google Scholar] [CrossRef] [PubMed]
  53. Uricchio, M.N.; Ramanan, R.; Esper, S.A.; Murray, H.; Kaczorowski, D.J.; D’Aloiso, B.; Gomez, H.; Sciortino, C.; Sanchez, P.G.; Sappington, P.L.; et al. Bivalirudin Versus Unfractionated Heparin in Patients With Cardiogenic Shock Requiring Venoarterial Extracorporeal Membrane Oxygenation. ASAIO J. Am. Soc. Artif. Intern. Organs 1992 2023, 69, 107–113. [Google Scholar] [CrossRef] [PubMed]
  54. Geli, J.; Capoccia, M.; Maybauer, D.M.; Maybauer, M.O. Argatroban Anticoagulation for Adult Extracorporeal Membrane Oxygenation: A Systematic Review. J. Intensive Care Med. 2022, 37, 459–471. [Google Scholar] [CrossRef] [PubMed]
  55. Menninger, L.; Körner, A.; Mirakaj, V.; Heck-Swain, K.-L.; Haeberle, H.A.; Althaus, K.; Baumgaertner, M.; Jost, W.; Schlensak, C.; Rosenberger, P.; et al. Membrane Oxygenator Longevity Was Higher in Argatroban-Treated Patients Undergoing vvECMO. Eur. J. Clin. Invest. 2023, 53, e13963. [Google Scholar] [CrossRef] [PubMed]
  56. Bhatt, D.L.; Lincoff, A.M.; Gibson, C.M.; Stone, G.W.; McNulty, S.; Montalescot, G.; Kleiman, N.S.; Goodman, S.G.; White, H.D.; Mahaffey, K.W.; et al. Intravenous Platelet Blockade with Cangrelor during PCI. N. Engl. J. Med. 2009, 361, 2330–2341. [Google Scholar] [CrossRef] [PubMed]
  57. Harrington, R.A.; Stone, G.W.; McNulty, S.; White, H.D.; Lincoff, A.M.; Gibson, C.M.; Pollack, C.V.; Montalescot, G.; Mahaffey, K.W.; Kleiman, N.S.; et al. Platelet Inhibition with Cangrelor in Patients Undergoing PCI. N. Engl. J. Med. 2009, 361, 2318–2329. [Google Scholar] [CrossRef]
  58. Yusuf, S.; Mehta, S.R.; Chrolavicius, S.; Afzal, R.; Pogue, J.; Granger, C.B.; Budaj, A.; Peters, R.J.G.; Bassand, J.-P.; Wallentin, L.; et al. Effects of Fondaparinux on Mortality and Reinfarction in Patients with Acute ST-Segment Elevation Myocardial Infarction: The OASIS-6 Randomized Trial. JAMA 2006, 295, 1519–1530. [Google Scholar] [CrossRef]
  59. Ranucci, M.; Baryshnikova, E. Sensitivity of Viscoelastic Tests to Platelet Function. J. Clin. Med. 2020, 9, 189. [Google Scholar] [CrossRef]
  60. Erdoes, G.; Koster, A.; Levy, J.H. Viscoelastic Coagulation Testing: Use and Current Limitations in Perioperative Decision-Making. Anesthesiology 2021, 135, 342–349. [Google Scholar] [CrossRef]
  61. Bartoli, C.R.; Kang, J.; Restle, D.J.; Zhang, D.M.; Shabahang, C.; Acker, M.A.; Atluri, P. Inhibition of ADAMTS-13 by Doxycycline Reduces von Willebrand Factor Degradation During Supraphysiological Shear Stress: Therapeutic Implications for Left Ventricular Assist Device-Associated Bleeding. JACC Heart Fail. 2015, 3, 860–869. [Google Scholar] [CrossRef]
  62. Pollack, C.V.; Kurz, M.A.; Hayward, N.J. EP-7041, a Factor XIa Inhibitor as a Potential Antithrombotic Strategy in Extracorporeal Membrane Oxygenation: A Brief Report. Crit. Care Explor. 2020, 2, e0196. [Google Scholar] [CrossRef] [PubMed]
  63. Gernhofer, Y.K.; Banks, D.A.; Golts, E.; Pretorius, V. Novel Use of Cangrelor With Heparin During Cardiopulmonary Bypass in Patients With Heparin-Induced Thrombocytopenia Who Require Cardiovascular Surgery: A Case Series. Semin. Thorac. Cardiovasc. Surg. 2020, 32, 763–769. [Google Scholar] [CrossRef] [PubMed]
  64. Ciolek, A.M.; Ma, K.; Garan, A.R.; Eisenberger, A.B.; Jennings, D.L. Use of Cangrelor during Venoarterial Extracorporeal Membrane Oxygenation Following Percutaneous Coronary Intervention. Artif. Organs 2020, 44, 339–340. [Google Scholar] [CrossRef] [PubMed]
  65. Colvin, B.M.; Rivosecchi, R. Parenteral Antiplatelet Agents in Three Patients Receiving VA-ECMO Support to Maintain Drug-Eluting Stent Patency. J. Heart Lung Transplant. 2021, 40, S533. [Google Scholar] [CrossRef]
  66. Gurnani, P.K.; Bohlmann, A.; March, R.J. Prolonged Use of Eptifibatide as a Bridge to Maintain Drug-Eluting Stent Patency in a Patient Receiving Extracorporeal Membrane Oxygenation. Perfusion 2018, 33, 699–703. [Google Scholar] [CrossRef] [PubMed]
  67. Baldetti, L.; Nardelli, P.; Ajello, S.; Melisurgo, G.; Calabrò, M.G.; Pieri, M.; Scandroglio, A.M. Anti-Thrombotic Therapy With Cangrelor and Bivalirudin in Venoarterial Extracorporeal Membrane Oxygenation Patients Undergoing Percutaneous Coronary Intervention: A Single-Center Experience. ASAIO J. Am. Soc. Artif. Intern. Organs 1992 2023, 69, e346–e350. [Google Scholar] [CrossRef] [PubMed]
  68. Batra, S.; Kamran, H.; Lech, T.; Montgomery, J.; Venesy, D.; Patten, R.; Shah, S. (128)—High Thromboembolic Event Rate in Patients Supported With an Impella CP Device With an Anti-Xa Level of Less Than 0.1 u/mL. J. Heart Lung Transplant. 2018, 37, S59. [Google Scholar] [CrossRef]
  69. Vandenbriele, C.; Arachchillage, D.J.; Frederiks, P.; Giustino, G.; Gorog, D.A.; Gramegna, M.; Janssens, S.; Meyns, B.; Polzin, A.; Scandroglio, M.; et al. Anticoagulation for Percutaneous Ventricular Assist Device-Supported Cardiogenic Shock: JACC Review Topic of the Week. J. Am. Coll. Cardiol. 2022, 79, 1949–1962. [Google Scholar] [CrossRef]
  70. Vidal, S.; Richebé, P.; Barandon, L.; Calderon, J.; Tafer, N.; Pouquet, O.; Fournet, N.; Janvier, G. Evaluation of Continuous Veno-Venous Hemofiltration for the Treatment of Cardiogenic Shock in Conjunction with Acute Renal Failure after Cardiac Surgery. Eur. J. Cardio-Thorac. Surg. Off. J. Eur. Assoc. Cardio-Thorac. Surg. 2009, 36, 572–579. [Google Scholar] [CrossRef]
  71. Gorog, D.A.; Price, S.; Sibbing, D.; Baumbach, A.; Capodanno, D.; Gigante, B.; Halvorsen, S.; Huber, K.; Lettino, M.; Leonardi, S.; et al. Antithrombotic Therapy in Patients with Acute Coronary Syndrome Complicated by Cardiogenic Shock or Out-of-Hospital Cardiac Arrest: A Joint Position Paper from the European Society of Cardiology (ESC) Working Group on Thrombosis, in Association with the Acute Cardiovascular Care Association (ACCA) and European Association of Percutaneous Cardiovascular Interventions (EAPCI). Eur. Heart J.-Cardiovasc. Pharmacother. 2021, 7, 125–140. [Google Scholar] [CrossRef]
  72. Schilder, L.; Nurmohamed, S.A.; Bosch, F.H.; Purmer, I.M.; den Boer, S.S.; Kleppe, C.G.; Vervloet, M.G.; Beishuizen, A.; Girbes, A.R.J.; Ter Wee, P.M.; et al. Citrate Anticoagulation versus Systemic Heparinisation in Continuous Venovenous Hemofiltration in Critically Ill Patients with Acute Kidney Injury: A Multi-Center Randomized Clinical Trial. Crit. Care 2014, 18, 472. [Google Scholar] [CrossRef] [PubMed]
  73. Liu, C.; Mao, Z.; Kang, H.; Hu, J.; Zhou, F. Regional Citrate versus Heparin Anticoagulation for Continuous Renal Replacement Therapy in Critically Ill Patients: A Meta-Analysis with Trial Sequential Analysis of Randomized Controlled Trials. Crit. Care 2016, 20, 144. [Google Scholar] [CrossRef] [PubMed]
  74. Link, A.; Girndt, M.; Selejan, S.; Rbah, R.; Böhm, M. Tirofiban Preserves Platelet Loss during Continuous Renal Replacement Therapy in a Randomised Prospective Open-Blinded Pilot Study. Crit. Care 2008, 12, R111. [Google Scholar] [CrossRef] [PubMed]
  75. Thiele, H.; Ohman, E.M.; de Waha-Thiele, S.; Zeymer, U.; Desch, S. Management of Cardiogenic Shock Complicating Myocardial Infarction: An Update 2019. Eur. Heart J. 2019, 40, 2671–2683. [Google Scholar] [CrossRef] [PubMed]
  76. Levy, B.; Girerd, N.; Amour, J.; Besnier, E.; Nesseler, N.; Helms, J.; Delmas, C.; Sonneville, R.; Guidon, C.; Rozec, B.; et al. Effect of Moderate Hypothermia vs Normothermia on 30-Day Mortality in Patients with Cardiogenic Shock Receiving Venoarterial Extracorporeal Membrane Oxygenation: A Randomized Clinical Trial. JAMA 2022, 327, 442–453. [Google Scholar] [CrossRef] [PubMed]
  77. Bian, W.; Bian, W.; Li, Y.; Feng, X.; Song, M.; Zhou, P. Hypothermia May Reduce Mortality and Improve Neurologic Outcomes in Adult Patients Treated with VA-ECMO: A Systematic Review and Meta-Analysis. Am. J. Emerg. Med. 2023, 70, 163–170. [Google Scholar] [CrossRef]
  78. Sandroni, C.; Nolan, J.P.; Andersen, L.W.; Böttiger, B.W.; Cariou, A.; Cronberg, T.; Friberg, H.; Genbrugge, C.; Lilja, G.; Morley, P.T.; et al. ERC-ESICM Guidelines on Temperature Control after Cardiac Arrest in Adults. Intensive Care Med. 2022, 48, 261–269. [Google Scholar] [CrossRef]
  79. Fuernau, G.; Beck, J.; Desch, S.; Eitel, I.; Jung, C.; Erbs, S.; Mangner, N.; Lurz, P.; Fengler, K.; Jobs, A.; et al. Mild Hypothermia in Cardiogenic Shock Complicating Myocardial Infarction. Circulation 2019, 139, 448–457. [Google Scholar] [CrossRef]
  80. Levi, M. Hemostasis and Thrombosis in Extreme Temperatures (Hypo- and Hyperthermia). Semin. Thromb. Hemost. 2018, 44, 651–655. [Google Scholar] [CrossRef]
  81. Van der Pals, J.; Götberg, M.I.; Götberg, M.; Hultén, L.M.; Magnusson, M.; Jern, S.; Erlinge, D. Hypothermia in Cardiogenic Shock Reduces Systemic T-PA Release. J. Thromb. Thrombolysis 2011, 32, 72–81. [Google Scholar] [CrossRef]
  82. Van Poucke, S.; Stevens, K.; Marcus, A.E.; Lancé, M. Hypothermia: Effects on Platelet Function and Hemostasis. Thromb. J. 2014, 12, 31. [Google Scholar] [CrossRef] [PubMed]
  83. Xavier, R.G.; White, A.E.; Fox, S.C.; Wilcox, R.G.; Heptinstall, S. Enhanced Platelet Aggregation and Activation under Conditions of Hypothermia. Thromb. Haemost. 2007, 98, 1266–1275. [Google Scholar] [CrossRef] [PubMed]
  84. Joffre, J.; Varenne, O.; Bougouin, W.; Rosencher, J.; Mira, J.-P.; Cariou, A. Stent Thrombosis: An Increased Adverse Event after Angioplasty Following Resuscitated Cardiac Arrest. Resuscitation 2014, 85, 769–773. [Google Scholar] [CrossRef] [PubMed]
  85. Penela, D.; Magaldi, M.; Fontanals, J.; Martin, V.; Regueiro, A.; Ortiz, J.T.; Bosch, X.; Sabaté, M.; Heras, M. Hypothermia in Acute Coronary Syndrome: Brain Salvage versus Stent Thrombosis? J. Am. Coll. Cardiol. 2013, 61, 686–687. [Google Scholar] [CrossRef] [PubMed]
  86. Shah, N.; Chaudhary, R.; Mehta, K.; Agarwal, V.; Garg, J.; Freudenberger, R.; Jacobs, L.; Cox, D.; Kern, K.B.; Patel, N. Therapeutic Hypothermia and Stent Thrombosis: A Nationwide Analysis. JACC Cardiovasc. Interv. 2016, 9, 1801–1811. [Google Scholar] [CrossRef] [PubMed]
  87. Bednar, F.; Kroupa, J.; Ondrakova, M.; Osmancik, P.; Kopa, M.; Motovska, Z. Antiplatelet Efficacy of P2Y12 Inhibitors (Prasugrel, Ticagrelor, Clopidogrel) in Patients Treated with Mild Therapeutic Hypothermia after Cardiac Arrest Due to Acute Myocardial Infarction. J. Thromb. Thrombolysis 2016, 41, 549–555. [Google Scholar] [CrossRef] [PubMed]
  88. Flierl, U.; Röntgen, P.; Zauner, F.; Tongers, J.; Berliner, D.; Bauersachs, J.; Schäfer, A. Platelet Inhibition with Prasugrel in Patients with Acute Myocardial Infarction Undergoing Therapeutic Hypothermia after Cardiopulmonary Resuscitation. Thromb. Haemost. 2016, 115, 960–968. [Google Scholar] [CrossRef]
  89. Frelinger, A.L.; Furman, M.I.; Barnard, M.R.; Krueger, L.A.; Dae, M.W.; Michelson, A.D. Combined Effects of Mild Hypothermia and Glycoprotein IIb/IIIa Antagonists on Platelet–Platelet and Leukocyte–Platelet Aggregation. Am. J. Cardiol. 2003, 92, 1099–1101. [Google Scholar] [CrossRef]
  90. Feistritzer, H.-J.; Desch, S.; Zeymer, U.; Fuernau, G.; de Waha-Thiele, S.; Dudek, D.; Huber, K.; Stepinska, J.; Schneider, S.; Ouarrak, T.; et al. Prognostic Impact of Atrial Fibrillation in Acute Myocardial Infarction and Cardiogenic Shock. Circ. Cardiovasc. Interv. 2019, 12, e007661. [Google Scholar] [CrossRef]
  91. Kalarus, Z.; Svendsen, J.H.; Capodanno, D.; Dan, G.-A.; De Maria, E.; Gorenek, B.; Jędrzejczyk-Patej, E.; Mazurek, M.; Podolecki, T.; Sticherling, C.; et al. Cardiac Arrhythmias in the Emergency Settings of Acute Coronary Syndrome and Revascularization: An European Heart Rhythm Association (EHRA) Consensus Document, Endorsed by the European Association of Percutaneous Cardiovascular Interventions (EAPCI), and European Acute Cardiovascular Care Association (ACCA). EP Europace 2019, 21, 1603–1604. [Google Scholar] [CrossRef]
  92. Lip, G.Y.H.; Collet, J.-P.; Haude, M.; Byrne, R.; Chung, E.H.; Fauchier, L.; Halvorsen, S.; Lau, D.; Lopez-Cabanillas, N.; Lettino, M.; et al. 2018 Joint European Consensus Document on the Management of Antithrombotic Therapy in Atrial Fibrillation Patients Presenting with Acute Coronary Syndrome and/or Undergoing Percutaneous Cardiovascular Interventions: A Joint Consensus Document of the European Heart Rhythm Association (EHRA), European Society of Cardiology Working Group on Thrombosis, European Association of Percutaneous Cardiovascular Interventions (EAPCI), and European Association of Acute Cardiac Care (ACCA) Endorsed by the Heart Rhythm Society (HRS), Asia-Pacific Heart Rhythm Society (APHRS), Latin America Heart Rhythm Society (LAHRS), and Cardiac Arrhythmia Society of Southern Africa (CASSA). EP Europace 2019, 21, 192–193. [Google Scholar] [CrossRef]
  93. Boriani, G.; Fauchier, L.; Aguinaga, L.; Beattie, J.M.; Blomstrom Lundqvist, C.; Cohen, A.; Dan, G.-A.; Genovesi, S.; Israel, C.; Joung, B.; et al. European Heart Rhythm Association (EHRA) Consensus Document on Management of Arrhythmias and Cardiac Electronic Devices in the Critically Ill and Post-Surgery Patient, Endorsed by Heart Rhythm Society (HRS), Asia Pacific Heart Rhythm Society (APHRS), Cardiac Arrhythmia Society of Southern Africa (CASSA), and Latin American Heart Rhythm Society (LAHRS). EP Europace 2019, 21, 7–8. [Google Scholar] [CrossRef]
  94. Geisler, T.; Poli, S.; Huber, K.; Rath, D.; Aidery, P.; Kristensen, S.D.; Storey, R.F.; Ball, A.; Collet, J.-P.; Berg, J.T. Resumption of Antiplatelet Therapy after Major Bleeding. Thromb. Haemost. 2023, 123, 135–149. [Google Scholar] [CrossRef] [PubMed]
  95. Bowman, S.; Gass, J.; Weeks, P. Antiplatelet Therapy Bridging with Cangrelor in Patients With Coronary Stents: A Case Series. Ann. Pharmacother. 2019, 53, 171–177. [Google Scholar] [CrossRef]
  96. Pagano, D.; Milojevic, M.; Meesters, M.I.; Benedetto, U.; Bolliger, D.; von Heymann, C.; Jeppsson, A.; Koster, A.; Osnabrugge, R.L.; Ranucci, M.; et al. 2017 EACTS/EACTA Guidelines on Patient Blood Management for Adult Cardiac Surgery. Eur. J. Cardio-Thorac. Surg. Off. J. Eur. Assoc. Cardio-Thorac. Surg. 2018, 53, 79–111. [Google Scholar] [CrossRef]
Figure 1. SCAI SHOCK Stage Classification according to [1]. AMI—acute myocardial infarction; CA—cardiac arrest; CS—cardiogenic shock; HF—heart failure; SCAI—Society for Cardiovascular Angiography and Interventions.
Figure 1. SCAI SHOCK Stage Classification according to [1]. AMI—acute myocardial infarction; CA—cardiac arrest; CS—cardiogenic shock; HF—heart failure; SCAI—Society for Cardiovascular Angiography and Interventions.
Jcm 13 00277 g001
Table 1. Pharmacological properties of different antithrombotic substances and special aspects in cardiogenic shock.
Table 1. Pharmacological properties of different antithrombotic substances and special aspects in cardiogenic shock.
DrugRoute of AdministrationMode of ActionPlasma Half-life/Duration of Action since Last DoseDosePharmacokinetics/
Pharmacodynamics in CS
Particular Aspects in CSReferences
AspirinOral/IVIrreversible COX-1 Inhibition20 min/7–10 daysloading dose of 150–300 mg perorally or 75–250 intravenously; 75–100 mg daily maintenance dosePlatelet inhibition by
oral aspirin may be reduced during TTM
IV preferred route[5,6,7]
P2Y12 receptor inhibitors
ClopidogrelOral/crushedIrreversible P2Y12-Inhibition30–60 min/3–10 days600 mg loading dose; 75 mg daily maintenance doseDelayed GI absorption
Decreased hepatic metabolism
Due to two-step hepatic metabolism, conversion to active metabolite may be substantially impaired in CS patients with liver failure
No sufficient antiplatelet effect in hypothermia
Potential Interaction with
CYP3A4, CYP3A5 or CYP2C19 inhibitors
[8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27]
PrasugrelOral/crushedIrreversible P2Y12-Inhibition30–60 min/7–10 days60 mg loading dose; 10 mg daily maintenance dose (5 mg daily in patients >75 years of age)Delayed GI absorption
Decreased hepatic metabolism
Potential interactions with strong CYP3A4/A5
and CYP2B6
inhibitors
Delayed onset with
opioids
Long offset of 7–10 days
TicagrelorOral/crushedReversible P2Y12-Inhibition6–12 h/3–5 days180 mg loading dose; 90 mg twice daily maintenance doseDelayed GI absorption
Decreased hepatic metabolism
Interactions with strong CYP3A4 inducers or inhibitors
Delayed onset with
opioids
Contraindicated in liver failure
CangrelorIVReversible P2Y12-Inhibition3–6 min/1–2 hBolus of 30 µg/kg IV followed by 4 µg/kg/min infusion for at least 2 h or the duration of the procedure (whichever is longer)
Bridge Dose: 0.75 µg/kg/min
Not dependent on hepatic/renal metabolism in CSNo drug interactions via CYP450 metabolism.
No interaction with opiates
[28,29,30,31,32,33,34,35,36,37,38,39,40,41]
GP IIb/IIIa inhibitors
EptifibatideIVBlockade of the GP IIb/IIIa receptor2–3 h/4 hIV bolus of 180 µg/kg
followed by a continuous infusion of 2 µg/kg/min
180 µg/kg by a continuous infusion dose of 1.0 µg/kg/min in patient with 30 ≤ CrCl < 50 mL/min)
Competitive inhibition of GP IIb/IIIa receptor
Rapid recovery of platelet function
Cautious use during TTM
because of reduced net benefit
Clearance reduced in renal impairment.
Contraindicated in thrombocytopenia (<100.000 cells/mm3), severe renal impairment (<30 mL/min)/renal dialysis and severe hepatic failure, patients with prior ICH, ischemic stroke within 30 days and prior fibrinolysis
[42,43,44,45,46,47,48,49,50]
TirofibanIVBlockade of the GP Iib/IIIa receptor1.5–2 h/4–8 hBolus of 25 µg/kg IV over 3 min, followed by an infusion of 0.15 µg/kg/min for up to 18 h.
For CrCl ≤ 60 mL/min: LD, 25 µg/kg IV over 5 min followed by a maintenance infusion of 0.075 µg/kg/min continued for up to 18 h.
Initial infusion rate of 0.4 μg/kg/min for 30 min followed by 0.1 µg/kg/min
(CrCl < 30 mL/min use 0.05 µg/kg/min)
Cautious use during TTM
because of reduced net benefit
Contraindicated in thrombocytopenia (<100.000 cells/mm3), severe hepatic failure, patients with prior ICH, ischemic stroke within 30 days and prior fibrinolysis
Anticoagulants
UFHIVThrombin inhibition and factor Xa inhibition by antithrombin complex formation/activation1.5 h/2–6 hIV bolus 70–100 U/kg during PCI. IV infusion titrated to achieve an aPTT of 60–80 s (or less) depending on further indications for anticoagulation (e.g., according to ECMO/Impella protocol)Variable response in CSIn TTM, UFH dose adjustment/reduction required under frequent aPTT/ACT monitoring
Can be antagonized by protamine in case of bleeding
[5,51,52]
LMWH (enoxaparin)SC/IVFactor Xa inhibition by antithrombin complex formation/activation, little effect on thrombin4–8 h/12 hEnoxaparin: During PCI: IV bolus of 0.3 mg/kg enoxaparin if the last s.c. administration was given > 8 h before balloon inflationLess variability in drug response compared to UFHImpaired subcutaneous absorption due to reduced tissue perfusion
Monitoring of anti-Fxa activity may be necessary in critically ill patients/patients with acute renal failure
[5]
Direct intravenous
thrombin inhibitors
BivalirudinIVReversible direct thrombin inhibitor25 min/1 h0.75 mg/kg IV bolus, followed by 1.75 mg/kg/h IV infusion for duration of procedure, extended duration for up 4 h after STEMI
Renal impairment:
No reduction of bolus dose
Reduction of IV infusion dose
CrCl < 30 mL/min: 1 mg/kg/h
Hemodialysis: 0.25 mg/kg/h
HIT (off-label): 0.15–0.2 mg/kg/h IV; adjust to aPTT 1.5–2.5 times baseline value
ECMO (off-label): Individual aPTT/ACT guided protocols (e.g., [53])
No need to titrate dose
No need for routine ACT monitoring
Alternative anticoagulant in HIT
No clinically relevant drug–drug interactions reported
Safe and efficacious alternative to UFH in CS/ECMO
[53]
ArgatrobanIVReversible direct thrombin inhibitor40–50 min/2–4 hInitial dose 2 µg/kg/minute, dose adjustments according to aPTT and ACTRapid onset of action, fast reversibility of its anticoagulant effect, inhibition of clot-bound thrombin
Hepatically cleared
Alternative anticoagulant in HIT
No dosage adjustment in renal-impairment
Contraindicated in patients with severe liver dysfunction
Alternative to UHF in ECMO
[54,55]
ACT—activated clotting time; aPTT—activated partial thromboplastin time; CrCl—creatinine clearance; CS—cardiogenic shock; COX-1—cyclooxygenase 1; ECMO—extracorporeal membrane oxygenation; GI—gastrointestinal; GP Iib/IIIa—glycoprotein Iib/IIIa; IV—intravenous; LMWH—low-molecular-weight heparins; PCI—percutaneous coronary intervention; SC—subcutaneous; TTM—target temperature management; UHF—unfractionated heparin.
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

Droppa, M.; Geisler, T. Optimal Antithrombotic Strategies in Cardiogenic Shock. J. Clin. Med. 2024, 13, 277. https://doi.org/10.3390/jcm13010277

AMA Style

Droppa M, Geisler T. Optimal Antithrombotic Strategies in Cardiogenic Shock. Journal of Clinical Medicine. 2024; 13(1):277. https://doi.org/10.3390/jcm13010277

Chicago/Turabian Style

Droppa, Michal, and Tobias Geisler. 2024. "Optimal Antithrombotic Strategies in Cardiogenic Shock" Journal of Clinical Medicine 13, no. 1: 277. https://doi.org/10.3390/jcm13010277

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