Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Clin Med ; 9(12)2020 Nov 25.
Article in English | MEDLINE | ID: mdl-33255530

ABSTRACT

Guidelines for the treatment of severe bleeding comprise viscoelastic-test-guided use of coagulation factor concentrates as part of their recommendations. The aim of this study is to investigate the effects of substituting fibrinogen, prothrombin complex concentrate, and a combination of both on conventional coagulation tests, viscoelastic test results, and thrombin generation. Blood was drawn from seven healthy volunteers to obtain platelet-free plasma, which later was diluted by replacing 40%, 60%, 80%, 90%, 95%, and 99% with a crystalloid solution. The diluted samples were spiked with fibrinogen concentrate, prothrombin complex concentrate, a combination of both, or a corresponding amount of crystalloid solution. Up to a dilution level of 95%, viscoelastically determined clotting time was significantly shorter in the group substituted with fibrinogen only in comparison with the additional use of prothrombin complex concentrate. Clot firmness and endogenous thrombin potential remained at relatively stable values up to a dilution level of 95% with the substitution of fibrinogen but not prothrombin complex concentrate. Substitution of prothrombin complex concentrate led to an excessive overshoot of thrombin generation. The results of our study question currently propagated treatment algorithms for bleeding patients that include the use of prothrombin complex concentrate for patients without former intake of oral anticoagulants. Even in severely bleeding patients, thrombin generation might be sufficient to achieve adequate hemostasis.

2.
Br J Anaesth ; 123(2): 186-195, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31202564

ABSTRACT

BACKGROUND: Andexanet alfa (andexanet) reverses the anticoagulant effects of factor Xa inhibitors, but it has not been assessed in clinical studies for apixaban reversal in trauma. This study evaluated andexanet for reversing apixaban anticoagulation in a porcine polytrauma model. METHODS: Oral apixaban (20 mg q.d., n=21) or placebo (n=7; sham group) was administered to male pigs for 4 days before blunt liver injury and bi-lateral femur fracture. After trauma, animals were randomised 1:1:1 to a single andexanet bolus (1000 mg), a bolus (1000 mg) plus infusion (1200 mg over 2 h), or vehicle (control). Haemodynamic and coagulation variables were monitored for 5 h or until death. The primary endpoint was blood loss. RESULTS: Mean blood loss in sham animals was 472 (standard deviation, 58) ml 12 min after injury and 658 (98) ml at 300 min, with 100% survival. Anticoagulation with apixaban significantly increased blood loss 12 min after injury [888 (133) ml, P<0.01]. Controls exhibited total blood loss of 3403 (766) ml, with 100% mortality. Andexanet bolus or bolus plus infusion significantly reduced blood loss to 1264 (205) and 1202 (95) ml, respectively), and increased survival to 100%. Haemodynamic parameters and markers of shock recovered to pre-trauma levels in andexanet-treated animals. CONCLUSION: Andexanet effectively reversed apixaban anticoagulation and reduced blood loss induced by severe trauma. Andexanet bolus alone had a similar impact on survival and blood loss as bolus plus infusion. Therefore, a 2 h andexanet infusion after the bolus may not be necessary to restore normal haemostatic mechanisms.


Subject(s)
Blood Coagulation/drug effects , Factor Xa Inhibitors/pharmacology , Factor Xa/pharmacology , Multiple Trauma , Pyrazoles/pharmacology , Pyridones/pharmacology , Recombinant Proteins/pharmacology , Animals , Anticoagulants/pharmacology , Disease Models, Animal , Male , Swine
3.
Anesthesiology ; 131(3): 543-554, 2019 09.
Article in English | MEDLINE | ID: mdl-31180918

ABSTRACT

BACKGROUND: The risk of thromboembolic complications with prothrombin complex concentrates (PCCs) appears low when used for reversal of vitamin K antagonists but might be different in other indications (e.g., trauma). A difference in risk could arise from the plasma ratio of pro- versus anticoagulant proteins. This study used a porcine trauma model to investigate combined treatment with PCC and antithrombin. The hypothesis was that antithrombin can modulate prothrombotic effects and prevent adverse events of PCC. METHODS: Nine treatment groups (n = 7 per group) were included: control (placebo), PCC (50 IU/kg), PCC plus antithrombin (three groups, with antithrombin doses of 12.5, 25, or 50 IU/kg), fibrinogen concentrate (100 mg/kg) plus PCC, fibrinogen concentrate plus PCC plus antithrombin dose of 50 IU/kg, tranexamic acid (15 mg/kg) plus fibrinogen concentrate plus PCC, and tranexamic acid plus fibrinogen concentrate plus PCC plus antithrombin dose of 50 IU/kg. In each group, bilateral femur fractures and thorax contusion were followed 60 min later by blunt liver injury. Study treatment was then administered, and animals were subsequently observed for 210 min. RESULTS: Total blood loss (mean ± SD) was statistically significantly lower in all three PCC plus antithrombin groups (PCC plus antithrombin dose of 50 IU/kg, 672 ± 63 ml; PCC plus antithrombin dose of 25 IU/kg, 535 ± 72 ml; and PCC plus antithrombin dose of 12.5 IU/kg, 538 ± 50 ml) than in the PCC group (907 ± 132 ml), which in turn had statistically significantly reduced bleeding versus the control group (1,671 ± 409 ml). Signs of disseminated intravascular coagulation were apparent with PCC monotherapy, and early deaths occurred with fibrinogen concentrate plus PCC, attributable to pulmonary emboli. Antithrombin was protective against both of these effects: signs of disseminated intravascular coagulation were absent from the PCC plus antithrombin groups, and there were no early deaths in the group with fibrinogen concentrate plus PCC plus antithrombin dose of 50 IU/kg. CONCLUSIONS: According to this trauma model, 50 IU/kg PCC increases the risk of disseminated intravascular coagulation and other thromboembolic complications, most notably when coadministered with fibrinogen concentrate. The addition of antithrombin appears to reduce this risk.


Subject(s)
Antithrombins/therapeutic use , Blood Coagulation Factors/adverse effects , Disseminated Intravascular Coagulation/prevention & control , Animals , Disease Models, Animal , Disseminated Intravascular Coagulation/complications , Male , Swine , Wounds, Nonpenetrating/complications
4.
PLoS One ; 14(1): e0209350, 2019.
Article in English | MEDLINE | ID: mdl-30615630

ABSTRACT

BACKGROUND: Idarucizumab is a humanized Fab fragment that specifically reverses dabigatran anticoagulation. In trauma, volume expanders are used for resuscitation to compensate for blood loss and hemorrhagic shock, but it is unknown whether volume expanders influence the binding of dabigatran to its antidote. Using a porcine dilutional coagulopathy model, this study investigated whether volume replacement strategies affect binding of dabigatran to idarucizumab. METHODS: Twenty-five male pigs were treated orally with dabigatran etexilate (30 mg/kg bid) for 3 days. The following day, animals were anesthetized, infused with dabigatran (total dose 0.645 mg/kg) to achieve supratherapeutic concentrations, and randomized 1:1:1:1:1 (n = 5 per group) to control (no hemodilution) or hemodilution where ~50% of blood volume was substituted with Ringer's solution, 6% hydroxyethyl starch 130/0.4, 6% hydroxyethyl starch 200/0.5 or 4% gelatin. Idarucizumab was then administered intravenously (30 mg/kg) and serial blood samples were taken for up to 24 hours to measure diluted thrombin time (corresponding with dabigatran activity), total dabigatran (bound to antidote and free drug) and a panel of coagulation parameters. RESULTS: Mean plasma dabigatran levels were 617 ± 16 ng/mL after infusion and 600 ± 114 ng/mL after ~50% hemodilution with no significant differences between groups. Following treatment with idarucizumab, plasma concentrations of unbound dabigatran decreased markedly, with similar reductions in all groups. Dabigatran-induced prolongation of coagulation parameters was rapidly reversed in all groups. CONCLUSION: This study indicates that several volume expanders used for resuscitation in trauma do not interfere with the binding of idarucizumab to dabigatran.


Subject(s)
Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antidotes/pharmacokinetics , Antithrombins/blood , Antithrombins/pharmacokinetics , Dabigatran/antagonists & inhibitors , Dabigatran/pharmacokinetics , Hemodilution , Animals , Blood Coagulation Disorders/blood , Blood Coagulation Disorders/therapy , Blood Volume/physiology , Dabigatran/blood , Male , Models, Animal , Plasma Substitutes/administration & dosage , Plasma Substitutes/metabolism , Sus scrofa
5.
Transfusion ; 59(4): 1376-1387, 2019 04.
Article in English | MEDLINE | ID: mdl-30586470

ABSTRACT

BACKGROUND: Idarucizumab (IDA) is approved for emergency reversal of dabigatran; prothrombin complex concentrates (PCCs) are recommended in the absence of specific antidote. The combined effects of IDA and PCC in trauma-related bleeding are unknown. The efficacy and safety of combined IDA + PCC were assessed in a lethal porcine model of double trauma under dabigatran anticoagulation. STUDY DESIGN AND METHODS: Male pigs (n = 28) received oral dabigatran etexilate (30 mg/kg bid) for 3 days; a non-dabigatran control group (n = 7) received placebo. On Day 4, dabigatran-treated animals were randomized 1:1:1:1 to receive placebo + placebo (dabigatran-treated control), IDA + PCC (60 mg/kg + 50 IU/kg), PCC + IDA, or IDA + IDA. Trauma was induced at t = 0 (bilateral femur fractures and blunt liver injury) and t = 60 minutes (second blunt liver injury). Study treatment was administered 15 minutes after each trauma. Animals were monitored for 5 hours or until death. RESULTS: Total blood loss in IDA + PCC, PCC + IDA, and IDA + IDA was 1673 ± 370, 1981 ± 361, and 1417 ± 135 mL, respectively, with 100% survival at 5 hours. Blood loss in dabigatran-treated controls was 4427 ± 162 mL with 100% mortality. With IDA + IDA, plasma coagulation parameters and thrombin generation were similar to non-dabigatran control group levels after the second dose and remained stable over time. In the IDA + PCC and PCC + IDA groups, thrombin generation and d-dimer levels after the second dose were higher than with IDA + IDA. No thromboembolic complications were found. CONCLUSION: IDA and PCC are effective in treating trauma-related bleeding with dabigatran anticoagulation. IDA is preferable for emergency reversal of dabigatran, but PCC may be valuable when the anticoagulant is unknown.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Blood Coagulation Factors/administration & dosage , Dabigatran/adverse effects , Hemorrhage/drug therapy , Multiple Trauma/complications , Animals , Blood Coagulation , Dabigatran/blood , Disease Models, Animal , Hemodynamics/drug effects , Male , Multiple Trauma/blood , Swine , Thrombin/biosynthesis
6.
Clin Appl Thromb Hemost ; 24(5): 724-733, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29534609

ABSTRACT

Idarucizumab, a humanized monoclonal antibody fragment (Fab), provides rapid and sustained reversal of dabigatran-mediated anticoagulation. Idarucizumab and dabigatran are mainly eliminated via the kidneys. This analysis aimed to characterize the renal elimination of idarucizumab and investigate the influence of idarucizumab on the pharmacokinetics (PK) of dabigatran and vice versa. Studies were conducted in 5/6 nephrectomized rats, in human volunteers with and without renal impairment, and in a porcine liver trauma model. In both rats and humans, renal impairment increased idarucizumab exposure and initial half-life but did not affect its terminal half-life. Urinary excretion of unchanged idarucizumab increased with increasing idarucizumab dose, suggesting saturation of renal tubular reuptake processes at higher doses. The PK of idarucizumab was unaffected by dabigatran. In contrast, idarucizumab administration resulted in redistribution of dabigatran to the plasma, where it was bound and inactivated by idarucizumab. Urinary excretion of dabigatran after administration of idarucizumab was delayed, but total dabigatran excreted in urine was unaffected. Idarucizumab and dabigatran were eliminated together via renal pathways.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Dabigatran/pharmacokinetics , Renal Elimination , Animals , Antibodies, Monoclonal, Humanized/metabolism , Blood Coagulation/drug effects , Dabigatran/metabolism , Humans , Kidney/metabolism , Rats , Renal Insufficiency , Swine
7.
Circ J ; 82(5): 1309-1318, 2018 04 25.
Article in English | MEDLINE | ID: mdl-29237991

ABSTRACT

BACKGROUND: In long-term left ventricular assist device (LVAD) therapy, recurrent bleeding events may justify cessation of anticoagulation therapy (AT). However, data about THE safety and risks of AT cessation in LVAD patients are scarce.Methods and Results:Between 2010 and 2015, 128 patients received a HeartMate II (HMII). Following recurrent bleeding events, we ceased vitamin K antagonist (VKA) therapy in 13 patients (10%) (no-VKA group). To characterize the hemostatic profile, we performed von Willebrand factor (vWF), platelet function (PF), and other hemostatic tests in all HMII patients. The incidence of pump thrombosis (PT), ischemic stroke (IS) and bleeding events in this HMII population was 4.7 %, 6.2% and 36.7%, respectively. Median survival without VKA was 435 days. No cases of PT and only 1 of IS occurred after AT discontinuation. All patients had impaired PF and acquired von Willebrand syndrome (AvWS). However, the vWF collagen-binding activity to antigen ratio before and after VKA cessation was significantly lower in the no-VKA group compared with the HMII population (0.60±0.12 vs. 0.73±0.14, P=0.006). The thrombin-antithrombin III complex (TAT) value was significantly higher in the no-VKA group (P=0.0005). CONCLUSIONS: We experienced good results with AT cessation in specific HMII patients. The simultaneous onset of AvWS and high TAT values could explain at least in part the low thromboembolic rate in HMII patients without VKA.


Subject(s)
Anticoagulants/administration & dosage , Heart-Assist Devices/adverse effects , Hemorrhage/mortality , Hemostasis , Prosthesis Implantation/adverse effects , Thromboembolism/mortality , Aged , Anticoagulants/adverse effects , Brain Ischemia/blood , Brain Ischemia/etiology , Brain Ischemia/mortality , Female , Hemorrhage/blood , Hemorrhage/etiology , Humans , Incidence , Male , Middle Aged , Prospective Studies , Retrospective Studies , Stroke/blood , Stroke/etiology , Stroke/mortality , Thromboembolism/blood , Thromboembolism/etiology , von Willebrand Diseases/blood , von Willebrand Diseases/etiology , von Willebrand Diseases/mortality , von Willebrand Factor/metabolism
8.
Anesthesiology ; 127(5): 852-861, 2017 11.
Article in English | MEDLINE | ID: mdl-28857806

ABSTRACT

BACKGROUND: Although idarucizumab is the preferred treatment for urgent dabigatran reversal, it is not always available. Prothrombin complex concentrate (PCC) may be an alternative and, with bleeding in trauma, additional hemostatic therapy may be required. The authors investigated multimodal treatment in a preclinical polytrauma model. METHODS: Dabigatran etexilate (30 mg/kg twice daily) was given orally to 45 male pigs for 3 days. On day 4, animals received a dabigatran infusion before blunt liver injury and bilateral femur fractures. After injury, animals were randomized 1:1:1:1:1 to receive placebo (control), tranexamic acid (TXA; 20 mg/kg) plus human fibrinogen concentrate (FCH; 80 mg/kg) (TXA-FCH group), PCC (25 U/kg or 50 U/kg) plus TXA plus FCH (PCC25 and PCC50 groups), or 60 mg/kg idarucizumab (IDA) plus TXA plus FCH (IDA group). Animals were monitored for 240 min after trauma, or until death. RESULTS: The degree of injury was similar in all animals before intervention. Control and TXA-FCH animals had the highest total postinjury blood loss (3,652 ± 601 and 3,497 ± 418 ml) and 100% mortality (mean survival time 96 and 109 min). Blood loss was significantly lower in the PCC50 (1,367 ± 273 ml) and IDA (986 ± 144 ml) groups, with 100% survival. Thrombin-antithrombin levels and thrombin generation were significantly elevated in the PCC50 group. CONCLUSIONS: Idarucizumab may be considered the optimal treatment for emergency reversal of dabigatran anticoagulation. However, this study suggests that PCC may be similarly effective as idarucizumab and could therefore be valuable when idarucizumab is unavailable. (Anesthesiology 2017; 127:852-61).


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Blood Coagulation Factors/therapeutic use , Blood Coagulation/drug effects , Dabigatran/toxicity , Disease Models, Animal , Multiple Trauma/drug therapy , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Anticoagulants/toxicity , Blood Coagulation/physiology , Blood Coagulation Factors/pharmacology , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Hemostasis/drug effects , Hemostasis/physiology , Humans , Male , Multiple Trauma/pathology , Random Allocation , Swine
9.
Shock ; 48(2): 144-158, 2017 08.
Article in English | MEDLINE | ID: mdl-28471371

ABSTRACT

Several direct oral anticoagulants (DOACs), including direct thrombin and factor Xa inhibitors, have been approved as alternatives to vitamin K antagonist anticoagulants. As with any anticoagulant, DOAC use carries a risk of bleeding. In patients with major bleeding or needing urgent surgery, reversal of DOAC anticoagulation may be required, presenting a clinical challenge. The optimal strategy for DOAC reversal is being refined, and may include use of hemostatic agents such as prothrombin complex concentrates (PCCs; a source of concentrated clotting factors), or DOAC-specific antidotes (which bind their target DOAC to abrogate its activity). Though promising, most specific antidotes are still in development.Preclinical animal research is the key to establishing the efficacy and safety of potential reversal agents. Here, we summarize published preclinical animal studies on reversal of DOAC anticoagulation. These studies (n = 26) were identified via a PubMed search, and used rodent, rabbit, pig, and non-human primate models. The larger of these animals have the advantages of similar blood volume/hemodynamics to humans, and can be used to model polytrauma. We find that in addition to varied species being used, there is variability in the models and assays used between studies; we suggest that blood loss (bleeding volume) is the most clinically relevant measure of DOAC anticoagulation-related bleeding and its reversal.The studies covered indicate that both PCCs and specific reversal agents have the potential to be used as part of a clinical strategy for DOAC reversal. For the future, we advocate the development and use of standardized, clinically, and pharmacologically relevant animal models to study novel DOAC reversal strategies.


Subject(s)
Antidotes/therapeutic use , Blood Coagulation Factors/therapeutic use , Disease Models, Animal , Hemorrhage , Administration, Oral , Animals , Anticoagulants/administration & dosage , Anticoagulants/therapeutic use , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Humans
10.
Thromb Haemost ; 117(7): 1370-1378, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28424818

ABSTRACT

Idarucizumab is licensed for emergency reversal of dabigatran. A single 5 g dose is usually sufficient, but higher doses may sometimes be required and optimum dosing has not been defined. It was the aim of this study to investigate the effect of idarucizumab, given once or as a split dose, after double trauma in pigs anticoagulated with dabigatran. Dabigatran etexilate (30 mg/kg bid) was given to 18 male pigs orally for 3 days. On day 4, animals were randomised 1:1:1 to receive idarucizumab 60+0, 60+60 or 120+0 mg/kg. Doses were administered 15 and 75 minutes after initial liver trauma. At 60 minutes, a second liver injury was undertaken. Animals were monitored for 5 hours after initial trauma or until death. Blood loss during the first hour was 990 ± 109 ml, 988 ± 84 ml and 964 ± 75 ml in the 60+0, 60+60 and 120+0 groups, respectively. In the 120+0 and 60+60 groups, total blood loss was 1659 ± 346 and 1426 ± 106 ml, respectively, and survival at 5 hours was 100 %. However, in the 60+0 group, total blood loss was 3561 ± 770 ml and survival was 50 %. Analysis of dabigatran plasma concentrations showed that equimolar concentrations of idarucizumab are necessary to bind all dabigatran and achieve sufficient thrombin generation. At sufficient doses, idarucizumab rapidly reduced blood loss and improved survival in this lethal porcine model of double trauma with dabigatran anticoagulation. In clinical practice, should bleeding continue after initial treatment with the approved 5 g dose of idarucizumab, a second dose may potentially be effective to control bleeding caused by redistribution of unbound dabigatran.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Dabigatran/antagonists & inhibitors , Hemorrhage/therapy , Animals , Antibodies, Monoclonal, Humanized/blood , Antidotes/administration & dosage , Antithrombins/blood , Antithrombins/toxicity , Blood Coagulation , Dabigatran/blood , Dabigatran/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Hemorrhage/etiology , Hemostasis , Liver/injuries , Male , Sus scrofa
11.
Eur J Cardiothorac Surg ; 51(3): 449-456, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27806995

ABSTRACT

Objectives: For patients with a severe acute respiratory distress syndrome (ARDS), extracorporeal membrane oxygenation (ECMO) represents a life-saving measure. Frequently, patients with severe ARDS also show signs of severe sepsis. As blood contact with the membrane oxygenator surface leads to adverse effects due to insufficient biocompatibility partly caused by activation of platelets, coagulation factors and leucocytes, we hypothesized that these adverse effects would be amplified if septic blood in a preactivated state came into contact with the membrane oxygenator. Methods: In a previously established in vitro 12-h ECMO test system (mock loop), we used septic or healthy domestic pig blood to analyse coagulation and inflammatory parameters. Sepsis was induced by a caecal ligation and puncture model in pigs. Results: At the beginning of the mock loop experiments, the septic blood showed significantly increased thrombin-antithrombin complexes (76.9 vs 27.7 µg/l), D-dimers (1.2 vs 0.3 mg/l) and fibrinogen concentration (1.8 vs 1.5 g/l), as well as elevated extrinsic coagulation activity (shorter EXTEM-CT: 44.2 vs 57 s) and higher lactate (3.4 vs 1.5 mmol/l) and cytokine levels (interleukin-6: 827 vs 31 pg/ml) when compared with the blood from healthy animals. Despite the preactivated status of the septic blood, no further increase of coagulation activity, inflammatory response or increased oxygenator resistance was observed in comparison to the control experiments. Conclusion: Septic porcine blood was not further activated due to the contact with an oxygenator, and no increased clot formation or biocompatibility problems were observed.


Subject(s)
Blood Coagulation/physiology , Extracorporeal Membrane Oxygenation/methods , Sepsis/blood , Animals , Biocompatible Materials , Blood Specimen Collection/methods , Cytokines/blood , Disease Models, Animal , Hemodynamics/physiology , Inflammation Mediators/metabolism , Oxygenators, Membrane , Platelet Activation , Sepsis/diagnosis , Sus scrofa
12.
Anesth Analg ; 123(1): 38-48, 2016 07.
Article in English | MEDLINE | ID: mdl-27192476

ABSTRACT

BACKGROUND: The potential clinical benefits of targeted therapy with coagulation factor concentrates (e.g., fibrinogen) and antifibrinolytic agents (e.g., tranexamic acid [TXA]) for the treatment of trauma-induced coagulopathy are increasingly recognized. We hypothesized that human fibrinogen concentrate (FC) and prothrombin complex concentrate (PCC), administered as combined therapy with TXA, would provide additive effects for reducing blood loss in an animal trauma model. METHODS: Thirty-six pigs were subjected to 2 consecutive blunt liver injuries, resulting in severe hemorrhagic shock and coagulopathy. Intervention comprised saline (control group); TXA (15 mg kg, TXA group); TXA and FC (90 mg kg, TXA-FC); or TXA, FC, and PCC (20 U kg, TXA-FC-PCC). Blood loss, thromboelastometry (ROTEM), measures of thrombin generation, platelet activation, and global coagulation variables were monitored for 4 hours. Tissue sections were examined to determine the occurrence of thromboembolic events. RESULTS: Total blood loss was similar in the TXA-FC and TXA-FC-PCC groups (mean ± SD: 1012 ± 86 mL and 1037 ± 118 mL, respectively; P = 1.000). These values were both lower (P < 0.001) than the TXA group (1579 ± 306 mL). Blood loss in all 3 intervention groups was lower (P < 0.001) than in the control group (2376 ± 478 mL). After trauma and resuscitation, but before study intervention, plasma fibrinogen levels were severely depleted (median for the whole study population: 66 mg dL; interquartile range: 51-108 mg dL) and clot strength was decreased (EXTEM whole-blood maximum clot firmness [MCF]: 53 ± 5 mm). Compared with controls, TXA inhibited fibrinolysis and stabilized MCF and clotting time. The addition of FC restored and stabilized hemostasis to a greater extent than TXA alone; the addition of PCC had no statistically significant impact on blood loss, clot strength (MCF), or clotting time, but it increased thrombin generation. There were no significant differences among the study groups regarding platelet activation. No thrombi or microthrombi were observed in any group at necropsy. CONCLUSIONS: The early use of TXA and FC reduced blood loss and improved coagulation measurements in a porcine model of blunt liver injury and hemorrhagic shock. FC, administered in addition to TXA, was highly effective in reducing blood loss. The lack of statistically significant reduction in blood loss when PCC was added to TXA and FC may be attributable to the absence of thrombin generation impairment in this model.


Subject(s)
Abdominal Injuries/drug therapy , Antifibrinolytic Agents/pharmacology , Blood Coagulation Factors/pharmacology , Fibrinogen/pharmacology , Hemostasis/drug effects , Liver/injuries , Shock, Hemorrhagic/drug therapy , Tranexamic Acid/pharmacology , Wounds, Nonpenetrating/drug therapy , Abdominal Injuries/blood , Abdominal Injuries/diagnosis , Animals , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blood Platelets/metabolism , Disease Models, Animal , Drug Therapy, Combination , Male , Platelet Activation/drug effects , Platelet Function Tests , Shock, Hemorrhagic/blood , Shock, Hemorrhagic/diagnosis , Sus scrofa , Thrombelastography , Thrombin/metabolism , Time Factors , Wounds, Nonpenetrating/blood , Wounds, Nonpenetrating/diagnosis
13.
Thromb Haemost ; 115(2): 271-84, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26333775

ABSTRACT

Clinical use of non-vitamin K antagonist oral anticoagulants is increasingly well established. However, specific agents for reversal of these drugs are not currently available. It was to objective of this study to investigate the impact of activated prothrombin complex concentrate (aPCC) on the anticoagulant effects of dabigatran in a randomised, controlled, porcine trauma model. Twenty-one pigs received oral and intravenous dabigatran, resulting in supratherapeutic plasma concentrations. Twelve minutes after injury (standardised bilateral femur fractures and blunt liver injury), animals (n=7/group) received 25 or 50 U/kg aPCC (aPCC25 and aPCC50) or placebo (control) and were followed for 5 hours. The primary endpoint was total volume of blood loss (BL). Haemodynamic and coagulation variables (prothrombin time [PT], activated partial thromboplastin time, diluted thrombin time, thrombin-antithrombin complexes, thromboelastometry, thrombin generation and D-dimers) were measured. Twelve minutes post-injury, BL was similar between groups. Compared with control (total BL: 3807 ± 570 ml) and aPCC25 (3690 ± 454 ml; p=0.77 vs control), a significant reduction in total BL (1639 ± 276 ml; p< 0.0001) and improved survival (p< 0.05) was observed with aPCC50. Dabigatran's anticoagulant effects were effectively treated in the aPCC50 group, as measured by several parameters including EXTEM clotting time (CT) and PT. In contrast, with aPCC25, laboratory values were initially corrected but subsequently deteriorated due to ongoing blood loss. Thromboembolic or bleeding effects were not detected. In conclusion, blood loss following trauma in dabigatran-anticoagulated pigs was successfully reduced by 50 U/kg aPCC. Optimal methodology for measuring amelioration of dabigatran anticoagulation by aPCC is yet to be determined.


Subject(s)
Blood Coagulation Factors/administration & dosage , Dabigatran/administration & dosage , Hemorrhage/drug therapy , Multiple Trauma/drug therapy , Administration, Oral , Animals , Antibodies, Monoclonal, Humanized/administration & dosage , Anticoagulants/administration & dosage , Area Under Curve , Blood Coagulation , Blood Coagulation Tests , Blood Platelets/cytology , Calibration , Femoral Fractures/drug therapy , Fibrin Fibrinogen Degradation Products/chemistry , Hemodynamics , Hemorrhage/chemically induced , Hemostasis , Male , Partial Thromboplastin Time , Platelet Aggregation , Platelet Function Tests , Prothrombin Time , Random Allocation , Swine , Thrombelastography , Thrombin/chemistry , Thrombin/metabolism , Time Factors , Vitamin K/antagonists & inhibitors
14.
Anesthesiology ; 123(6): 1350-61, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26414500

ABSTRACT

BACKGROUND: In the event of trauma, emergency reversal of anticoagulation therapy may be required. However, no specific reversal agents are routinely available for the direct oral anticoagulants such as dabigatran. The authors investigated four-factor prothrombin complex concentrate (PCC) for treating dabigatran-induced anticoagulation in a porcine polytrauma model. METHODS: Dabigatran etexilate was given orally for 3 days and intravenously on day 4 to 32 pigs. Animals were randomized 1:1:1:1 to PCC (25, 50, or 100 U/kg) or saline. Study medication was administered 12 min after bilateral femur fractures and blunt liver injury. The primary endpoint was blood loss at 300 min. RESULTS: The mean plasma concentration of dabigatran was 487 ± 161 ng/ml after intravenous administration. Blood loss was 3,855 ± 258 ml in controls and 3,588 ± 241 ml in the PCC25 group. In the PCC50 and PCC100 groups, blood loss was significantly lower: 1,749 ± 47 ml and 1,692 ± 97 ml, respectively. PCC50 and PCC100 effectively reduced dabigatran's effects on coagulation parameters, whereas control and (to a lesser extent) PCC25 animals developed severe coagulopathy. Sustained increases in endogenous thrombin potential occurred with PCC50 and PCC100. CONCLUSION: Four-factor PCC (50 or 100 U/kg) is effective in reducing blood loss in dabigatran-anticoagulated pigs, but higher doses may induce a procoagulant state.


Subject(s)
Blood Coagulation Factors/pharmacology , Blood Coagulation/drug effects , Dabigatran/pharmacology , Hemorrhage/drug therapy , Multiple Trauma/complications , Animals , Antithrombins/pharmacology , Blood Coagulation Tests , Dabigatran/antagonists & inhibitors , Disease Models, Animal , Femur/injuries , Hemorrhage/etiology , Liver/injuries , Male , Sodium Chloride/administration & dosage , Swine
16.
ASAIO J ; 61(5): 548-55, 2015.
Article in English | MEDLINE | ID: mdl-26273935

ABSTRACT

In vitro test systems for extracorporeal membrane oxygenation (mock loop) represent an interesting alternative to complex and expensive in vivo test systems to analyze the pathomechanisms leading to insufficient biocompatibility. Data on mock loop systems are limited, and operation times are constricted to a maximum duration of 6 hr. This study aims at a 12 hr operation time and frequent monitoring of markers for insufficient biocompatibility in two experimental settings. Porcine blood circulated in a mock loop without any modifications, or the circuit was operated with a CO2-enhanced gas (5% CO2/21% O2/74% N2) in combination with a nutrient solution (phosphate-adenine-glucose-guanosine-saline-mannitol). Coagulation parameters changed over time without differences between the two groups. In the unmodified test setting, a pH increase was detected after 1 hr, followed by significantly increased levels of free hemoglobin as a marker for hemolysis and elevated numbers of activated platelets, which correlate with detected von Willebrand factor, microparticles, and interleukin-ß. Proinflammatory cytokine levels were significantly increased after 12 hr. In contrast, these parameters remained constant in the modified test setting providing proof of a stable operating in vitro mock loop system with an extended/prolonged operation time.


Subject(s)
Blood Circulation/physiology , Materials Testing , Oxygenators, Membrane , Animals , Extracorporeal Membrane Oxygenation/adverse effects , Female , Flow Cytometry , Hematologic Tests , Hemolysis , In Vitro Techniques , Male , Models, Cardiovascular , Oxygenators, Membrane/adverse effects , Swine , Thrombelastography , Time Factors
17.
Thromb Haemost ; 113(4): 728-40, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25567155

ABSTRACT

Urgent surgery or life-threatening bleeding requires prompt reversal of the anticoagulant effects of dabigatran. This study assessed the ability of three- and four-factor prothrombin complex concentrate (PCC) and idarucizumab (specific antidote for dabigatran) to reverse the anticoagulant effects of dabigatran in a porcine model of trauma. Twelve animals were given dabigatran etexilate (DE) orally and dabigatran intravenously, before infliction of trauma. Six animals received tranexamic acid plus fibrinogen concentrate 12 minutes post-injury. Six PCCs (each 30 and 60 U/kg) and idarucizumab (30 and 60 mg/kg) were added to blood samples ex vivo. Coagulation was assessed by several coagulation assays. All coagulation parameters were altered after dabigatran infusion (plasma level: 442 ± 138 ng/ml). Both three- and four-factor PCCs mostly or completely reversed the effects of dabigatran on thromboelastometry variables and PT but not on aPTT. Idarucizumab neutralised plasma concentrations of dabigatran, and reversed the effects of the drug on coagulation variables. Thrombin generation showed dose-dependent over-correction following the addition of PCC, implying that elevated levels of thrombin are required to overcome dabigatran-induced coagulopathy. In contrast, treatment with idarucizumab returned thrombin generation to baseline levels. Following trauma, therapy with tranexamic acid plus fibrinogen improved correction of coagulation parameters by PCC, and thromboelastometry parameters by idarucizumab. All investigated PCCs improved dabigatran- and trauma-induced coagulopathy to a similar degree. In conclusion, this study shows that three- and four-factor PCCs are similarly effective for dabigatran reversal. Idarucizumab also reversed the effects of dabigatran and, unlike PCCs, was not associated with over-correction of thrombin generation.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antithrombins/pharmacology , Blood Coagulation Factors/pharmacology , Blood Coagulation/drug effects , Coagulants/pharmacology , Dabigatran/pharmacology , Multiple Trauma/drug therapy , Administration, Oral , Animals , Antifibrinolytic Agents/pharmacology , Antithrombins/administration & dosage , Blood Coagulation Tests , Dabigatran/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrinogen/metabolism , Infusions, Intravenous , Male , Multiple Trauma/blood , Swine , Thrombin/metabolism , Time Factors , Tranexamic Acid/pharmacology
18.
Thromb Haemost ; 106(4): 724-33, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21901227

ABSTRACT

Although prothrombin complex concentrate (PCC) is increasingly used for the treatment of trauma-induced coagulopathy, few studies have investigated the impact and safety of PCC for this indication. The present study was performed to assess PCC for treatment of coagulopathy after blunt liver injury under severe hypothermia. Coagulopathy in 14 anaesthetised pigs was induced by haemodilution. Subsequently, standardised blunt liver injury was induced under severe hypothermia (32.8-33.2°C). Animals were randomised to receive either PCC (35 IU kg⁻¹) or saline (control). Coagulation was assessed over the following 2 hours by thromboelastometry and thrombin generation. Internal organs were examined to determine presence of emboli. The administration of PCC showed a significant reduction in blood loss (p=0.002 vs. controls) and a significant increase in the rate of survival (p=0.022 vs. controls). Plasma thrombin generation in the PCC group increased considerably above baseline levels, with significant increases in peak thrombin levels and endogenous thrombin potential versus controls throughout the follow-up period. In addition, PT decreased significantly in the PCC group versus the control group. However, only slight improvements in thromboelastometry variables were observed. Histology showed an equal degree of liver injury in both groups, and no thromboembolism. In severely hypothermic pigs, the application of PCC corrected trauma-induced coagulopathy and reduced blood loss. Thus, the infusion of PCC might be a reasonable approach to reduce the need for blood cell transfusion in trauma. Furthermore, the impact and safety of PCC application can be monitored through thrombin generation and thromboelastometry under hypothermia.


Subject(s)
Blood Coagulation Disorders/drug therapy , Blood Coagulation Factors/administration & dosage , Liver Diseases/drug therapy , Liver/drug effects , Wounds, Nonpenetrating/drug therapy , Animals , Blood Coagulation Disorders/etiology , Blood Coagulation Disorders/pathology , Blood Coagulation Disorders/physiopathology , Blood Coagulation Factors/adverse effects , Disease Models, Animal , Hemorrhage , Humans , Hypothermia , Liver/injuries , Liver/metabolism , Liver/pathology , Liver Diseases/complications , Liver Diseases/pathology , Liver Diseases/physiopathology , Swine , Thrombelastography , Thrombin/metabolism , Wounds, Nonpenetrating/complications , Wounds, Nonpenetrating/pathology , Wounds, Nonpenetrating/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...