Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 60
1.
CJC Open ; 5(1): 1-7, 2023 Jan.
Article En | MEDLINE | ID: mdl-36700178

Background: Dabigatran is effective and safe for stroke prevention in patients with atrial fibrillation and for venous thromboembolism prevention and treatment. In Canada, APO-dabigatran, a generic formulation, has been approved based on a bioequivalence study, but its bioavailability in settings of reduced gastric acidity has not been examined. Methods: Treatment With APO-Dabigatran Absorption (TADA) was an open-label crossover study in 46 healthy male volunteers, comparing the absorption of APO-dabigatran (150 mg) with vs without rabeprazole. The primary outcome was the 24-hour total dabigatran exposure as measured by area under the curve (AUC) and peak concentration (Cmax). Results: Compared with no rabeprazole pretreatment, the total dabigatran AUC (geometric mean [gmean] AUC0-tz: 567.2 vs 804 ngh/mL, and gmean AUC0-∞: 609.7 vs 804) and Cmax (gmean: 64.1 vs 104.4 ng/mL) were significantly reduced with rabeprazole. The percent gmean ratios for AUC0-tz, AUC0-∞, and Cmax (with rabeprazole vs without) were 70.5% (95% confidence interval [CI]: 51.9% to 95.7%), 71.8% (95% CI: 53.1% to 96.9%), and 61.4% (95% CI: 44.1% to 85.5%), respectively. With rabeprazole, the proportions of participants with > 50% reduction in AUC0-tz, AUC0-∞, and Cmax were 32.6%, 30.4%, and 39.1%, respectively. Conclusions: When APO-dabigatran is administered with rabeprazole, the exposure to dabigatran is reduced by about 30%, which is similar to the level observed with Pradaxa when it was co-administered with a proton pump inhibitor. However, the finding that one-third of participants had a > 50% reduction in exposure is concerning, and it highlights the need for caution in patients who have, or are at risk of, reduced gastric acidity.


Contexte: Le dabigatran est une option thérapeutique sûre et efficace pour prévenir les accidents vasculaires cérébraux chez les patients atteints de fibrillation auriculaire et pour prévenir et traiter les thromboembolies veineuses. Au Canada, APO-dabigatran (une version générique) a été homologué sur la base d'une étude de bioéquivalence, mais sa biodisponibilité en contexte d'acidité gastrique réduite n'a pas été évaluée. Méthodologie: L'étude TADA ( T reatment with A PO- D abigatran A bsorption) est une étude croisée menée en mode ouvert auprès de 46 hommes volontaires en bonne santé, afin de comparer l'absorption d'APO-dabigatran (à 150 mg) avec et sans rabéprazole. Le critère d'évaluation principal était l'exposition totale sur 24 heures au dabigatran, telle que mesurée par la surface sous la courbe (SSC) et la concentration maximale (Cmax). Résultats: Par rapport à une administration sans prétraitement par rabéprazole, une réduction significative de la SSC totale du dabigatran (moyenne géométrique [MG] SSC0-tz : 567,2 par rapport à 804 ngh/ml; MG SSC0-∞ : 609,7 par rapport à 804 ngh/ml) et de la Cmax (MG : 64,1 par rapport à 104,4 ng/ml) a été observée avec la prise de rabéprazole. Les ratios des MG en pourcentage de la SSC0-tz, de la SSC0-∞, et de la Cmax (avec et sans rabéprazole) étaient de 70,5 % (intervalle de confiance [IC] à 95 % : 51,9 % à 95,7 %), 71,8 % (IC à 95 % : 53,1 % à 96,9 %) et 61,4 % (IC à 95 % : 44,1 % à 85,5 %), respectivement. Les proportions de participants chez qui une réduction de > 50 % de la SSC0-tz, de la SSC0-∞ ou de la Cmax a été notée avec l'administration du rabéprazole s'élevaient à 32,6 %, 30,4 % et 39,1 %, respectivement. Conclusions: Lors de l'administration d'APO-dabigatran en concomitance avec le rabéprazole, l'exposition au dabigatran était réduite d'environ 30 %, une valeur comparable à la réduction observée lors de l'administration de Pradaxa en concomitance avec un inhibiteur de la pompe à protons. La réduction de > 50 % de l'exposition médicamenteuse chez le tiers des participants n'en est pas moins préoccupante et démontre la nécessité de faire preuve de prudence lorsque l'acidité gastrique est réduite ou risque d'être réduite chez un patient.

2.
J Thromb Haemost ; 19(5): 1348-1363, 2021 05.
Article En | MEDLINE | ID: mdl-33687782

INTRODUCTION: Vitamin K antagonists (VKA) and non-vitamin K oral antagonist anticoagulants (NOAC) are used in the clinic to reduce risk of thrombosis. However, they also exhibit vascular off-target effects. The aim of this study is to compare VKA and NOAC on atherosclerosis progression and calcification in an experimental setup. MATERIAL AND METHODS: Female Apoe-/- mice (age 12 weeks) were fed Western-type diet as control or supplemented with dabigatran etexilate or warfarin for 6 or 18 weeks. Vascular calcification was measured in whole aortic arches using µCT and [18 F]-NaF. Atherosclerotic burden was assessed by (immuno)histochemistry. Additionally, in vitro effects of warfarin, thrombin, and dabigatran on primary vascular smooth muscle cells (VSMC) were assessed. RESULTS: Short-term treatment with warfarin promoted formation of atherosclerotic lesions with a pro-inflammatory phenotype, and more rapid plaque progression compared with control and dabigatran. In contrast, dabigatran significantly reduced plaque progression compared with control. Long-term warfarin treatment significantly increased both presence and activity of plaque calcification compared with control and dabigatran. Calcification induced by warfarin treatment was accompanied by increased presence of uncarboxylated matrix Gla protein. In vitro, both warfarin and thrombin significantly increased VSMC oxidative stress and extracellular vesicle release, which was prevented by dabigatran. CONCLUSION: Warfarin aggravates atherosclerotic disease activity, increasing plaque inflammation, active calcification, and plaque progression. Dabigatran lacks undesired vascular side effects and reveals beneficial effects on atherosclerosis progression and calcification. The choice of anticoagulation impacts atherosclerotic disease by differential off target effect. Future clinical studies should test whether this beneficial effect also applies to patients.


Atherosclerosis , Atrial Fibrillation , Animals , Anticoagulants , Atherosclerosis/drug therapy , Dabigatran , Female , Humans , Mice , Vitamin K , Warfarin
3.
Ann Surg ; 274(3): e204-e211, 2021 09 01.
Article En | MEDLINE | ID: mdl-31599808

OBJECTIVE: To further examine anticoagulation reversal and clinical outcomes in dabigatran treated patients requiring urgent surgery or procedural interventions. BACKGROUND: Idarucizumab, a humanized monoclonal antibody fragment, reverses dabigatran anticoagulation. METHODS: Data from surgical and procedural patients in RE-VERSE AD, a multicenter, open-label, single-arm, prospective cohort of dabigatran reversal were evaluated. A total of 202 patients in this group received 5 g of idarucizumab before surgery or procedures. RESULTS: The interventions included 49 abdominal, 45 orthopedic, 34 vascular, 8 neurologic, and 4 genitourinary surgical procedures, or 29 catheter-based cases, 20 cases for drainage, and 8 diagnostic procedures. Five patients did not undergo their intended intervention after receiving idarucizumab. Complete reversal of the dabigatran anticoagulant effect occurred within minutes in almost all patients, with normal hemostasis in more than 91% of patients. The median time from the first vial of idarucizumab to surgery or procedures was less than 2 hours in all groups except neurosurgery, where it was 3.3 hours. Fresh frozen plasma and packed red cells were the most frequently transfused blood products. Postreversal thromboembolic events occurred in 10 (5%) patients at 30 days, 5 of whom had restarted anticoagulation before the event. Overall 30-day mortality was 12.6%. There were no serious adverse safety signals due to idarucizumab dosing. CONCLUSIONS: Idarucizumab facilitates management of patients requiring urgent procedures by providing rapid dabigatran reversal, and is the only agent of its class studied in surgical patients.


Antibodies, Monoclonal, Humanized/administration & dosage , Antithrombins/administration & dosage , Dabigatran/administration & dosage , Hemorrhage/prevention & control , Surgical Procedures, Operative , Adult , Aged , Blood Coagulation/drug effects , Blood Loss, Surgical/prevention & control , Emergencies , Female , Hemorrhage/chemically induced , Humans , Male , Middle Aged , Prospective Studies
6.
J Am Coll Cardiol ; 74(14): 1760-1768, 2019 10 08.
Article En | MEDLINE | ID: mdl-31582135

BACKGROUND: Dabigatran and idarucizumab, its reversal agent, are renally cleared. OBJECTIVES: The purpose of this study was to determine the extent of reversal and outcomes according to baseline renal function in dabigatran-treated nondialysis patients receiving idarucizumab. METHODS: In 503 patients in RE-VERSE AD (Reversal of Effects of Idarucizumab in Patients on Active Dabigatran), the extent of dabigatran reversal and clinical outcomes were compared according to baseline renal function (creatinine clearance: normal ≥80, mild 50 to <80, moderate 30 to <50, and severe <30 ml/min). RESULTS: Compared with patients with normal renal function, those with impaired renal function were older, were more often women, and had lower body mass indexes, more comorbidities, higher CHADS2 scores, and higher dabigatran plasma levels despite more frequent use of lower-dose dabigatran regimens. Regardless of renal function, median reversal measured by dilute thrombin time was 100% within 4 h of idarucizumab administration, and over 98% of patients achieved this with corresponding undetectable levels of unbound dabigatran. By 12 or 24 h, 56% of patients with severe, 29.1% with moderate, and 9.2% with mild renal impairment had dabigatran levels >20 ng/ml compared with 8.3% of patients with normal renal function at baseline. Time to cessation of bleeding and the proportion with normal hemostasis with procedures were similar regardless of renal function, but patients with severe renal impairment had higher 30- and 90-day mortality rates. CONCLUSIONS: Idarucizumab completely reverses dabigatran in >98% of patients regardless of renal function. Although re-elevation of dabigatran levels within 12 to 24 h is more common with renal impairment, the time to bleeding cessation and the extent of hemostasis during procedures are similar. (Reversal of Dabigatran Anticoagulant Effect With Idarucizumab; NCT02104947).


Antibodies, Monoclonal, Humanized/therapeutic use , Antithrombins/adverse effects , Dabigatran/adverse effects , Hemorrhage/chemically induced , Hemorrhage/drug therapy , Renal Insufficiency/drug therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/blood , Antibodies, Monoclonal, Humanized/pharmacology , Antithrombins/blood , Antithrombins/therapeutic use , Blood Coagulation/drug effects , Blood Coagulation/physiology , Cohort Studies , Dabigatran/blood , Dabigatran/therapeutic use , Female , Hemorrhage/blood , Humans , Male , Middle Aged , Prospective Studies , Renal Insufficiency/blood , Treatment Outcome
7.
Front Neurol ; 10: 966, 2019.
Article En | MEDLINE | ID: mdl-31611836

Oral anticoagulants are a critical component of stroke prevention, but carry a risk of brain hemorrhage. These hemorrhagic complications tend to occur in elderly individuals, especially those with predisposing conditions such as cerebral amyloid angiopathy (CAA). Clinical evidence suggests that non-vitamin K antagonist oral anticoagulants are safer than traditional oral anticoagulants. We analyzed whether the anticoagulant dabigatran produces cerebral microhemorrhage (the pathological substrate of MRI-demonstrable cerebral microbleeds) or intracerebral hemorrhage in aged mice with and without hemorrhage-predisposing angiopathy. We studied aged (22 months old) Tg2576 (a model of CAA) and wild-type (WT) littermate mice. Mice received either dabigatran etexilate (DE) (Tg N = 7; WT N = 10) or vehicle (Tg N = 9; WT N = 7) by gavage for 4 weeks. Anticoagulation effects of DE were confirmed using thrombin time assay. No mice experienced intracerebral hemorrhage. Cerebral microhemorrhage analysis, performed using Prussian-blue and H&E staining, showed no significant change in either number or size of cerebral microhemorrhage in DE-treated animals. Analysis of biochemical parameters for endothelial activation (ICAM-1), blood-brain barrier disruption (IgG, claudin-5, fibrinogen), microglial activation (Iba-1), or astrocyte activation (GFAP) showed neither exacerbation nor protective effects of DE in either Tg2576 or WT mice. Our study provides histological and biochemical evidence that aged mice, with or without predisposing factors for brain hemorrhage, tolerate anticoagulation with dabigatran. The absence of dabigatran-induced intracerebral hemorrhage or increased frequency of acute microhemorrhage may provide some reassurance for its use in high-risk patient populations.

8.
Thromb Haemost ; 119(6): 916-929, 2019 Jun.
Article En | MEDLINE | ID: mdl-31005062

The direct thrombin inhibitor (DTI) dabigatran is a non-vitamin K antagonist oral anticoagulant for the prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation. In addition to its anti-thrombotic efficacy, dabigatran has been suggested to exert some pro-thrombotic effect due to fostering the ligation of thrombin to its high affinity platelet receptor glycoprotein (GP) Ibα in patients with atrial fibrillation. On the other hand, we provided evidence that a member of another class of DTIs, lepirudin, stimulates the inhibitory cyclic guanosine monophosphate (cGMP)/soluble guanylate cyclase pathway in human platelets. Here, we investigated the effect of lepirudin and dabigatran spiked to platelets from healthy volunteers on GPIbα-mediated platelet aggregation and agglutination. Ristocetin/von Willebrand factor (vWF)-induced aggregation of platelets in the presence or absence of plasma was significantly inhibited by lepirudin, dabigatran and D-phenylalanyl-L-prolyl-L-arginine chloromethyl ketone (PPACK). However, ristocetin/vWF-mediated platelet agglutination and binding of vWF to platelets were not affected by the DTIs. The anti-aggregatory effect was confirmed by using the GPIbα-specific agonist echicetin beads for human and murine platelets. DTIs diminished echicetin beads-induced Syk Y352 phosphorylation (used here as readout for an early signal occurring during echicetin-induced platelet aggregation), but did not inhibit adenosine diphosphate- or thromboxane A2-induced platelet aggregation. Thrombin was not generated in response to ristocetin/vWF or echicetin beads and therefore did not explain the inhibitory effect of the DTIs. Therapeutic concentration of lepirudin and dabigatran did not affect significantly platelet vasodilator-stimulated phosphoprotein S239 phosphorylation or cGMP and cyclic adenosine monophosphate levels. These data suggest that the DTIs, lepirudin and dabigatran, impair platelet activation measured during platelet aggregation induced by ristocetin/vWF or echicetin beads.


Antithrombins/therapeutic use , Atrial Fibrillation/drug therapy , Blood Platelets/physiology , Dabigatran/therapeutic use , Platelet Aggregation/drug effects , Animals , Cells, Cultured , Female , Hirudins , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Platelet Glycoprotein GPIb-IX Complex/metabolism , Protein Binding , Recombinant Proteins/therapeutic use , Ristocetin/pharmacology , von Willebrand Factor/metabolism
9.
Transfusion ; 59(7): 2436-2445, 2019 07.
Article En | MEDLINE | ID: mdl-30946491

BACKGROUND: Comparative studies on the restoration of hemostasis with different reversal agents after dabigatran therapy have not been performed. We compared the efficacy and prothrombotic potential of the specific antidote idarucizumab with that of previously recommended non-specific procoagulant concentrates. STUDY DESIGN AND METHODS: We explored the in vitro effects of dabigatran (184 ng/mL) on fibrin and platelet-aggregate formation onto a damaged vessel under flow conditions (600 s-1 ). The reversal mechanisms and efficacy of idarucizumab (0.3-3 mg/mL) were compared with that of the non-specific procoagulant concentrates aPCC (25-75 U/Kg), PCC (70 U/Kg), or rFVIIa (120 µg/Kg). Generation of thrombin and prothrombin fragment (F1 + 2), and thromboelastometry parameters of clot formation were measured. RESULTS: Dabigatran caused pronounced reductions in fibrin (87%) and platelet interactions (36%) with damaged vessels (p < 0.01) and significantly impaired thrombin generation and thromboelastometric parameters (delayed dynamics and reduced firmness). Idarucizumab completely normalized rates of fibrin and platelet coverage to baseline values in flow studies; and reversed the alterations in thrombin generation, F1 + 2 and thromboelastometry parameters produced by dabigatran. In comparison, aPCC and PCC only partially compensated for the dabigatran-induced alterations in fibrin deposition, but were unable to fully restore them to baseline values. Reversal with aPCC or PCC improved the majority of alterations in coagulation-related tests, but tended to overcompensate thrombin generation kinetics and significantly increased F1 + 2 levels. CONCLUSION: Idarucizumab antagonizes alterations of direct and indirect biomarkers of hemostasis caused by dabigatran. In our studies, idarucizumab was clearly more efficacious than strategies with non-specific procoagulant concentrates and devoid of the excessive procoagulant tendency observed with the latter.


Antibodies, Monoclonal, Humanized/pharmacology , Blood Coagulation/drug effects , Blood Platelets/metabolism , Dabigatran/pharmacology , Fibrin/metabolism , Platelet Aggregation/drug effects , Animals , Female , Humans , Kinetics , Male , Rabbits , Thrombelastography
10.
PLoS One ; 14(1): e0209350, 2019.
Article En | MEDLINE | ID: mdl-30615630

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.


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
11.
Circulation ; 139(6): 748-756, 2019 02 05.
Article En | MEDLINE | ID: mdl-30586692

BACKGROUND: Although dabigatran has a favorable risk-benefit profile compared with vitamin K antagonist therapy for venous thromboembolism and nonvalvular atrial fibrillation, major bleeding events, including gastrointestinal (GI) bleeding, may occur. Therefore, our aim was to provide insights into the efficacy and safety of idarucizumab for urgent dabigatran reversal in patients with major GI bleeding. METHODS: Patients with uncontrollable GI bleeding requiring reversal were enrolled from June 2014 through July 2016 in the RE-VERSE AD study (Reversal of Dabigatran Anticoagulant Effect With Idarucizumab), a prospective, multicenter, open-label study of idarucizumab, and were followed up for 90 days for primary and secondary outcomes. Patients were to receive a 5-g dose of intravenous idarucizumab, administered as 2 bolus infusions of 2.5 g no more than 15 minutes apart. The primary end point was the maximum reversal of dabigatran anticoagulation within 4 hours after administration of idarucizumab as measured by the dabigatran-specific assays diluted thrombin time and ecarin clotting time. Further end points included investigator-reported bleeding cessation within the first 24 hours and incidence of rebleeding, thromboembolic events, or mortality. RESULTS: GI bleeding occurred in 137 patients enrolled in RE-VERSE AD, of which 84% was adjudicated as major or life-threatening, 48 (35.0%) was upper GI tract in origin, 43 (31.4%) was lower GI in origin, and 46 (33.6%) was either both or unknown. Complete reversal of dabigatran was observed in 118 of 121 patients (97.5%) with an elevated diluted thrombin time at presentation and 95 of 131 patients (72.5%) with an elevated ecarin clotting time and was similar for upper and lower GI bleeding. Bleeding cessation within 24 hours was reported in 92 of 134 evaluable patients (68.7%) after a median duration of 2.4 hours (interquartile range, 2.0-3.9 hours). During the 90-day follow-up, 6 patients (4.4%) had a postreversal thromboembolic event, and 20 patients (14.6%) died. CONCLUSIONS: Idarucizumab showed a rapid and complete reversal of dabigatran activity in nearly all patients presenting with GI bleeding, facilitating emergency patient care without the additional presence of anticoagulation. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov . Unique identifier: NCT02104947.


Antibodies, Monoclonal, Humanized/therapeutic use , Anticoagulants/therapeutic use , Dabigatran/therapeutic use , Drug-Related Side Effects and Adverse Reactions/epidemiology , Gastrointestinal Hemorrhage/epidemiology , Venous Thromboembolism/drug therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Anticoagulants/adverse effects , Dabigatran/adverse effects , Drug Substitution , Female , Follow-Up Studies , Gastrointestinal Hemorrhage/etiology , Humans , Male , Middle Aged , Prospective Studies , Risk Assessment , United States/epidemiology , Venous Thromboembolism/epidemiology , Vitamin K/antagonists & inhibitors
12.
Clin Appl Thromb Hemost ; 24(5): 724-733, 2018 Jul.
Article En | MEDLINE | ID: mdl-29534609

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.


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
13.
Stroke ; 48(8): 2248-2254, 2017 08.
Article En | MEDLINE | ID: mdl-28706123

BACKGROUND AND PURPOSE: Cerebral microbleeds (CMBs) predispose patients to intracerebral hemorrhage. Preclinical models to examine the effects of antithrombotic treatments on the development of clinically overt intracerebral hemorrhage are needed. We examined the natural course of CMB development and the effects of long-term anticoagulation with warfarin or dabigatran on cerebral micro- and macrohemorrhage in mice overexpressing the APP23 (amyloid precursor protein). METHODS: Repeated susceptibility-weighted magnetic resonance imaging was performed in APP23 mice at the age of 18 and 21 months, respectively. After establishing stable long-term anticoagulation effects of warfarin and dabigatran on number and total volume of CMBs, the outcome parameters were compared with nonanticoagulated control. RESULTS: CMBs were equally located in lobar and deep brain regions, and number and total volume of CMBs increased over time. Anticoagulation with either warfarin or dabigatran did not increase CMBs in APP23 significantly. Mice treated with warfarin numerically had a higher mortality (nonanticoagulated: 31%; dabigatran: 35% versus warfarin: 55%; P=0.21). In postmortem brains of prematurely dying animals warfarin caused significantly more frequently large intracerebral hemorrhage than control and dabigatran. CONCLUSIONS: Anticoagulation with warfarin or dabigatran for 3 to 4 months does not promote the formation of CMBs in aged APP23 mice. Nevertheless, warfarin but not dabigatran is associated with a higher risk of extensive intracerebral hemorrhage, suggesting that this model may allow preclinical safety evaluation of antithrombotic therapies.


Anticoagulants/therapeutic use , Cerebral Amyloid Angiopathy/diagnostic imaging , Cerebral Amyloid Angiopathy/drug therapy , Cerebral Hemorrhage/diagnostic imaging , Cerebral Hemorrhage/drug therapy , Microvessels/diagnostic imaging , Amyloid beta-Protein Precursor/genetics , Animals , Anticoagulants/pharmacology , Cerebral Amyloid Angiopathy/genetics , Cerebral Hemorrhage/genetics , Female , Magnetic Resonance Imaging/trends , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/drug effects , Random Allocation , Treatment Outcome
14.
J Clin Invest ; 127(8): 3152-3166, 2017 Aug 01.
Article En | MEDLINE | ID: mdl-28737512

Obesity promotes a chronic inflammatory and hypercoagulable state that drives cardiovascular disease, type 2 diabetes, fatty liver disease, and several cancers. Elevated thrombin activity underlies obesity-linked thromboembolic events, but the mechanistic links between the thrombin/fibrin(ogen) axis and obesity-associated pathologies are incompletely understood. In this work, immunohistochemical studies identified extravascular fibrin deposits within white adipose tissue and liver as distinct features of mice fed a high-fat diet (HFD) as well as obese patients. Fibγ390-396A mice carrying a mutant form of fibrinogen incapable of binding leukocyte αMß2-integrin were protected from HFD-induced weight gain and elevated adiposity. Fibγ390-396A mice had markedly diminished systemic, adipose, and hepatic inflammation with reduced macrophage counts within white adipose tissue, as well as near-complete protection from development of fatty liver disease and glucose dysmetabolism. Homozygous thrombomodulin-mutant ThbdPro mice, which have elevated thrombin procoagulant function, gained more weight and developed exacerbated fatty liver disease when fed a HFD compared with WT mice. In contrast, treatment with dabigatran, a direct thrombin inhibitor, limited HFD-induced obesity development and suppressed progression of sequelae in mice with established obesity. Collectively, these data provide proof of concept that targeting thrombin or fibrin(ogen) may limit pathologies in obese patients.


Fibrin/metabolism , Inflammation/metabolism , Obesity/metabolism , Obesity/therapy , Thrombin/metabolism , Adipose Tissue/metabolism , Adiposity , Amino Acid Motifs , Animals , Blood Glucose/metabolism , Body Composition , Body Weight , Coagulants/pharmacology , Dabigatran/pharmacology , Diet, High-Fat , Fatty Liver/metabolism , Female , Genotype , Homozygote , Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Weight Gain
15.
N Engl J Med ; 377(5): 431-441, 2017 08 03.
Article En | MEDLINE | ID: mdl-28693366

BACKGROUND: Idarucizumab, a monoclonal antibody fragment, was developed to reverse the anticoagulant effect of dabigatran. METHODS: We performed a multicenter, prospective, open-label study to determine whether 5 g of intravenous idarucizumab would be able to reverse the anticoagulant effect of dabigatran in patients who had uncontrolled bleeding (group A) or were about to undergo an urgent procedure (group B). The primary end point was the maximum percentage reversal of the anticoagulant effect of dabigatran within 4 hours after the administration of idarucizumab, on the basis of the diluted thrombin time or ecarin clotting time. Secondary end points included the restoration of hemostasis and safety measures. RESULTS: A total of 503 patients were enrolled: 301 in group A, and 202 in group B. The median maximum percentage reversal of dabigatran was 100% (95% confidence interval, 100 to 100), on the basis of either the diluted thrombin time or the ecarin clotting time. In group A, 137 patients (45.5%) presented with gastrointestinal bleeding and 98 (32.6%) presented with intracranial hemorrhage; among the patients who could be assessed, the median time to the cessation of bleeding was 2.5 hours. In group B, the median time to the initiation of the intended procedure was 1.6 hours; periprocedural hemostasis was assessed as normal in 93.4% of the patients, mildly abnormal in 5.1%, and moderately abnormal in 1.5%. At 90 days, thrombotic events had occurred in 6.3% of the patients in group A and in 7.4% in group B, and the mortality rate was 18.8% and 18.9%, respectively. There were no serious adverse safety signals. CONCLUSIONS: In emergency situations, idarucizumab rapidly, durably, and safely reversed the anticoagulant effect of dabigatran. (Funded by Boehringer Ingelheim; RE-VERSE AD ClinicalTrials.gov number, NCT02104947 .).


Antibodies, Monoclonal, Humanized/therapeutic use , Dabigatran/antagonists & inhibitors , Hemorrhage/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies/blood , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/immunology , Anticoagulants/adverse effects , Blood Coagulation/drug effects , Dabigatran/adverse effects , Dabigatran/blood , Drug Hypersensitivity , Female , Hemorrhage/chemically induced , Humans , Infusions, Intravenous , Male , Middle Aged , Prospective Studies , Thrombin Time , Thrombosis/chemically induced , Time Factors
16.
Thromb Haemost ; 117(7): 1370-1378, 2017 06 28.
Article En | MEDLINE | ID: mdl-28424818

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.


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
18.
Clin Pharmacokinet ; 56(1): 41-54, 2017 01.
Article En | MEDLINE | ID: mdl-27317414

BACKGROUND AND OBJECTIVES: Idarucizumab is an antibody fragment that specifically reverses dabigatran-mediated anticoagulation. Safety, pharmacokinetics and pharmacodynamics of idarucizumab were investigated in dabigatran-treated, middle-aged, elderly and renally impaired volunteers with characteristics similar to patients receiving anticoagulant therapy. METHODS: In this randomized, double-blind, crossover study, 46 subjects (12 middle-aged, 45-64 years; 16 elderly, 65-80 years; and 18 with mild or moderate renal impairment) received dabigatran etexilate (DE; 220 or 150 mg twice daily) for 4 days. Idarucizumab doses of 1, 2.5 and 5 g or 2 × 2.5 g 1 h apart, or placebo, were administered as a rapid (5 min) infusion ~2 h after DE at steady state. RESULTS: Dabigatran-prolonged diluted thrombin time, ecarin clotting time and activated partial thromboplastin time were reversed to baseline immediately after idarucizumab infusion in all groups. Reversal was sustained with doses ≥2.5 g. Idarucizumab was well tolerated under all conditions. No impact of age on idarucizumab pharmacokinetics was observed; however, subjects with mild or moderate renal impairment demonstrated increased exposure (up to 84 %), decreased clearance and prolonged (by up to 49 %) initial half-life of idarucizumab compared with healthy middle-aged subjects. CONCLUSIONS: Impaired renal function was associated with increased exposure and decreased clearance of idarucizumab. Idarucizumab resulted in immediate, complete and sustained reversal of dabigatran anticoagulant activity, and was safe and well tolerated in middle-aged, elderly and renally impaired volunteers. The results support the clinical use of a 5 g dose of idarucizumab. CLINICAL TRIAL REGISTRATION: http://www.clinicaltrials.gov . Unique identifier: NCT01955720.


Antibodies, Monoclonal, Humanized/pharmacology , Antithrombins/pharmacology , Blood Coagulation/drug effects , Dabigatran/pharmacology , Age Factors , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/adverse effects , Area Under Curve , Cross-Over Studies , Dose-Response Relationship, Drug , Double-Blind Method , Female , Half-Life , Humans , Kidney Function Tests , Male , Metabolic Clearance Rate , Middle Aged , Partial Thromboplastin Time , Renal Insufficiency/metabolism , Time Factors
20.
Oncotarget ; 7(51): 85291-85305, 2016 Dec 20.
Article En | MEDLINE | ID: mdl-27852034

Cancer is often associated with an increased risk of thrombotic complications which can be aggravated by treatment with chemotherapeutics such as cisplatin. Multiple lines of evidence suggest that thrombin activity promotes tumor growth and metastasis. We examined the effect of co-treatment with dabigatran etexilate, a direct thrombin inhibitor, and cisplatin using the murine ID8 ovarian cancer model. Mice receiving co-treatment with both dabigatran etexilate and low dose cisplatin had significantly smaller tumors, developed less ascites and had lower levels of circulating activated platelets and tissue factor (TF) positive microparticles than those treated with dabigatran etexilate or cisplatin alone. Co-treatment with dabigatran etexilate and cisplatin significantly decreased the number of Gr1+/CD11b+ myeloid derived suppresser cells and CD11b+/CD11c+ dendritic cells in the ascites of ID8 tumor-bearing mice. Co-treatment also significantly reduced levels of pro-tumorigenic cytokines including TGF-ß, VEGF, IL-6, IL-10, and MCP-1 in the ascites while increasing IFN-γ production by CD8+ effector T cells in the tumor ascites. These results demonstrate that co-treatment with dabigatran etexilate significantly augments the anti-tumor activity of cisplatin in ovarian tumor progression by alleviating the immunosuppressive microenvironment, suggesting that thrombin may be a potential therapeutic target for treatment of ovarian cancer.


Antithrombins/therapeutic use , Blood Platelets/immunology , Cisplatin/therapeutic use , Dabigatran/therapeutic use , Dendritic Cells/immunology , Myeloid-Derived Suppressor Cells/immunology , Ovarian Neoplasms/drug therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinogenesis , Cell Growth Processes , Cells, Cultured , Cytokines/metabolism , Cytotoxicity, Immunologic , Drug Synergism , Drug Therapy, Combination , Female , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Ovarian Neoplasms/immunology , Tumor Burden , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
...