Background

The use of prothrombin complex concentrates and the role of plasma concentration of anticoagulants in the management of bleeding in patients treated with direct oral anticoagulants are still debated. Our aim was to describe management strategies and outcomes of severe bleeding events in patients treated with direct oral anticoagulants.

Methods

We performed a prospective cohort study of 732 patients treated with dabigatran, rivaroxaban, or apixaban hospitalized for severe bleeding, included prospectively in the registry from June 2013 to November 2015.

Results

Bleeding was gastrointestinal or intracranial in 37% (212 of 732) and 24% (141 of 732) of the cases, respectively. Creatinine clearance was lower than 60 ml/min in 61% (449 of 732) of the cases. The plasma concentration of direct oral anticoagulants was determined in 62% (452 of 732) of the cases and was lower than 50 ng/ml or higher than 400 ng/ml in 9.2% (41 of 452) and in 6.6% (30 of 452) of the cases, respectively. Activated or nonactivated prothrombin complex concentrates were administered in 38% of the cases (281 of 732). Mortality by day 30 was 14% (95% CI, 11 to 16).

Conclusions

Management of severe bleeding in patients treated with direct oral anticoagulants appears to be complex. The use of prothrombin complex concentrates differs depending on bleeding sites and direct oral anticoagulant plasma concentrations. Mortality differs according to bleeding sites and was similar to previous estimates.

What We Already Know about This Topic
  • Specific reversal strategies are currently available for dabigatran and in development for the anti-Xa agents. Other strategies used to manage bleeding in patients treated with these direct oral anticoagulants include measuring plasma levels of anticoagulants and therapy with prothrombin complex concentrates. However, there is little information currently available regarding these management modalities.

What This Article Tells Us That Is New
  • In a prospective cohort registry study of 732 patients treated with direct oral anticoagulants and hospitalized for severe bleeding, bleeding sites were gastrointestinal in 37% and intracranial in 24% of the cases. Activated or nonactivated prothrombin complex concentrates were administered in 38% of the cases with a day 30 mortality of 13.5% and varied according to bleeding sites but was similar to previous reports. This report provides a detailed assessment of direct oral anticoagulant-treated patients managed in clinical settings.

DIRECT oral anticoagulants (DOACs), anti-IIa (dabigatran) or anti-Xa (rivaroxaban, apixaban, and edoxaban), are now recognized as a major step forward for patients with nonvalvular atrial fibrillation or recurrent venous thromboembolism requiring long-term anticoagulation for the prevention of thromboembolic events. The efficacy–safety ratio of DOACs is similar or even better in comparison with vitamin K antagonists (VKA) in clinical trials,1,2  and this benefit appears to be reproduced in the real world,3  especially with the reduction of approximately one half of intracranial hemorrhages compared with VKA. However, spontaneous severe bleeding still occurs at significant rates during DOAC treatment. Based on the reported experience of severe bleeding in clinical trials,4–6  a limited number of case reports, and conflicting results of prothrombin complex concentrates (PCCs) and activated PCCs in animal studies or in vitro or ex vivo studies in humans,7–10  guidelines to manage bleeding in patients treated with DOACs have been published.11,12  Owing to the lack of clinical trials until the recent introduction of specific antidotes of dabigatran (idarucizumab13 ) or xabans (andexanet alfa14 ), these guidelines mainly reflect expert opinions and have a low evidence level.

To obtain more data on the current management of these patients, we set up a large multicenter prospective observational study on a European binational basis. The present article reports the results obtained in 732 patients with major bleeding requiring hospitalization when specific antidotes were not available. The primary objective of the study was to describe the management of bleeding episodes and subsequent outcomes until day 30. The secondary objectives were to describe the typology of bleeding, to describe the clinical and biologic characteristics of the patients on admission, and to analyze how these data have influenced the choice of treatment, in particular the use of reversal agents (four-factor PCC or activated PCC).

The GIHP-NACO registry (NCT02185027) (Gestes Invasifs et Hémorragies chez les Patients Traités par les Nouveaux Anticoagulants Oraux) is a large, prospective, multicenter registry, set up by the Groupe d′Intérêt en Hémostase Périopératoire (GIHP), enrolling patients treated with a DOAC and hospitalized for spontaneous or posttraumatic bleeding or who needed urgent invasive procedures (for a list of participating centers, see the appendix) in 36 centers in university, general, and private hospitals in France and Belgium (www.ClinicalTrials.gov Identifier NCT02185027). The registry opened in June 2013 and closed in November 2015. A subsample of these data was presented at the American Society of Hematology Meeting in San Francisco, California, in 2014 and published as an abstract (Blood 2014; 124:2877). This report presents the data from patients with severe bleeding under treatment with DOACs (dabigatran, rivaroxaban, or apixaban) for indications including atrial fibrillation and treatment of deep venous thrombosis or pulmonary embolism. Patients treated with DOACs for prevention of venous thromboembolism after major orthopedic surgery were not included. The institutional review board (Comité d’Ethique des Centres d’Investigation Clinique de l’Inter-région Rhône-Alpes-Auvergne, France; institutional review board number 5891) approved the study (reference 2013-02). Oral consent was obtained from all patients or proxies. Written informed consent of the patients was not necessary according to French law regarding observational studies.

Local investigators screened hospitalized patients presenting with bleeding events. Data including demographic, clinical, laboratory tests, and treatment were documented using an electronic case report form providing immediate and continuous monitoring for completeness and accuracy (ClinInfo S.A., France).

Bleeding management was assessed by collecting the following: rates and counts of erythrocyte, plasma or platelet transfusions, the use of agents such as PCC, factor VIII inhibitor bypass activator (FEIBA®, France), recombinant activated factor VII concentrate, fibrinogen concentrates, or tranexamic acid. Three-factor PCCs were not available in the participating centers. In France, three four-factor PCCs with similar contents in factors II, VII, IX, and X and proteins C and S were used: KANOKAD® (LFB, Les Ulis, France), OCTAPLEX® (Octapharma, Boulogne-Billancourt, France), and CONFIDEX® (CSL-Behring, Marburg, Germany).15,16 

Clinical Outcome Definitions

All clinical outcomes were assessed 30 days after admission. All bleeding events were classified as major or nonmajor clinically relevant.

Major bleeding was defined as overt bleeding with any of the following: fatal bleeding and/or symptomatic bleeding in a critical area or organ such as intracranial, intraspinal, intraocular, retroperitoneal, intraarticular or pericardial, or intramuscular with compartment syndrome and/or bleeding causing a fall in hemoglobin level of 20 g/l or more or leading to transfusion of two or more units of whole blood or red cells.17 

Since all patients included in this cohort were admitted to a hospital, nonmajor bleeding fulfilled nonmajor clinically relevant criteria defined as any sign or symptom of hemorrhage that did not fit the criteria for the International Society of Thrombosis and Hemostasis (ISTH) definition of major bleeding but did meet at least one of the following criteria: (1) requiring medical intervention by a healthcare professional; (2) leading to hospitalization or increased level of care; or (3) prompting a face-to-face (i.e., not just a telephone or electronic communication) evaluation.18 

On day 30 postbleeding, patients were also evaluated for suspected major cerebral and cardiovascular events (MACCEs) after the acute bleeding event. MACCEs were defined as fatal or nonfatal cardiovascular complication events as follows: acute coronary syndrome, stroke or transient ischemic attack or systemic embolism, deep venous thrombosis or pulmonary embolism, pulmonary edema, cardiogenic shock, or any other fatal cardiovascular event as assessed by investigators. It excluded rebleeding or any new bleeding episode. The fatality rate was assessed at day 30 postbleeding.

Statistical Analysis

Descriptive results are expressed as frequency and percentage for categoric variables and were compared using the chi-square test (or Fisher exact test for small samples). P < 0.05 was considered significant. For continuous variables, statistics are expressed as median, range, interquartile range, and 95% CIs (V.13.0; Stata Corp, USA).

From June 2013 to November 2015, 732 patients treated with DOACs and admitted to 36 hospitals were consecutively included in the registry. Eligible patients were identified from different departments: emergency rooms, intensive care units, and the departments of neurology, cardiology, gastroenterology, and surgery. There were 535 patients (73%) admitted to the hospital from the emergency room, and 156 (21%) and 39 (5.3%) were admitted directly to intensive care units or to other medical or surgical departments, respectively.

The demographic and clinical characteristics of the patients are presented in table 1. Among the patients included in this analysis, 207 (28%) were treated with dabigatran, 472 (64%) were treated with rivaroxaban, and 53 (7.2%) were treated with apixaban. When patients were treated with rivaroxaban (10 mg) or dabigatran (75 mg twice daily; unlicensed therapeutic protocols in Europe for atrial fibrillation or venous thromboembolism treatment), the indication was confirmed with the local investigator. As a result, 85% of the patients included in the registry were treated for atrial fibrillation, and 34% of all patients had been treated for fewer than 6 months.

Table 1.

Demographic and Clinical Characteristics

Demographic and Clinical Characteristics
Demographic and Clinical Characteristics

Among the patients, 42% were older than 80 yr, and 69% of the patients had moderate to severe renal dysfunction. There were 26% of the patients who were also receiving other medications with antithrombotic properties such as nonsteroidal antiinflammatory drugs, aspirin, VKA, or heparin, concomitantly or within days before admission for bleeding. In 64% of the cases, patients were also treated with medications interacting with P-glycoprotein or CYP450.

The sites, types, and severity of bleeding events are summarized in table 2. Severe bleeding events were mainly major according to ISTH criteria19  (74%) and spontaneous (79%).

Table 2.

Site Type and Severity of Bleeding

Site Type and Severity of Bleeding
Site Type and Severity of Bleeding

In 94% of the cases, conventional routine hemostasis lab tests were performed. In 62% of the cases, a plasma concentration of dabigatran, rivaroxaban, or apixaban was determined on admission (table 3). The median concentrations of dabigatran, rivaroxaban, and apixaban were 162, 124, and 111 ng/ml, respectively. Among these patients, 9.2% had a plasma concentration that was lower than 50 ng/ml at the time of blood sampling. On the other hand, 6.6% of these patients had a DOAC concentration higher than 400 ng/ml, up to 3,500, 1,245, and 537 ng/ml for dabigatran, rivaroxaban, and apixaban, respectively. The median time between the last intake of DOACs and blood sampling was 12 h. In patients with a plasma concentration of less than 50 ng/ml, intracranial hemorrhage was observed in 42%, gastrointestinal bleeding was observed in 27%, and bleeding at other sites was observed in 31% of the cases. Plasma concentrations were not significantly higher in patients treated with medications interacting with P-glycoprotein or CYP450, compared to patients who were not (table 4).

Table 3.

Laboratory Results on Admission

Laboratory Results on Admission
Laboratory Results on Admission
Table 4.

Bleeding Management

Bleeding Management
Bleeding Management

Bleeding management is described in table 4. PCCs (activated or nonactivated) were more frequently administered when bleeding occurred in a critical organ (intracranial or spinal; 57%) than in other sites (28%). PCCs were used in 28% of the patients for whom DOAC plasma concentrations were not determined. PCCs were used in 39% of cases where plasma concentrations were less than 50 ng/ml, 44% with plasma concentrations between 50 and 400 ng/ml, and 50% with plasma concentrations higher than 400 ng/ml (P = 0.001). This study was not designed to address the hemostatic efficacy of PCC in specific bleeding sites. The adequacy of hemostasis provided by the PCC treatment was therefore assessed subjectively by local investigators with an ordinal scale: totally, partially, or not at all in 44, 37, and 19% of the cases, respectively.

Thirty days after bleeding, MACCEs occurred in 7.4% of the cases. On day 30, case fatality was 13.5% (95% CI, 11.0 to 16.2) with central nervous system causes in 42% of the cases (table 5).

Table 5.

30-day Outcome

30-day Outcome
30-day Outcome

Patients with gastrointestinal bleeding events were elderly (mean age, 78.6 ± 10). Of these patients, 76% had a CHA2DS2Vasc score higher than 2, 24% had a history of heart failure, 25% had a history of stroke, and 20% had previous bleeding. The mortality in patients presenting with gastrointestinal bleeding was high (11.9% 30-day mortality).

Patients with bleeding during dabigatran, rivaroxaban, or apixaban treatment have been analyzed in several substudies of pivotal trials evaluating the efficacy and safety of dabigatran, rivaroxaban, and apixaban.4,6,20–23  These studies aimed to compare the management and prognosis of major bleeding in patients treated with dabigatran or rivaroxaban versus warfarin. However, bleeding management during these treatments was not formally analyzed. Our registry aimed to describe the actual management of patients regarding the use of PCC and plasma concentration determination in a real world cohort and association with the outcome. This registry was not designed to obtain an estimation of incidence or the relative importance of bleeding in patients treated with dabigatran, rivaroxaban, or apixaban.

As observed in patients with bleeding events in phase III trials, the patients included in our registry were mainly elderly. Creatinine clearance was lower than 60 ml/min in 61% of the patients. Similar results have been previously reported.13,14  Indeed, renal function may have been altered in different ways: (1) slow deterioration of renal function before the bleeding events due to comorbidities, treatment, or an acute illness (infection or dehydration) owing to accumulation of DOACs; (2) acute renal failure associated with the bleeding event; or (3) off-label use of DOACs (not recommended when creatinine clearance is lower than 15 ml/mn for rivaroxaban, apixaban, or edoxaban and lower than 30 ml/mn for dabigatran). Monitoring renal function is recommended, at least yearly, to detect changes in renal function and adapt treatment doses, particularly in patients with previously altered renal function and elderly or frail patients.12 

A major consideration of DOACs is that no specific reversal strategy was proposed before the manufacturers made them available for clinical use. Antidotes were developed several years after the first approval of DOACs.24  To date, except for dabigatran, reversal strategies rely on nonspecific measures and on the administration of hemostatic agents, mainly activated or nonactivated PCC.12  Idarucizumab, a humanized monoclonal antibody that selectively binds dabigatran, has been recently approved.13  Andexanet alfa is a recombinant modified human factor Xa decoy protein that has been shown to reverse the inhibition of factor Xa in healthy volunteers25  and patients.14  Ciraparantag (PER977) is a small cationic and water-soluble molecule designed to bind with high affinity to oral FXa inhibitors (edoxaban, rivaroxaban, and apixaban) and to dabigatran.26 

Little information has been made available on the use and timing of specific reversal strategies in pivotal studies on DOACs.4,6,23  In these studies, few patients with major bleeding have been treated with PCCs. In the Dresden registry, among 1,082 bleeding events observed during rivaroxaban exposure, 59% were classified as ISTH minor bleeding, 35% were classified as ISTH nonmajor clinically relevant bleeding, and 66 (6.1%) patients had major bleeding treated with PCC in 9.1% of the cases.27  Our registry includes a population principally with major bleeding events that is large enough to obtain a reasonable estimate for specific management strategies and outcome.

The use of PCCs to reverse the anticoagulation action of DOACs has not yet been assessed in clinical practice but is still suggested in international and national guidelines.11,12  Only animal8,28  or healthy volunteer studies7,9,29  have assessed the efficacy of PCCs (activated or not) on different endpoints. A study of healthy volunteers compared the effect of a three-factor with a four-factor PCC on reversal of the anticoagulant effects of rivaroxaban.30  Both four-factor PCCs and three-factor PCCs partially reversed the anticoagulant effects of rivaroxaban with good tolerance and no signs of thromboembolic events. In our registry, only four-factor PCCs or FEIBA were used. The present study shows that 38.4% of the patients were treated with PCCs with a distribution between activated or nonactivated PCC that is similar in patients treated with each DOAC. The use of PCC was related to the anatomic site of the bleeding. A majority of investigators consider that the use of PCCs partially or totally contributed to cessation of bleeding.

The use of coagulation testing during the clinical development of DOACs was not used in clinical trials based on considerations they would not require monitoring because of their highly predictable pharmacokinetics and therapeutic effects. Although this position has been challenged in subsequent publications regarding dabigatran,31  specific plasma concentration techniques have been developed32  for each DOAC, in theory either to answer specific questions (i.e., compliance to treatment) or to guide periprocedural or bleeding management.11  Indeed, numerous case reports and series have shown that bleeding events could be related to the high plasma concentrations of these new agents,31,33  as already observed with other anticoagulant agents.

In the present study, the plasma concentration of DOACs was determined on admission in 62% of the patients. Among these patients, 9.2% had a plasma concentration that was lower than 50 ng/ml at the time of blood sampling. These results could in part be explained by the time between the last dose and sampling. On the other hand, 6.6% of these patients had a DOAC concentration higher than 400 ng/ml, suggesting that in several patients, DOACs could accumulate owing to renal failure or major drug interaction. Very high DOAC plasma concentrations (higher than 400 ng/ml) have been observed in previous studies, either in cases of intentional overdose cohorts34  and particularly in the context of bleeding.13,14 

In our registry, 63% of the patients were also treated with medication interacting with the pharmacokinetics of DOACs. However, DOAC plasma concentration was not significantly higher than in patients not treated with these medications. The importance of these potential interactions in explaining high DOAC plasma concentration was not explored in the present study.

In addition to these observations, the usefulness of DOAC plasma concentration determination to guide major bleeding management has never been evaluated. The present study shows that plasma concentration was positively related to the use of PCCs. Hence, plasma concentration could be of major importance in identifying patients in whom the use of an antidote or, if not available, PCC could be useful in reversing the anticoagulant effects of DOACs. However, our study does not show that this strategy lowers mortality.

Several analyses of mortality related to major bleeds have been performed from pivotal studies of DOACs. Thirty-day mortality after the first major bleed in phase III trials comparing dabigatran with warfarin was 9.1% in the dabigatran group and 13.0% in the warfarin group.23  Using data from the ROCKET AF study, the outcomes after major bleeding, including all causes of death, were similar in patients treated with rivaroxaban and warfarin (20.4 vs. 25.6% in the rivaroxaban and warfarin group, respectively).4  In the ARISTOTLE study, 8.9% with major nonintracranial bleeding died in the apixaban arm, and 9.5% died in the warfarin arm. Of those with intracranial hemorrhage, 45.3% died in the apixaban arm, and 42.3% died in the warfarin arm.6 

The outcomes of intracranial hemorrhage in the ROCKET AF and RE-LY studies have been analyzed more specifically.21,22  In the RE-LY study, 154 intracranial hemorrhages occurred in 153 subjects.22  The mortality rate ranged from 24 to 49% depending on the intracranial location of the bleeding in both the dabigatran and the warfarin groups. During ROCKET AF follow-up, 172 patients experienced 174 intracranial bleeding episodes.21  Mortality ranged from 20 to 69% depending on the intracranial location of the bleeding in both the rivaroxaban and the warfarin groups.4  In the present study, the mortality for spontaneous intracranial hemorrhage (n = 141) was 36, 28, and 17% in patients treated with dabigatran, rivaroxaban, and apixaban, respectively, with large confidence intervals.

Several factors impair the comparability with different studies on bleeding complications: history, severity, cause, and sites of bleeding. Depending on the cause of bleeding, mortality can differ greatly owing to the rate of intracranial hemorrhage, whose mortality rate is high,35  and to other anatomic sites with lower mortality.

Our study has several strengths. The majority of the bleeding events described are classified based on major ISTH criteria. A detailed clinical and biologic assessment was available for most of the patients in this real world cohort. This included the plasma concentrations of dabigatran, rivaroxaban, or apixaban in a large subset of patients. Step-by-step management of these bleeding episodes is described including the use of hemostatic agents as suggested in most guidelines.

Our study has the limitations inherent to the observational nature of the study. This impacts the distribution of the bleeding sites and the severity of the cases. Moreover, the rate of inclusion among the 36 participating centers ranged from 1 to 67 depending on the investigator’s department.

The data available in the GIHP-NACO registry provide an instant picture of the complex management of DOAC-induced major bleeding events in two European countries. Despite compliance to specific management, including the use of hemostatic agents or plasma concentration determination, mortality remains high. Specific analysis of bleeding sites is needed to assess the role of present or future (antidote) reversal strategies to improve the outcome of major bleeding in patients treated with DOACs.

This study was supported by an unrestricted grant from Octapharma (Boulogne-Billancourt, France), CSL-Behring (Marburg, Germany), and LFB (Les Ulis, France).

Dr. Albaladejo and Dr. Samama report personal fees and nonfinancial support from Bayer (Lyon, France), Boehringer Ingelheim (Paris, France), Daiichi Sankyo (Rueil-Malmaison, France), Bristol-Myers Squibb (Rueil-Malmaison, France), and Pfizer (Paris, France) outside the submitted work; Dr. Gruel reports personal fees from Boehringer Ingelheim (Paris, France) and Bristol-Myers Squibb (Rueil-Malmaison, France) and personal fees and nonfinancial support from Bayer (Lyon, France) outside the submitted work. The other authors declare no competing interests.

1.
van der Hulle
T
,
Kooiman
J
,
den Exter
PL
,
Dekkers
OM
,
Klok
FA
,
Huisman
MV
:
Effectiveness and safety of novel oral anticoagulants as compared with vitamin K antagonists in the treatment of acute symptomatic venous thromboembolism: A systematic review and meta-analysis.
J Thromb Haemost
2014
;
12
:
320
8
2.
Ruff
CT
,
Giugliano
RP
,
Braunwald
E
,
Hoffman
EB
,
Deenadayalu
N
,
Ezekowitz
MD
,
Camm
AJ
,
Weitz
JI
,
Lewis
BS
,
Parkhomenko
A
,
Yamashita
T
,
Antman
EM
:
Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: A meta-analysis of randomised trials.
Lancet
2014
;
383
:
955
62
3.
Larsen
TB
,
Skjøth
F
,
Nielsen
PB
,
Kjældgaard
JN
,
Lip
GY
:
Comparative effectiveness and safety of non-vitamin K antagonist oral anticoagulants and warfarin in patients with atrial fibrillation: Propensity weighted nationwide cohort study.
BMJ
2016
;
353
:
i3189
4.
Piccini
JP
,
Garg
J
,
Patel
MR
,
Lokhnygina
Y
,
Goodman
SG
,
Becker
RC
,
Berkowitz
SD
,
Breithardt
G
,
Hacke
W
,
Halperin
JL
,
Hankey
GJ
,
Nessel
CC
,
Mahaffey
KW
,
Singer
DE
,
Califf
RM
,
Fox
KA
;
ROCKET AF Investigators
:
Management of major bleeding events in patients treated with rivaroxaban vs. warfarin: Results from the ROCKET AF trial.
Eur Heart J
2014
;
35
:
1873
80
5.
Majeed
A
,
Meijer
K
,
Larrazabal
R
,
Arnberg
F
,
Luijckx
GJ
,
Roberts
RS
,
Schulman
S
:
Mortality in vitamin K antagonist-related intracerebral bleeding treated with plasma or 4-factor prothrombin complex concentrate.
Thromb Haemost
2014
;
111
:
233
9
6.
Held
C
,
Hylek
EM
,
Alexander
JH
,
Hanna
M
,
Lopes
RD
,
Wojdyla
DM
,
Thomas
L
,
Al-Khalidi
H
,
Alings
M
,
Xavier
D
,
Ansell
J
,
Goto
S
,
Ruzyllo
W
,
Rosenqvist
M
,
Verheugt
FW
,
Zhu
J
,
Granger
CB
,
Wallentin
L
:
Clinical outcomes and management associated with major bleeding in patients with atrial fibrillation treated with apixaban or warfarin: Insights from the ARISTOTLE trial.
Eur Heart J
2015
;
36
:
1264
72
7.
Zahir
H
,
Brown
KS
,
Vandell
AG
,
Desai
M
,
Maa
JF
,
Dishy
V
,
Lomeli
B
,
Feussner
A
,
Feng
W
,
He
L
,
Grosso
MA
,
Lanz
HJ
,
Antman
EM
:
Edoxaban effects on bleeding following punch biopsy and reversal by a 4-factor prothrombin complex concentrate.
Circulation
2015
;
131
:
82
90
8.
Martin
AC
,
Le Bonniec
B
,
Fischer
AM
,
Marchand-Leroux
C
,
Gaussem
P
,
Samama
CM
,
Godier
A
:
Evaluation of recombinant activated factor VII, prothrombin complex concentrate, and fibrinogen concentrate to reverse apixaban in a rabbit model of bleeding and thrombosis.
Int J Cardiol
2013
;
168
:
4228
33
9.
Marlu
R
,
Hodaj
E
,
Paris
A
,
Albaladejo
P
,
Cracowski
JL
,
Crackowski
JL
,
Pernod
G
:
Effect of non-specific reversal agents on anticoagulant activity of dabigatran and rivaroxaban: a randomised crossover ex vivo study in healthy volunteers.
Thromb Haemost
2012
;
108
:
217
24
10.
Lee
FM
,
Chan
AK
,
Lau
KK
,
Chan
HH
:
Reversal of new, factor-specific oral anticoagulants by rFVIIa, prothrombin complex concentrate and activated prothrombin complex concentrate: A review of animal and human studies.
Thromb Res
2014
;
133
:
705
13
11.
Pernod
G
,
Albaladejo
P
,
Godier
A
,
Samama
CM
,
Susen
S
,
Gruel
Y
,
Blais
N
,
Fontana
P
,
Cohen
A
,
Llau
JV
,
Rosencher
N
,
Schved
JF
,
de Maistre
E
,
Samama
MM
,
Mismetti
P
,
Sié
P
;
Working Group on Perioperative Haemostasis
:
Management of major bleeding complications and emergency surgery in patients on long-term treatment with direct oral anticoagulants, thrombin or factor-Xa inhibitors: Proposals of the working group on perioperative haemostasis (GIHP) - March 2013.
Arch Cardiovasc Dis
2013
;
106
:
382
93
12.
Heidbuchel
H
,
Verhamme
P
,
Alings
M
,
Antz
M
,
Diener
HC
,
Hacke
W
,
Oldgren
J
,
Sinnaeve
P
,
Camm
AJ
,
Kirchhof
P
:
Updated European Heart Rhythm Association Practical Guide on the use of non-vitamin K antagonist anticoagulants in patients with non-valvular atrial fibrillation.
Europace
2015
;
17
:
1467
507
13.
Pollack
CV
Jr
,
Reilly
PA
,
Eikelboom
J
,
Glund
S
,
Verhamme
P
,
Bernstein
RA
,
Dubiel
R
,
Huisman
MV
,
Hylek
EM
,
Kamphuisen
PW
,
Kreuzer
J
,
Levy
JH
,
Sellke
FW
,
Stangier
J
,
Steiner
T
,
Wang
B
,
Kam
CW
,
Weitz
JI
:
Idarucizumab for dabigatran reversal.
N Engl J Med
2015
;
373
:
511
20
14.
Connolly
SJ
,
Milling
TJ
Jr
,
Eikelboom
JW
,
Gibson
CM
,
Curnutte
JT
,
Gold
A
,
Bronson
MD
,
Lu
G
,
Conley
PB
,
Verhamme
P
,
Schmidt
J
,
Middeldorp
S
,
Cohen
AT
,
Beyer-Westendorf
J
,
Albaladejo
P
,
Lopez-Sendon
J
,
Goodman
S
,
Leeds
J
,
Wiens
BL
,
Siegal
DM
,
Zotova
E
,
Meeks
B
,
Nakamya
J
,
Lim
WT
,
Crowther
M
;
ANNEXA-4 Investigators
:
Andexanet alfa for acute major bleeding associated with factor Xa inhibitors.
N Engl J Med
2016
;
375
:
1131
41
15.
Ghadimi
K
,
Levy
JH
,
Welsby
IJ
:
Prothrombin complex concentrates for bleeding in the perioperative setting.
Anesth Analg
2016
;
122
:
1287
300
16.
Dickneite
G
,
Hoffman
M
:
Reversing the new oral anticoagulants with prothrombin complex concentrates (PCCs): What is the evidence?
Thromb Haemost
2014
;
111
:
189
98
17.
Schulman
S
,
Kearon
C
:
Definition of major bleeding in clinical investigations of antihemostatic medicinal products in non-surgical patients.
J Thromb Haemost
2005
;
3
:
692
4
18.
Kaatz
S
,
Ahmad
D
,
Spyropoulos
AC
,
Schulman
S
;
Subcommittee on Control of Anticoagulation
:
Definition of clinically relevant non-major bleeding in studies of anticoagulants in atrial fibrillation and venous thromboembolic disease in non-surgical patients: Communication from the SSC of the ISTH.
J Thromb Haemost
2015
;
13
:
2119
26
19.
Schulman
S
,
Carrier
M
,
Lee
AY
,
Shivakumar
S
,
Blostein
M
,
Spencer
FA
,
Solymoss
S
,
Barty
R
,
Wang
G
,
Heddle
N
,
Douketis
JD
;
Periop Dabigatran Study Group
:
Perioperative management of dabigatran: A prospective cohort study.
Circulation
2015
;
132
:
167
73
20.
Hylek
EM
,
Held
C
,
Alexander
JH
,
Lopes
RD
,
De Caterina
R
,
Wojdyla
DM
,
Huber
K
,
Jansky
P
,
Steg
PG
,
Hanna
M
,
Thomas
L
,
Wallentin
L
,
Granger
CB
:
Major bleeding in patients with atrial fibrillation receiving apixaban or warfarin: The ARISTOTLE Trial (Apixaban for Reduction in Stroke and Other Thromboembolic Events in Atrial Fibrillation): Predictors, characteristics, and clinical outcomes.
J Am Coll Cardiol
2014
;
63
:
2141
7
21.
Hankey
GJ
,
Stevens
SR
,
Piccini
JP
,
Lokhnygina
Y
,
Mahaffey
KW
,
Halperin
JL
,
Patel
MR
,
Breithardt
G
,
Singer
DE
,
Becker
RC
,
Berkowitz
SD
,
Paolini
JF
,
Nessel
CC
,
Hacke
W
,
Fox
KA
,
Califf
RM
;
ROCKET AF Steering Committee and Investigators
:
Intracranial hemorrhage among patients with atrial fibrillation anticoagulated with warfarin or rivaroxaban: The rivaroxaban once daily, oral, direct factor Xa inhibition compared with vitamin K antagonism for prevention of stroke and embolism trial in atrial fibrillation.
Stroke
2014
;
45
:
1304
12
22.
Hart
RG
,
Diener
HC
,
Yang
S
,
Connolly
SJ
,
Wallentin
L
,
Reilly
PA
,
Ezekowitz
MD
,
Yusuf
S
:
Intracranial hemorrhage in atrial fibrillation patients during anticoagulation with warfarin or dabigatran: The RE-LY trial.
Stroke
2012
;
43
:
1511
7
23.
Majeed
A
,
Hwang
HG
,
Connolly
SJ
,
Eikelboom
JW
,
Ezekowitz
MD
,
Wallentin
L
,
Brueckmann
M
,
Fraessdorf
M
,
Yusuf
S
,
Schulman
S
:
Management and outcomes of major bleeding during treatment with dabigatran or warfarin.
Circulation
2013
;
128
:
2325
32
24.
Greinacher
A
,
Thiele
T
,
Selleng
K
:
Reversal of anticoagulants: An overview of current developments.
Thromb Haemost
2015
;
113
:
931
42
25.
Siegal
DM
,
Curnutte
JT
,
Connolly
SJ
,
Lu
G
,
Conley
PB
,
Wiens
BL
,
Mathur
VS
,
Castillo
J
,
Bronson
MD
,
Leeds
JM
,
Mar
FA
,
Gold
A
,
Crowther
MA
:
Andexanet alfa for the reversal of factor Xa inhibitor activity.
N Engl J Med
2015
;
373
:
2413
24
26.
Ansell
JE
,
Bakhru
SH
,
Laulicht
BE
,
Steiner
SS
,
Grosso
M
,
Brown
K
,
Dishy
V
,
Noveck
RJ
,
Costin
JC
:
Use of PER977 to reverse the anticoagulant effect of edoxaban.
N Engl J Med
2014
;
371
:
2141
2
27.
Beyer-Westendorf
J
,
Förster
K
,
Pannach
S
,
Ebertz
F
,
Gelbricht
V
,
Thieme
C
,
Michalski
F
,
Köhler
C
,
Werth
S
,
Sahin
K
,
Tittl
L
,
Hänsel
U
,
Weiss
N
:
Rates, management, and outcome of rivaroxaban bleeding in daily care: Results from the Dresden NOAC registry.
Blood
2014
;
124
:
955
62
28.
Honickel
M
,
Treutler
S
,
van Ryn
J
,
Tillmann
S
,
Rossaint
R
,
Grottke
O
:
Reversal of dabigatran anticoagulation ex vivo: Porcine study comparing prothrombin complex concentrates and idarucizumab.
Thromb Haemost
2015
;
113
:
728
40
29.
Eerenberg
ES
,
Kamphuisen
PW
,
Sijpkens
MK
,
Meijers
JC
,
Buller
HR
,
Levi
M
:
Reversal of rivaroxaban and dabigatran by prothrombin complex concentrate: A randomized, placebo-controlled, crossover study in healthy subjects.
Circulation
2011
;
124
:
1573
9
30.
Levi
M
,
Moore
KT
,
Castillejos
CF
,
Kubitza
D
,
Berkowitz
SD
,
Goldhaber
SZ
,
Raghoebar
M
,
Patel
MR
,
Weitz
JI
,
Levy
JH
:
Comparison of three-factor and four-factor prothrombin complex concentrates regarding reversal of the anticoagulant effects of rivaroxaban in healthy volunteers.
J Thromb Haemost
2014
;
12
:
1428
36
31.
Reilly
PA
,
Lehr
T
,
Haertter
S
,
Connolly
SJ
,
Yusuf
S
,
Eikelboom
JW
,
Ezekowitz
MD
,
Nehmiz
G
,
Wang
S
,
Wallentin
L
;
RE-LY Investigators
:
The effect of dabigatran plasma concentrations and patient characteristics on the frequency of ischemic stroke and major bleeding in atrial fibrillation patients: The RE-LY Trial (Randomized Evaluation of Long-Term Anticoagulation Therapy).
J Am Coll Cardiol
2014
;
63
:
321
8
32.
Samama
MM
,
Contant
G
,
Spiro
TE
,
Perzborn
E
,
Guinet
C
,
Gourmelin
Y
,
Le Flem
L
,
Rohde
G
,
Martinoli
JL
;
Rivaroxaban Anti-Factor Xa Chromogenic Assay Field Trial Laboratories
:
Evaluation of the anti-factor Xa chromogenic assay for the measurement of rivaroxaban plasma concentrations using calibrators and controls.
Thromb Haemost
2012
;
107
:
379
87
33.
Warkentin
TE
,
Margetts
P
,
Connolly
SJ
,
Lamy
A
,
Ricci
C
,
Eikelboom
JW
:
Recombinant factor VIIa (rFVIIa) and hemodialysis to manage massive dabigatran-associated postcardiac surgery bleeding.
Blood
2012
;
119
:
2172
4
34.
Cuker
A
,
Siegal
DM
,
Crowther
MA
,
Garcia
DA
:
Laboratory measurement of the anticoagulant activity of the non-vitamin K oral anticoagulants.
J Am Coll Cardiol
2014
;
64
:
1128
39
35.
Kuramatsu
JB
,
Gerner
ST
,
Schellinger
PD
,
Glahn
J
,
Endres
M
,
Sobesky
J
,
Flechsenhar
J
,
Neugebauer
H
,
Jüttler
E
,
Grau
A
,
Palm
F
,
Röther
J
,
Michels
P
,
Hamann
GF
,
Hüwel
J
,
Hagemann
G
,
Barber
B
,
Terborg
C
,
Trostdorf
F
,
Bäzner
H
,
Roth
A
,
Wöhrle
J
,
Keller
M
,
Schwarz
M
,
Reimann
G
,
Volkmann
J
,
Müllges
W
,
Kraft
P
,
Classen
J
,
Hobohm
C
,
Horn
M
,
Milewski
A
,
Reichmann
H
,
Schneider
H
,
Schimmel
E
,
Fink
GR
,
Dohmen
C
,
Stetefeld
H
,
Witte
O
,
Günther
A
,
Neumann-Haefelin
T
,
Racs
AE
,
Nueckel
M
,
Erbguth
F
,
Kloska
SP
,
Dörfler
A
,
Köhrmann
M
,
Schwab
S
,
Huttner
HB
:
Anticoagulant reversal, blood pressure levels, and anticoagulant resumption in patients with anticoagulation-related intracerebral hemorrhage.
JAMA
2015
;
313
:
824
36

Appendix: GIHP-NACO Study Group: Severe Bleedings

Sophie Kauffmann, M.D., Jean Etienne Bazin, M.D., Ph.D., Nicolas Combaret, M.D., Anne Laure Cherprenet, M.D., Moustafa Fares, M.D., Sophie Dufraisse, M.D., Clermont-Ferrand University Hospital, Clermont-Ferrand, France (n = 67); Pierre Sié, M.D., Ph.D., Vincent Minville, M.D., Ph.D., Vincent Mémier, M.D., Toulouse University Hospital, Toulouse, France (n = 66); Pierre Morange, M.D., Ph.D., Marc Leone, M.D., Ph.D., William Cohen, M.D., Nadège Néant, M.D., Pierre Suchon, M.D., Assistance Publique- Hôpitaux de Marseille, Marseille, France (n = 64); Pierre Albaladejo, M.D., Ph.D., Gilles Pernod, M.D., Ph.D., Raphaël Marlu, M.D., Ph.D., FrançoiseCarpentier, M.D., Ph.D., Pascal Defaye, M.D., Ph.D., Grenoble University Hospital, Grenoble, France (n = 56); Patrick Mismetti, M.D., Ph.D., Alain Viallon, M.D., Ph.D., Serge Molliex, M.D., Ph.D., Julien Lanoiselée, M.D., Saint Etienne University Hospital, Saint Etienne, France (n = 55); Vincent Piriou, M.D., Ph.D., Jean Stéphane David, M.D., Ph.D., Carine Delaleu-Rague, M.D., Mathilde Lefèvre, M.D., Sophie Lagrange, M.D., Hospices Civils de Lyon, Lyon-Sud University Hospital, Pierre Benite, France (n = 48); Yves Gruel, M.D., Ph.D., Laurent Ardillon, M.D., Tours University Hospital, Tours, France (n = 46); Lorenn Bellamy, M.D., Yves Ozier, M.D., Ph.D., Brest University Hospital, Brest, France (n = 42); Emmanuel de Maistre, M.D., Ph.D., Julien Bovet, M.D., Dijon University Hospital, Dijon, France (n = 39); Annick Steib, M.D., Ph.D., Sonia Mebaouj, M.D., Louis Profumo, M.D., Strasbourg University Hospital, Strasbourg, France (n = 29); Stéphanie Roullet, M.D., Ph.D., Bordeaux University Hospital, Bordeaux, France (n = 27); David Smadja, M.D., Ph.D., Guy Meyer, M.D., Ph.D., Didier Journois, M.D., Ph.D., Assistance Publique-Hôpitaux de Paris, Georges Pompidou University Hospital, Paris, France (n = 23); Albert Trinh-Duc, M.D., Thibault Viard, M.D., Agen General Hospital, Agen, France (n = 21); Loic Belle, M.D., Ph.D., Albrice Levrat, M.D., Etienne Gautheron, M.D., Frédéric Heluwaert, M.D., Annecy Regional Hospital, Annecy, France (n = 19); Sylvie Schlumberger, M.D., Centre Médico-Chirurgical Foch, Suresnes, France (n = 18); Antonia Blanié, M.D., Dan Benhamou, M.D., Ph.D., Assistance Publique-Hôpitaux de Paris, Bicêtre University Hospital, Le Kremlin Bicêtre, France (n = 17); Charles Marc Samama, M.D., Ph.D., Claire Flaujac, M.D., Nadia Rosencher, M.D., Assistance Publique-Hôpitaux de Paris, Cochin University Hospital, Paris, France (n = 13); Hélène Beloeil, M.D., Ph.D., Catherine Blery, M.D., Lauren Daviet, M.D., Pierre Gueret, M.D., Ph.D., Rennes University Hospital, Rennes, France (n = 11); David Faraoni, M.D., Ph.D., Brugmann University Hospital, Brussels, Belgium, (n = 10); Jean François Schved, M.D., Ph.D., Pauline Deras, M.D., Samir Jaber, M.D., Ph.D., Montpellier University Hospital, Montpellier, France (n = 10); Pierre Yves Gueugniaud, M.D., Ph.D., Bernard Floccard, M.D., Hospices Civils de Lyon, Edouard Herriot University Hospital, Lyon, France (n = 8); Karim Asehnoune, M.D., Ph.D., Jérôme Paulus, M.D., Omar Amrani, M.D., Nantes University Hospital, Nantes, France (n = 7); Marie Caroline Gonthier, M.D., Marie Geneviève Huisse, M.D., Assistance Publique-Hôpitaux de Paris, Bichat University Hospital, Paris, France (n = 6); Julien Josserand, M.D., Emmanuelle de Raucourt, M.D., Assistance Publique-Hôpitaux de Paris, Beaujon University Hospital, Clichy, France (n = 5); Cécile Defournel, M.D., Alpes Léman Hospital, Contamine sur Arve, France (n = 5); Yohann Dubois, M.D., Stéphane Bouvier, M.D., Groupe Hospitalier Mutualiste, Grenoble, France (n = 4); Pierre-Marie Roy, M.D., Ph.D., Sigismond Lasocki, M.D., Ph.D., Angers University Hospital, Angers, France (n = 4); Christian Carmagnac, M.D., Catherine Chastagner, M.D., Jean Marc Thouret, M.D., Chambéry General Hospital, Chambéry, France (n = 3); Marie Antoinette Sevestre, M.D., Ph.D., Amiens University Hospital, Amiens, France (n = 3); Eric Meaudre, M.D., Pierre-Julien Cungi, M.D., Saint Anne Military Hospital, Toulon, France (n = 2); Sophie Susen, M.D., Ph.D., Lille University Hospital, Lille, France (n = 1); Jean Yves Lefrant, M.D., Ph.D., Nimes University Hospital, Nimes, France (n = 1); Stéphane Macey, M.D., Gap General Hospital, Gap, France (n = 1); Jean Baptiste Rieu, M.D., Castres General Hospital, Castres, France (n = 1); Anne Godier, M.D., Ph.D., Rotschild Foundation, Paris, France (n = 1).