Sie sind auf Seite 1von 13

J Neurol (2012) 259:212224

DOI 10.1007/s00415-011-6153-3

REVIEW

Oral anticoagulant-associated intracerebral hemorrhage


lvaro Cervera Sergio Amaro A
ngel Chamorro
A

Received: 30 May 2011 / Accepted: 16 June 2011 / Published online: 5 July 2011
Springer-Verlag 2011

Abstract The incidence of oral anticoagulation-associated intracerebral hemorrhage (OAC-ICH) is growing


due to the increasing use of warfarin and the older age of
treated patients. Recent population studies reveal that
OAC-ICH currently occurs at a frequency comparable to
that of subarachnoid hemorrhage. Most frequently, OACICH are located in deep or lobar regions of the brain,
although it may also occur in the brainstem. These
hemorrhages are larger than spontaneous hematomas and
may be fatal in at least 50% of cases. The primary cause
of brain injury in patients with OAC-ICH is the direct
mechanical disruption of the brain tissue but secondary
damage may occur through the intervention of matrix
metalloproteinases, glutamate, cytokines, heme, iron, and
the chemical toxicity of products such as thrombin, which
are released from the clot. The pathogenesis of OAC-ICH
also includes the effects of aging, the level of anticoagulation, genetic factors, and a high prevalence of concurrent cerebrovascular conditions, such as cerebral
amyloid angiopathy, leukoaraiosis or previous strokes.
The treatment of OAC-ICH is challenging and involves
rapid reversal of anticoagulation with hemostatic drug
therapies such as vitamin K, fresh frozen plasma, prothrombin complex concentrates or recombinant factor
VIIa. These therapies may not always be sufficient to
stabilize the patients clinical condition and lacking randomized controlled trials, the best hematological approach
to reverse oral anticoagulation is debated. Other difficult

. Cervera  S. Amaro  A
. Chamorro (&)
A
Comprehensive Stroke Center, Hospital Clinic, Institut
dInvestigacions Biome`diques August Pi i Sunyer (IDIBAPS),
University of Barcelona, 170 Villarroel, 08036 Barcelona, Spain
e-mail: achamorro@ub.edu

123

decisions reviewed in this article are whether anticoagulation should be restarted after OAC-ICH, and when
anticoagulant treatment should be resumed. The newer
oral anticoagulants, which are increasingly being introduced for thromboembolism prevention, may confer a
lower risk of intracranial bleeding than warfarin, although
they do not have an antidote and their anticoagulant effect
is difficult to monitor.
Keywords Intracerebral hemorrhage  Warfarin 
Oral anticoagulation  Outcome

Introduction
Warfarin is the most widely used oral anticoagulant (OAC)
in the world, although acenocoumarol, phenprocoumon or
anisindione are also frequently prescribed in several
countries. Warfarin inhibits the vitamin K-dependent posttranslational carboxylation of glutamate residues on the
N-terminal regions of coagulation factors II, VII, IX and X
by inhibiting the conversion of vitamin K 2, 3-epoxide to
reduced vitamin K [1]. The mechanism of action is comparable in the above cited OAC, being the major differences their half-lives and the duration of effect.
The benefits of OAC for thromboembolic protection are
supported by a high level of evidence in patients with
several cardiac conditions, atrial fibrillation or a history of
venous thromboembolism [2]. Nonetheless, OAC are also
notorious for having a narrow therapeutic index, numerous
drug and dietary interactions, and a significant risk of
serious bleeding that includes hemorrhagic stroke [3]. In
this review, we analyze the epidemiology, time trends,
main risk factors and pathophysiology of OAC-associated
intracerebral hemorrhage (OAC-ICH). The review also

J Neurol (2012) 259:212224

critically discusses highly controversial therapeutic decisions that are required during the early and late phases of
this devastating condition, including the pros and cons of
different hematological approaches aimed to control the
acute bleeding, the dilemma of deciding whether oral
anticoagulation should or not be restarted, and how and
when to do it in individual cases.

Epidemiology
Approximately 1% of the population in Europe is currently
receiving OAC with vitamin K antagonists, and this proportion has increased to 1.7% in some countries [4]. About
512% of ICH is related to OAC, leading to an estimated
annual incidence in the USA of nearly 3,000 [57]. In
randomized trials, the risk of major bleeding associated
with vitamin K antagonists varied according to the clinical
condition that motivated the treatment, mechanical heart
valves (18.3%), atrial fibrillation (06.6%), coronary heart
disease (019.3%), venous thromboembolism (016.7%),
or ischemic cerebrovascular disease (213%) [8]. The most
frequent complication of OAC is gastrointestinal bleeding,
but intracranial hemorrhage (ICH) is the main cause of
fatal bleeding.
In a pooled analysis of five trials using warfarin in
patients with AF the annual rate of OAC-ICH was 0.3%
[9]. The rate of OAC-ICH is about 29 per 100,000 population/year, an incidence 7- to 10-fold higher than in the
not treated population [10]. Moreover, the incidence of
OAC-ICH is increasing and ranked only slightly behind the
incidence of subarachnoid hemorrhage in recent studies
[7]. This relentless increment can be explained by the
larger number of elderly patients that receive OAC, the
increased use of combined anticoagulant regimens or the
addition of antiplatelets, or the expanded use of OAC for
secondary stroke prevention [11]. Although the increased
number of bleeding complications is counterbalanced by a
more effective prophylaxis of thromboembolic events, the
identification of those patients at higher risk of bleeding
complications must be improved.

Pathophysiology
The primary cause of brain injury in patients with ICH or
OAC-ICH is the direct mechanical disruption of the brain
tissue [12], although secondary damage frequently occurs
as the result of mass effect, midline shift and herniation
of brain structures, particularly in subjects with large
brain hemorrhages. Several molecules have been implicated in the mechanisms of brain injury after ICH

213

including matrix metalloproteinases [13], glutamate [14],


activated cytokines [15], heme [16], and iron [17]. It is
more controversial whether a reduction of cerebral blood
flow in areas surrounding the hematoma is also relevant
in patients [18, 19], as it appears to be in animal studies
[20].
An important clinical question is whether the majority of
brain damage is caused at the time of vessel rupture, or if
secondary damage may also occur through more protracted
mechanisms. The relevance of secondary damage is supported by neuroimaging and pathological studies that
identify areas of necrotic brain tissue and edema formation
around the clot, which are attributed to the direct effects of
mechanical forces and the chemical toxicity of products
such as thrombin which are released from the clot [21].
This observation has encouraged the design of clinical
trials aimed at improving outcome at expenses of limiting
secondary hematoma growth [22].
The role of thrombin in brain edema formation
The formation of brain edema around the hematoma is
attributed to the effects of increased hydrostatic pressure
[23], increased bloodbrain barrier permeability [24], and
clot retraction with movement of serum molecules from the
hematoma into the surrounding tissue [2527]. The latter
process emphasizes that intracerebral blood may itself be
toxic and contribute to secondary brain damage [12].
Thrombin is a serine protease that is released from the
hematoma and may affect the behavior of the hemorrhage.
At low concentrations, thrombin is essential to stop the
bleeding [28], but at higher concentrations, it can kill
neurons and astrocytes [29] and may facilitate perihematomal edema formation [30]. Therefore, it is plausible that
thrombin might contribute to amplify the clinical effects of
the initial bleeding. The toxic effect of blood (and
thrombin) is further supported by experimental studies
showing that the injection of blood into the brain is more
harmful than the injection of an oilwax mixture [21].
Contrarily, injection of heparinized blood results in less
perihematomal edema formation [31], and brain edema
formation is also reduced after the administration of
thrombin inhibitors [32]. Thrombin formation is inhibited
by the effect of warfarin or other related compounds [33],
and it is conceivable that a lower concentration of thrombin
at the clot in patients with OAC-ICH facilitates the formation of larger hematomas and longer bleedings. Yet, the
lower disposal of thrombin within the clot would also result
in less toxicity to the surrounding brain parenchyma.
Overall, the available clinical data highlight the predominant negative effect of larger hematoma volumes in anticoagulated patients.

123

214

J Neurol (2012) 259:212224

Risk factors of OAC-ICH

Table 1 Predisposing factors of cerebral hemorrhage in anticoagulated patients [3, 5]

Pharmacological and non pharmacological interactions

Proven risk factors


Advancing age (especially [75 years)

Many factors contribute to the variability in warfarin dose


requirements, the fluctuation in the anticoagulant response
and the risk of bleeding [34]. Warfarindrug interactions
have been shown to increase the risk of serious bleeding
[35]. In a recent retrospective study [36], warfarin interaction with at least one drug was considered the main
contributor to bleeding in almost a half of the cases.
Groups of drugs prone to interact with warfarin include
anti-fungal agents, lipid-lowering drugs, acetaminophen,
non-steroidal anti-inflammatory drugs, selective serotonin
reuptake inhibitors, amiodarone, omeprazole, fluorouracil,
and cimetidine [37]. The main mechanism of interaction
implies the inhibition of cytochrome P450 2C9 isoenzyme
(CYP2C9), which increases the plasma concentration of
S-warfarin and leads to enhanced anticoagulation and
higher bleeding risk. Also, drugs that impair platelet
function raise the risk associated with warfarin therapy
[38]. Conversely, the anticoagulant effect of warfarin could
be inhibited by drugs which increase hepatic clearance, like
barbiturates, rifampicin, and carbamazepine.
Microhemorrhages, leukoaraiosis, previous stroke
and their relationship with OAC-ICH
There are a number of predisposing factors of cerebral
hemorrhage in anticoagulated patients, as shown in
Table 1. Several brain abnormalities have also been associated with a greater risk of complications in anticoagulated patients, including microhemorrhages, leukoaraiosis,
and previous strokes [3941]. Microhemorrhages are small,
dot-like signal loss best visualized on gradient-echo magnetic resonance imaging, which are regarded as hemosiderin deposits caused by minor bleeding from advanced
microangiopathy [42, 43]. Individuals with microhemorrhages are more likely to be hypertensive, and have history
of stroke, leukoaraiosis, or old hemorrhages. In patients
with acute ischemic stroke, those with coexistent microhemorrhages more frequently develop subsequent ICH
[44]. Sporadic reports also suggest that microhemorrhages
are preferential focuses of ICH in patients receiving warfarin, most likely because the drug unmasks ICH that
would otherwise remain asymptomatic [39]. Microhemorrhages, leukoaraiosis and previous stroke frequently coexist and this may confound the interpretation of the risk of
spontaneous or drug related cerebral bleeding.
Leukoaraiosis has been independently associated with
OAC-ICH in a dose-dependent manner [40], even among
patients with an INR B 3.0 [41]. In other studies, patients
with advanced leukoaraiosis but without microhemorrhages

123

Hypertension (especially systolic blood pressure [160 mm Hg)


History of cerebrovascular disease
Intensity of anticoagulation (mainly if INR [ 4.0)
Possible risk factors
Increased variation of INR
Concomitant use of aspirin
Cerebral amyloid angiopathy
Tobacco smoking
Heavy alcohol consumption
Diabetes
Serious heart disease
Liver disease
Malignancy
Imaging and genetic markers
Leukoaraiosis detected by brain CT/MRI
Microbleeds by T2*-weighted MRI
Apo e2 or e4 genotype

tended to experience ischemic strokes rather than ICH [45].


Yet, the presence of microhemorrhages was a predictor of
ICH in patients with no or mild leukoaraiosis [45]. Recently,
leukoaraiosis was not found independently associated with
the incidence of OAC-ICH after adjustment for the presence
of microhemorrhages [46]. In light of these inconsistent
findings, the use of OAC should not be avoided in patients
with leukoaraiosis or microhemorrhages.
Prediction of OAC-ICH
Several methods have been developed to estimate the risk
for OAC-related bleeding and improve the identification of
patients in whom the risks of therapy might outweigh the
benefits [4749]. The main characteristics and predictive
value of these models are shown in Table 2. These methods
are valid but should not be used in isolation to decide
whether to initiate or not a therapy with OAC in individual
patients. Moreover, none of these methods have differentiated between the risks of intracranial or systemic bleedings, or incorporated the predictive role of INR values.
More recently, the European Society of Cardiology has
proposed a new score, the HAS-BLED (hypertension,
abnormal renal/liver function, stroke, bleeding history,
labile INR, [65 years-old, intake of drugs or alcohol) to
assess the bleeding risk in AF patients [50]. A score C3
indicates a high-risk, and caution is advised before starting
antithrombotic treatment.

J Neurol (2012) 259:212224

215

Table 2 Bleeding prediction models


Author

Follow-up
(months)

High-risk
(%)

Medium-risk
(%)

Low-risk
(%)

Variables included in models

Beyth et al. [47]

48

53

12

Age [65 years, history of GI bleeding, history of stroke, at


least one of recent AMI, renal insufficiency, anemia or
diabetes mellitus

Kuijer et al. [48]

Age C60 years, gender, malignancy, body surface area [2,


coumarin type (long vs. short acting agents)

Shireman et al. [49]

5.4

2.0

0.9

Age C70 years, gender, remote bleeding; recent bleeding,


alcohol/drug abuse, diabetes; anemia, antiplatelet use

Genetics of OAC-ICH
It has been estimated that a third of all cases of lobar ICH
are attributable to the presence of an apolipoprotein e4 or
e2 allele [51], but testing for them does not appear to
facilitate the selection of patients that can safely be treated
with warfarin [52]. Randomized clinical trials have consistently demonstrated an increased risk of bleeding among
patients in whom the target intensity of anticoagulation is
higher [3]. While a minor part of the variability in dose
requirement has been attributed to demographic and clinical factors such as age, drug interactions, concomitant
diseases, or vitamin K intake, the major contributors to the
risk of bleeding are genetic factors [53].
Genetic variants of cytochrome P450 2C9 (CYP2C9)
and vitamin K epoxide reductase (VKORC1) may influence warfarin dose and appear to modulate the risk of
bleeding, mainly at the beginning of drug intake [54, 55].
CYP2C9 encodes the enzyme cytochrome P450 2C9 that
mediates the metabolic clearance of the enantiomer
S-warfarin, which is more potent than R-warfarin, and
three different alleles termed CYP2C9*1 (wild allele),
CYP2C9*2 and CYP2C9*3 have been described. Patients
with allele variants require a lower dose of anticoagulants,
take a longer time to reach a stable dose, and are at higher
risk for over-anticoagulation and serious bleeding during
the start of treatment [54]. It has been recently suggested
that CYP2C9 genotyping could identify patients at higher
risk of bleeding [56].
VKORC1 recycles vitamin K epoxide to the reduced
form of vitamin K, an essential cofactor in the activation of
clotting factors II, VII, IX, and X [57]. VKORC1 polymorphisms account for over a 30% of the variance in the
dose of warfarin [58], and they are associated with a lower
warfarin dose during long-term therapy [55]. The increased
risk of bleeding attributable to VKORC1 is limited to the
beginning of anticoagulant therapy, while CYP2C9
sequence variants may be associated with a continued risk
of hemorrhage [59]. However, recent prospective data
show that [70% of OAC-ICH occur in patients who have
been receiving warfarin for 1 year or longer, suggesting

that interventions aimed to adjust initial warfarin dose


based on genetic information are unlikely to prevent the
majority of intracerebral bleedings [60].

Clinical aspects and prognosis of OAC-ICH


ICH is the most threatening stroke subtype, particularly in
men, with a mortality rate between 30 and 55% [61, 62].
The higher mortality risk of ICH compared with ischemic
stroke (HR, 1.56) is maintained after adjustment for age,
gender, stroke severity and cardiovascular risk factors [63].
The increased mortality risk is more relevant during the
first 3 months of stroke, and detrimental factors include
decreased level of consciousness, hemorrhage volume,
hematoma expansion, and intraventricular extension of
hemorrhage [6466]. The prognosis of OAC-ICH varied in
different studies, as shown in Table 3, although most
studies were small, retrospective, lacked a control group,
did not have clear diagnostic protocols, and the care of
patients was not standardized. Aggregated data shows that
the outcome of patients with OAC-ICH is worse than in
patients with spontaneous ICH, and that increased mortality obeys as much to a higher incidence of larger hemorrhages, as to the older age and severe comorbidity
frequently observed in patients with OAC-ICH [61, 67].
The severity of OAC-ICH is influenced by the specific
brain anatomy of the bleeding. The localization of the
hemorrhage is similar to the observed in spontaneous ICH
[68]. Thus, the relative frequency of lobar bleedings is
similar in anticoagulated and non anticoagulated patients.
OAC-ICH showed a predilection for the cerebellum in
several studies [69, 70], although this preference was not
confirmed in aggregate data [71]. OAC-ICH is reported to
be significantly larger than spontaneous ICH [66, 72],
although this is not confirmed in other studies [73]. In one
study patients with OAC-ICH showed a greater risk of
hematoma expansion, and more protracted bleedings than
patients with spontaneous ICH [73]. Higher initial systolic
BP [74] and hyperglycemia [73] were found to be associated with greater ICH expansion in some studies, although

123

123

2000

2001

[97]

[119]

[75]

[73]

[67]

[61]

[76]

[65]

[72]

[74]

Bertram

Berwaerts
Sjoblom

Flibotte

Rosand

Flaherty

Huttner

Cucchiara

Flaherty

Zukov

88

51

21

55

190

102

42

151

42

15

79

OAC-ICH
n

No

207

267

No

891

333

142

No

No

No

No

ICH control

76

69

75

69

75

76

[18

75

71

62

67

Mean age

GCS, Hyperglycemia
ICH volume expansion

4043%
(730 days)

Warfarin use
INR [ 2.1

(90 days)
UK

Hematoma growth
Age, Male gender

(1 year)
62%

Age, ICH volume,

78%

(1 day1 year)

Warfarin use

Diabetes, Lobar ICH

3366%

Warfarin use, Age,

52%

Hyperglycemia

(30 days)

Warfarin use,

53%
(90 days)

ICH volume

(30 days6 months)

ICH volume
IV extension,

SBP

Lobar hemorrhage

Initial ICH volume


Warfarin use

Higher INR

UK

UK

Warfarin use

UK

No

INR prolongation

UK

UK
UK

Predictors of ICH
expansion volume

Predictors of
mortality or
rebleeding

5464%

43%

(4 days)

20%

4667%
(230 days)

Mortality or
poor outcome
(time of assessment)

No

Only if INR [ 3

UK

UK

UK

Yes

No

No

UK

No

Correlation of
INR with ICH ICH
expansion

GCS Glasgow Coma Scale, ICH Intracerebral hemorrhage, INR International normalized ratio, IV Intraventricular, SBP Systolic blood pressure, UK Unknown

2008

2008

2008

2006

2006

2004

2004

2000

1990

[66]

Franke

Year

Ref

Study

Table 3 Prognosis in OAC-ICH in different studies

17

UK

56

27

UK

54

47

47

UK

Early
worsening
(%)

216
J Neurol (2012) 259:212224

J Neurol (2012) 259:212224

217

acute blood pressure was not predictive of mortality in a


retrospective case control study [75]. Larger hematomas
[72], and greater hematoma growth have been correlated
with higher INR values in some studies [76]. Accordingly,
excessive anticoagulation may increase morbidity and
mortality in patients with OAC-ICH [77, 78].

should be actively reversed after OAC-ICH [80]. The


requirement of OAC reversal is mandatory if surgery of the
hematoma is planned. There are different options to reverse
OAC, and usually these are combined in order to achieve a
permanent reduction in the INR (Table 5).
Discontinuation of OAC therapy

Treatment of AOC-ICH
Reversal of anticoagulation after OAC-ICH
There is scanty evidence-based information about the proper
treatment of OAC-ICH; there are no randomized clinical
trials, current therapeutic guidelines differ in their recommendations (Table 4), and expert opinions are also variable
[79]. Nevertheless, the consensus is that anticoagulation

To restore INR to its normal values is essential to stop


OAC therapy. This is not associated with a relevant
increase in thromboembolic complications, as shown in a
large retrospective series of patients with OAC-ICH [81].
In patients with mechanical heart valves, there were no
embolic events or valve thrombosis after stopping OAC
[82]. Therefore, a short discontinuation of OAC in patients
at a high embolic risk can be performed safely in most
instances.

Table 4 Guidelines on OAC reversal


Guidelines

Bleeding definition

Preferred treatment

Indications

Australasian Society of
Thrombosis and Haemostasis
[120]

Any clinical significant


bleeding

IV Vitamin K1, PCC and


FFP

Cease warfarin

British Committee for Standards in


Hematology [121]

Major bleeding

IV Vitamin K ? PCC

Reversal of anticoagulation in patients with


major bleeding requires administration of a
factor concentrate in preference to FFP (grade
B, level III), and administration of intravenous
rather than oral vitamin K (grade B, level IIa)

European Stroke Initiative [122]

OAC-associated ICH

IV Vitamin K ? PCC or
FFP

AHA/ASA [123]

Warfarin-associated ICH

Vitamin K ? PCC or factor


IX complex or rFVIIa

In patients with OAC-associated ICH and


INR [ 1.4, OAC should be discontinued, and
the INR should be normalized with PCC or
FFP. Intravenous vitamin K should be added
(Class IV evidence)
Patients with warfarin-associated ICH should be
treated with IV Vitamin K to reverse the effects
of warfarin and with treatment to replace
clotting factors (Class I, Level of Evidence B)

IV Vitamin K1 510 mg
Prothrombinex (2550 IU/kg) and FFP
(150300 mL)

PCC, factor IX complex concentrate, and rFVIIa


normalize the laboratory elevation of the INR
very rapidly and with lower volumes of fluid
than FFP but with greater potential of
thromboembolism. FFP is another potential
choice but is associated with greater volumes
and much longer infusion times (Class IIb,
Level of Evidence B)
ACCP [124]

Life-threatening
bleeding

Vitamin K ? FFP, PCC or


rFVIIa

In patients with life-threatening bleeding (e.g,


intracranial hemorrhage) and elevated INR,
regardless of the magnitude of the elevation,
we recommend holding warfarin therapy and
administering FFP, PCC, or rFVIIa
supplemented with vitamin K, 10 mg by slow
IV infusion, repeated, if necessary, depending
on the INR (Grade 1C)

123

218

J Neurol (2012) 259:212224

Table 5 Recommendations for use of specific reversal agents in OAC-ICH [78, 93]
Treatment

Dose

Timing of OAC reversal

Special considerations

Discontinuing OAC therapy

514 days

Vitamin K

2.510 mg IV over 30 min

224 h

Anaphylactoid reaction

Fresh frozen plasma

15 mL/kg

Infusion 36 h

Volume overload

Prothrombin complex concentrate

25100 UI/kg

Reversal 1232 h
Infusion 10 min to 1 h
Reversal 15 min

Recombinant factor VIIa

1090 lg/kg

Bolus injection
Reversal 15 min

Vitamin K
Vitamin K slowly reverts INR to its normal values, needing
224 h to be effective. All patients with OAC-ICH must be
given vitamin K. Otherwise, INR will not be corrected
completely and a rebound coagulopathy might develop
[83]. Vitamin K should be administered IV, because the
effect is too slow using the oral route. The usual doses and
infusion time are shown in Table 5.
Fresh frozen plasma
Fresh frozen plasma (FFP) has been used for many years as
the main therapy for OAC reversal, and is still the most
frequent treatment in North America. The administration of
FFP rapidly restores clotting factor levels, although rapid
correction of the INR has not been shown to improve
mortality and morbidity [84]. FFP is given IV at a dose of
15 mL/kg, although a lower dose can be used, and it
requires the concomitant administration of vitamin K. FFP
has some important disadvantages: the administration can
suffer delays due to thawing and preparation, the risk of
volume overload has to be considered in older patients, and
there is a small risk of viral transmission, passive alloimmune thrombocytopenia, anaphylactoid reactions and septicemia [85].
Prothrombin complex concentrates
Prothrombin complex concentrates (PCC) are prepared
from pooled plasma that is virally inactivated and contains
vitamin K-dependent factors. PCC is the most widely OAC
reversal agent used in Europe. There are two main types of
PCC: the three-factor concentrates contain therapeutic
amounts of factors II, IX, and X, while the four-factor
concentrates additionally contain factor VII. [86]. PCC are
given as shown in Table 5, although some authors defend
dose adjustment based on baseline INR [87]. The INR has

123

Three-factor concentrates may not


adequately correct INR, limited
availability, cost, variable cofactor
content based on manufacturer,
potentially prothrombotic
Short half-life, cost, potentially
prothrombotic, uncertain safety

to be monitored within 15 min of the dosage [88], and


vitamin K should be added to avoid a rebound coagulopathy [83]. PPC are administered more quickly than FFP, as
they not require blood checking or thawing, and there is no
risk of volume overload [86]. INR reversal with PPT is
very fast, and it may prevent hematoma enlargement and
poor outcome after OAC-ICH [89]. The main disadvantage
of PCC is a higher cost. In some patients it is not possible
to lower INR despite the administration of vitamin K and
factor supplementation. In these cases, liver disease, or
some coagulopathy have to be suspected [83].
Recombinant factor VIIa
Recombinant factor VIIa (rFVIIa) is able to reverse anticoagulation in patients with OAC-ICH and yields a very
rapid normalization of the INR [90, 91]. However, due to
its short half-life, a rebound increase in INR may occur.
The combination of rFVIIa and FFP achieves a better
correction of INR, and reduces the total dose of FFP needed [92], although this effect has not been reflected in a
better outcome. rFVIIa is also costly, and may increase the
risk of thromboembolic events [22].
PCC has been shown in some [80, 93, 94] but not all
[75, 76] studies to be more effective than FFP to reverse
the INR. In an experimental model of OAC-ICH in mice
PPC and FFP were similarly able to prevent hematoma
growth, and they were more effective than rFVIIa [95].
Yet, based on the available evidence, PCC plus IV vitamin
K should be the treatment of choice for most patients with
OAC-ICH [96].
Risk/benefits of early anticoagulation in patients
with OAC-ICH
A therapeutic dilemma arises when a patient who requires
full-dose anticoagulation for high-risk of thromboembolism is admitted with OAC-ICH. As shown in Table 6, the

J Neurol (2012) 259:212224


Table 6 Main results of early
versus late anticoagulation with
full-dose heparin or warfarin in
patients with OAC-ICH

Early anticoagulation (AC) was


restarted within 72 h and in
many of these patients within
24 h of stroke onset; late AC
was restarted after day 14

219

Author

Anticoagulation

Rebleeding

Embolism

Lieberman et al. [98]

Early

Kawamata et al. [68]

12

Early

Nagakawa [101]

Early

Leker and Abramsky [100]

Early

Butler and Tait [99]

13

Early

Bertram et al. [97]

15

Early

Phan et al. [81]

34

Late

Summary of findings

83

5 (6.0%)

9 (10.8%)

Early AC

49

5 (10.2%)

6 (12.2%)

Late AC

34

3 (8.8%)

database addressing this difficult issue is rather small.


Bertram and colleges [97] retrospectively studied 15
patients with serious cardiac conditions and ICH which
occurred under anticoagulation. In all instances, INR normalization was attempted as early as possible and, therefore, patients received full-dose intravenous or low-dose
subcutaneous heparin. All patients that achieved a 1.5- to
2-fold elevation in activated partial thromboplastin time
(aPTT) after normalization of the INR were discharged
without complications. On the other hand, patients with
incomplete correction of the INR experienced rebleeding
within 3 days, and patients with normalized INR and no
significant increase in aPTT developed cerebral embolism.
Small retrospective studies also showed with few exceptions [98, 99] that none of the patients experienced rebleeding or embolic events after initiation of full-dose
heparin within 3672 h of OAC-ICH onset [100, 101]. In
one study [68], early resumption of anticoagulation did not
cause intracranial rebleeding even in patients that undergo
early surgery of the hematoma.
A retrospective series of 141 patients at the Mayo Clinic
showed that the risk of having an ischemic stroke after
discontinuation of warfarin therapy within 30 days of the
ICH was less than 5%, although the mortality was 48%
[81]. All embolic events occurred within the first 5 days
after warfarin discontinuation. Of the 34 patients in whom
anticoagulation therapy was restarted by day 14, none had
recurrence of ICH during hospitalization. Tinker and Tarhan [102] also observed in 159 patients with mechanical
heart valves undergoing elective surgery that none of the
patients had in-hospital thromboembolic complications
after discontinuing warfarin therapy.
Based on this small data base, the early initiation of fulldose anticoagulation in patients with OAC-ICH cannot be
recommended (or opposed). While early initiation of fulldose anticoagulation was not followed in these studies by a
clear reduction of the risk of embolism, the low statistical
power of the studies cannot rule out significant differences.
Moreover, few patients had the biological effects of

heparin adequately monitored in these studies [97], which


is a crucial step for heparin safety [103]. Also, weightadjusted heparin has been used safely in other bleeding
conditions including hemorrhagic stroke [104, 105] or
cerebral venous infarctions [106, 107]. Lacking randomized clinical trials to establish the value of full anticoagulation in patients with OAC-ICH, therapeutic decisions
should be consider case by case. Yet, clinicians should be
aware that full anticoagulation must not be started before
stable blockade of warfarin effects. The greater safety of
weight-adjusted anticoagulant nomograms, and the
importance of strict monitoring of the biological effects of
heparin have also to be considered [108].
Long-term secondary stroke prevention after OAC-ICH
Another difficult decision in clinical practice is whether
anticoagulants should be restarted and maintained indefinitely in patients with a history of OAC-ICH. Stroke prevention in this situation needs to balance the risk/benefit of
different antithrombotic options and the estimated risk of
CNS bleeding recurrence. To this aim, an important step is
to establish the most likely cause of the bleeding. Whereas
hypertensive vasculopathy appears to be the most important mechanism for ICH in deep hemispheric regions of the
brain, cerebral amyloid angiopathy, may be the most
common underlying pathophysiology for lobar ICH [51,
109]. The risk of recurrent hypertensive ICH can be
decreased by an adequate control of hypertension [110],
whereas cerebral amyloid angiopathy lacks any known
treatment. In a prospective study of elderly patients who
survived lobar ICH, recurrent ICH occurred in 22% at
2 years [111]. The rate of recurrent ICH in survivors of
deep hemispheric ICH was estimated to be 2.1% per
patient-year [112]. Therefore, in patients with lobar hemorrhage and major sources of embolism, decision analysis
models based on retrospective data suggest that the strategy
of do not anticoagulate appears robust [112]. Contrarily,
the risks and benefits of anticoagulation are more closely

123

220

balanced when applied to patients with deep hemispheric


ICH. In the latter case, oral anticoagulation might be justified if the estimated risk of ischemic stroke is high.
New oral anticoagulants
Dabigatran is a potent, direct, competitive inhibitor of
thrombin that does not require regular monitoring [113]. In
the RE-LY trial (Randomized Evaluation of Long-Term
Anticoagulation Therapy) two fixed doses of dabigatran
(110 or 150 mg, twice daily) administered in a blinded
manner were compared to open-label use of warfarin in
18,113 patients with AF [114]. Both doses were non-inferior to warfarin, and the 150 mg dose was shown to be
superior to warfarin (RR 0.66, 95% CI 0.530.82). Hemorrhagic stroke happened in 0.38% per year with warfarin,
0.12% per year with 110 mg dabigatran, and 0.10% per
year with 150 mg dabigatran. The conclusion of this trial
was that both doses of dabigatran were non-inferior to
warfarin in the prevention of stroke or systemic embolism.
Moreover, the dose of 150 mg was superior to warfarin for
embolic prevention, and the dose of 110 mg produced less
hemorrhagic events. Therefore, the authors suggested that
the dose of dabigatran could potentially be tailored to take
into consideration the risk characteristics of a specific
patient [114]. Nevertheless, it has to be taken in consideration that the number of patients needed to be treated
with dabigatran at a dose of 150 mg to prevent one nonhemorrhagic stroke, in comparison to warfarin, is approximately 357 [115]. There is no antidote to dabigatran, but in
case of life-threatening hemorrhage, administration of
rFVIIa or PCC can be considered [116].
Another kind of OACs are the factor Xa inhibitors, such
as rivaroxaban and apixaban. There is an unpublished study
that shows the non-inferiority of rivaroxaban versus warfarin to prevent ischemic events in patients with AF
(ROCKET-AF study) with less ICH (HR 0.67) and fatal
bleeding [117]. On the other hand, in the AVERROES
study, apixaban was superior to aspirin for thromboembolic
prevention in patients with AF unsuitable for warfarin
treatment, with a similar risk of ICH [118]. As it occurs
with dabigatran, there is not a known antidote for apixaban
or rivaroxaban.
Concluding remarks
The benefits of warfarin are well established in many
patients with AF and other prevalent clinical conditions.
Recent clinical trials have shown very encouraging results
using new OAC agents [114] and this new knowledge
anticipates that warfarin might not be in the short future the
most frequently used OAC. However, a predictable wider
use of OAC also foretells that the incidence of OAC-ICH

123

J Neurol (2012) 259:212224

will remain a sizeable clinical problem. Therefore, several


diagnostic and therapeutic questions remain to be clarified
in patients with this condition, including the best tools to
recognize the individuals at greater risk of bleeding, the
value of pharmacogenetics, and the most appropriate hemostatic therapy.
Acknowledgments Search strategy and selection criteria
References for this Review were identified through searches of
PubMed by use of the search terms warfarin or oral anticoagulation and intracranial hemorrhage, from December 1986 to
February 2011. Only papers in English were reviewed in detail.
Further references were obtained from the bibliographies of the
papers identified through our searches. The final reference list was
generated on the basis of relevance to the topic of this Review.
Chamorro has given lectures and received
Conflict of interest A
consultancy fees from Servier, Sanofi-Synthelabo, Takeda, Bristol Cervera has received
Myers Squibb and Boehringer Ingelheim. A
consultancy fees from Boehringer Ingelheim. S Amaro declares to
have no conflict of interests.

References
1. Whitlon DS, Sadowski JA, Suttie JW (1978) Mechanism of
coumarin action: significance of vitamin K epoxide reductase
inhibition. Biochemistry 17(8):13711377
2. Albers GW, Amarenco P, Easton JD, Sacco RL, Teal P (2008)
Antithrombotic and thrombolytic therapy for ischemic stroke:
American College of Chest Physicians evidence-based clinical
practice guidelines (8th Edition). Chest 133(6 Suppl):630S669S
3. Schulman S, Beyth RJ, Kearon C, Levine MN (2008) Hemorrhagic complications of anticoagulant and thrombolytic treatment: American College of Chest Physicians evidence-based
clinical practice guidelines (8th Edition). Chest 133(6 Suppl):
257S298S
4. Schurgers LJ, Aebert H, Vermeer C, Bultmann B, Janzen J
(2004) Oral anticoagulant treatment: friend or foe in cardiovascular disease? Blood 104(10):32313232
5. Hart RG, Tonarelli SB, Pearce LA (2005) Avoiding central
nervous system bleeding during antithrombotic therapy: recent
data and ideas. Stroke 36(7):15881593
6. Hart RG, Boop BS, Anderson DC (1995) Oral anticoagulants
and intracranial hemorrhage. Facts and hypotheses. Stroke
26(8):14711477
7. Flaherty ML, Kissela B, Woo D, Kleindorfer D, Alwell K, Sekar
P, Moomaw CJ, Haverbusch M, Broderick JP (2007) The
increasing incidence of anticoagulant-associated intracerebral
hemorrhage. Neurology 68(2):116121
8. Levine MN, Raskob G, Beyth RJ, Kearon C, Schulman S (2004)
Hemorrhagic complications of anticoagulant treatment: the
Seventh ACCP Conference on antithrombotic and thrombolytic
therapy. Chest 126(3 Suppl):287S310S
9. (1994) Risk factors for stroke and efficacy of antithrombotic
therapy in atrial fibrillation. Analysis of pooled data from five
randomized controlled trials. Arch Intern Med 154 (13):
14491457
10. Steiner T, Rosand J, Diringer M (2006) Intracerebral hemorrhage associated with oral anticoagulant therapy: current practices and unresolved questions. Stroke 37(1):256262
11. Smith NL, Psaty BM, Furberg CD, White R, Lima JA, Newman
AB, Manolio TA (1999) Temporal trends in the use of

J Neurol (2012) 259:212224

12.
13.

14.

15.
16.

17.

18.

19.

20.
21.

22.

23.

24.

25.

26.

27.

28.

29.

30.

anticoagulants among older adults with atrial fibrillation. Arch


Intern Med 159(14):15741578
Xi G, Keep RF, Hoff JT (2006) Mechanisms of brain injury after
intracerebral haemorrhage. Lancet Neurol 5(1):5363
Rosenberg GA, Navratil M (1997) Metalloproteinase inhibition
blocks edema in intracerebral hemorrhage in the rat. Neurology
48(4):921926
Castillo J, Davalos A, Alvarez-Sabin J, Pumar JM, Leira R, Silva
Y, Montaner J, Kase CS (2002) Molecular signatures of brain
injury after intracerebral hemorrhage. Neurology 58(4):624629
Wang J, Dore S (2007) Inflammation after intracerebral hemorrhage. J Cereb Blood Flow Metab 27(5):894908
Figueiredo RT, Fernandez PL, Mourao-Sa DS, Porto BN, Dutra
FF, Alves LS, Oliveira MF, Oliveira PL, Graca-Souza AV,
Bozza MT (2007) Characterization of heme as activator of Tolllike receptor 4. J Biol Chem 282(28):2022120229
Kruman I, Bruce-Keller AJ, Bredesen D, Waeg G, Mattson MP
(1997) Evidence that 4-hydroxynonenal mediates oxidative
stress-induced neuronal apoptosis. J Neurosci 17(13):50895100
Zazulia AR, Diringer MN, Videen TO, Adams RE, Yundt K,
Aiyagari V, Grubb RL Jr, Powers WJ (2001) Hypoperfusion
without ischemia surrounding acute intracerebral hemorrhage.
J Cereb Blood Flow Metab 21(7):804810
Mayer SA, Lignelli A, Fink ME, Kessler DB, Thomas CE,
Swarup R, Van Heertum RL (1998) Perilesional blood flow and
edema formation in acute intracerebral hemorrhage: a SPECT
study. Stroke 29(9):17911798
Ropper AH, Zervas NT (1982) Cerebral blood flow after experimental basal ganglia hemorrhage. Ann Neurol 11(3):266271
Suzuki J, Ebina T (1980) Sequential changes in tissue surrounding ICH. In: Pia HWLC, Zierski J (eds) Spontaneous
intracerebral hematomas. Springer, Berlin, pp 121128
Mayer SA, Brun NC, Begtrup K, Broderick J, Davis S, Diringer
MN, Skolnick BE, Steiner T (2008) Efficacy and safety of
recombinant activated factor VII for acute intracerebral hemorrhage. N Engl J Med 358(20):21272137
Katayama Y, Becker DP, Tamura T, Ikezaki K (1990) Early
cellular swelling in experimental traumatic brain injury: a phenomenon mediated by excitatory amino acids. Acta Neurochir
51:271273
Yang GY, Betz AL, Chenevert TL, Brunberg JA, Hoff JT (1994)
Experimental intracerebral hemorrhage: relationship between
brain edema, blood flow, and blood-brain barrier permeability in
rats. J Neurosurg 81(1):93102
Wagner KR, Xi G, Hua Y, Kleinholz M, de Courten-Myers GM,
Myers RE, Broderick JP, Brott TG (1996) Lobar intracerebral
hemorrhage model in pigs: rapid edema development in perihematomal white matter. Stroke 27(3):490497
Brott T, Broderick J, Barsan W, Kothari R, Tomsick T, Spilker
J, Huster G (1992) Hyper-acute clot retraction in spontaneous
intracerebral hemorrhage. Stroke 23:141
Gebel JM, Brott TG, Sila CA, Tomsick TA, Jauch E, Salisbury
S, Khoury J, Miller R, Pancioli A, Duldner JE, Topol EJ,
Broderick JP (2000) Decreased perihematomal edema in
thrombolysis-related intracerebral hemorrhage compared with
spontaneous intracerebral hemorrhage. Stroke 31(3):596600
Hua Y, Keep RF, Hoff JT, Xi G (2007) Brain injury after
intracerebral hemorrhage: the role of thrombin and iron. Stroke
38(2 Suppl):759762
Xi G, Reiser G, Keep RF (2003) The role of thrombin and
thrombin receptors in ischemic, hemorrhagic and traumatic
brain injury: deleterious or protective? J Neurochem 84(1):39
Lee KR, Colon GP, Betz AL, Keep RF, Kim S, Hoff JT (1996)
Edema from intracerebral hemorrhage: the role of thrombin.
J Neurosurg 84(1):9196

221
31. Lee KR, Betz AL, Keep RF, Chenevert TL, Kim S, Hoff JT
(1995) Intracerebral infusion of thrombin as a cause of brain
edema. J Neurosurg 83(6):10451050
32. Kitaoka T, Hua Y, Xi G, Hoff JT, Keep RF (2002) Delayed
argatroban treatment reduces edema in a rat model of intracerebral hemorrhage. Stroke 33(12):30123018
33. Brodin E, Seljeflot I, Arnesen H, Hurlen M, Appelbom H,
Hansen JB (2009) Endogenous thrombin potential (ETP) in
plasma from patients with AMI during antithrombotic treatment.
Thromb Res 123(4):573579
34. Holbrook AM, Pereira JA, Labiris R, McDonald H, Douketis
JD, Crowther M, Wells PS (2005) Systematic overview of
warfarin and its drug and food interactions. Arch Intern Med
165(10):10951106
35. Gasse C, Hollowell J, Meier CR, Haefeli WE (2005) Drug
interactions and risk of acute bleeding leading to hospitalisation
or death in patients with chronic atrial fibrillation treated with
warfarin. Thromb Haemost 94(3):537543
36. Jonsson AK, Spigset O, Jacobsson I, Hagg S (2007) Cerebral
haemorrhage induced by warfarin: the influence of drugdrug
interactions. Pharmacoepidemiol Drug Saf 16(3):309315
37. Hirsh J, Fuster V, Ansell J, Halperin JL (2003) American Heart
Association/American College of Cardiology Foundation guide
to warfarin therapy. Circulation 107(12):16921711
38. Battistella M, Mamdami MM, Juurlink DN, Rabeneck L, Laupacis A (2005) Risk of upper gastrointestinal hemorrhage in
warfarin users treated with nonselective NSAIDs or COX-2
inhibitors. Arch Intern Med 165(2):189192
39. Lee GH, Kwon SU, Kang DW (2008) Warfarin-induced intracerebral hemorrhage associated with microbleeds. J Clin Neurol
(Seoul, Korea) 4(3):131133
40. The Stroke Prevention in Reversible Ischemia Trial (SPIRIT)
Study Group (1997) A randomized trial of anticoagulants versus
aspirin after cerebral ischemia of presumed arterial origin. Ann
Neurol 42(6):857865
41. Smith EE, Rosand J, Knudsen KA, Hylek EM, Greenberg SM
(2002) Leukoaraiosis is associated with warfarin-related hemorrhage following ischemic stroke. Neurology 59(2):193197
42. Tanaka A, Ueno Y, Nakayama Y, Takano K, Takebayashi S
(1999) Small chronic hemorrhages and ischemic lesions in
association with spontaneous intracerebral hematomas. Stroke
30(8):16371642
43. Fazekas F, Kleinert R, Roob G, Kleinert G, Kapeller P, Schmidt
R, Hartung HP (1999) Histopathologic analysis of foci of signal
loss on gradient-echo T2*-weighted MR images in patients with
spontaneous intracerebral hemorrhage: evidence of microangiopathy-related microbleeds. AJNR 20(4):637642
44. Fan YH, Zhang L, Lam WW, Mok VC, Wong KS (2003)
Cerebral microbleeds as a risk factor for subsequent intracerebral hemorrhages among patients with acute ischemic stroke.
Stroke 34(10):24592462
45. Naka H, Nomura E, Takahashi T, Wakabayashi S, Mimori Y,
Kajikawa H, Kohriyama T, Matsumoto M (2006) Combinations
of the presence or absence of cerebral microbleeds and advanced
white matter hyperintensity as predictors of subsequent stroke
types. AJNR 27(4):830835
46. Lee SH, Ryu WS, Roh JK (2009) Cerebral microbleeds are a
risk factor for warfarin-related intracerebral hemorrhage. Neurology 72(2):171176
47. Beyth RJ, Quinn LM, Landefeld CS (1998) Prospective evaluation of an index for predicting the risk of major bleeding in
outpatients treated with warfarin. Am J Med 105(2):9199
48. Kuijer PM, Hutten BA, Prins MH, Buller HR (1999) Prediction
of the risk of bleeding during anticoagulant treatment for venous
thromboembolism. Arch Intern Med 159(5):457460

123

222
49. Shireman TI, Mahnken JD, Howard PA, Kresowik TF, Hou Q,
Ellerbeck EF (2006) Development of a contemporary bleeding
risk model for elderly warfarin recipients. Chest
130(5):13901396
50. Camm AJ, Kirchhof P, Lip GY, Schotten U, Savelieva I, Ernst
S, Van Gelder IC, Al-Attar N, Hindricks G, Prendergast B,
Heidbuchel H, Alfieri O, Angelini A, Atar D, Colonna P, De
Caterina R, De Sutter J, Goette A, Gorenek B, Heldal M, Hohloser SH, Kolh P, Le Heuzey JY, Ponikowski P, Rutten FH
(2010) Guidelines for the management of atrial fibrillation: the
task force for the management of atrial fibrillation of the
European Society of Cardiology (ESC). Eur Heart J
31(19):23692429
51. Woo D, Sauerbeck LR, Kissela BM, Khoury JC, Szaflarski JP,
Gebel J, Shukla R, Pancioli AM, Jauch EC, Menon AG, Deka R,
Carrozzella JA, Moomaw CJ, Fontaine RN, Broderick JP (2002)
Genetic and environmental risk factors for intracerebral hemorrhage: preliminary results of a population-based study. Stroke
33(5):11901195
52. Eckman MH, Wong LK, Soo YO, Lam W, Yang SR, Greenberg
SM, Rosand J (2008) Patient-specific decision-making for
warfarin therapy in nonvalvular atrial fibrillation: how will
screening with genetics and imaging help? Stroke
39(12):33083315
53. Loebstein R, Yonath H, Peleg D, Almog S, Rotenberg M, Lubetsky A, Roitelman J, Harats D, Halkin H, Ezra D (2001)
Interindividual variability in sensitivity to warfarinNature or
nurture? Clin Pharmacol Ther 70(2):159164
54. Aithal GP, Day CP, Kesteven PJ, Daly AK (1999) Association
of polymorphisms in the cytochrome P450 CYP2C9 with warfarin dose requirement and risk of bleeding complications.
Lancet 353(9154):717719
55. Rieder MJ, Reiner AP, Gage BF, Nickerson DA, Eby CS,
McLeod HL, Blough DK, Thummel KE, Veenstra DL, Rettie
AE (2005) Effect of VKORC1 haplotypes on transcriptional
regulation and warfarin dose. N Engl J Med 352(22):22852293
56. Klein TE, Altman RB, Eriksson N, Gage BF, Kimmel SE, Lee
MT, Limdi NA, Page D, Roden DM, Wagner MJ, Caldwell MD,
Johnson JA (2009) Estimation of the warfarin dose with clinical
and pharmacogenetic data. N Engl J Med 360(8):753764
57. Li T, Chang CY, Jin DY, Lin PJ, Khvorova A, Stafford DW
(2004) Identification of the gene for vitamin K epoxide reductase. Nature 427(6974):541544
58. Wadelius M, Chen LY, Downes K, Ghori J, Hunt S, Eriksson N,
Wallerman O, Melhus H, Wadelius C, Bentley D, Deloukas P
(2005) Common VKORC1 and GGCX polymorphisms associated with warfarin dose. Pharmacogenomics J 5(4):262270
59. Schwarz UI, Ritchie MD, Bradford Y, Li C, Dudek SM, FryeAnderson A, Kim RB, Roden DM, Stein CM (2008) Genetic
determinants of response to warfarin during initial anticoagulation. N Engl J Med 358(10):9991008
60. Genes for Cerebral Hemorrhage on Anticoagulation (GOCHA)
Collaborative Group (2009) Exploiting common genetic variation to make anticoagulation safer. Stroke 40(3 Suppl):S64S66
61. Flaherty ML, Haverbusch M, Sekar P, Kissela B, Kleindorfer D,
Moomaw CJ, Sauerbeck L, Schneider A, Broderick JP, Woo D
(2006) Long-term mortality after intracerebral hemorrhage.
Neurology 66(8):11821186
62. Foulkes MA, Wolf PA, Price TR, Mohr JP, Hier DB (1988) The
Stroke Data Bank: design, methods, and baseline characteristics.
Stroke 19(5):547554
63. Andersen KK, Olsen TS, Dehlendorff C, Kammersgaard LP
(2009) Hemorrhagic and ischemic strokes compared: stroke
severity, mortality, and risk factors. Stroke 40(6):20682072
64. Leira R, Davalos A, Silva Y, Gil-Peralta A, Tejada J, Garcia M,
Castillo J (2004) Early neurologic deterioration in intracerebral

123

J Neurol (2012) 259:212224

65.

66.

67.

68.

69.

70.

71.
72.

73.

74.

75.

76.

77.

78.

79.

80.

81.

hemorrhage: predictors and associated factors. Neurology


63(3):461467
Cucchiara B, Messe S, Sansing L, Kasner S, Lyden P (2008)
Hematoma growth in oral anticoagulant related intracerebral
hemorrhage. Stroke 39(11):29932996
Franke CL, de Jonge J, van Swieten JC, Op de Coul AA, van
Gijn J (1990) Intracerebral hematomas during anticoagulant
treatment. Stroke 21(5):726730
Rosand J, Eckman MH, Knudsen KA, Singer DE, Greenberg
SM (2004) The effect of warfarin and intensity of anticoagulation on outcome of intracerebral hemorrhage. Arch Intern Med
164(8):880884
Kawamata T, Takeshita M, Kubo O, Izawa M, Kagawa M,
Takakura K (1995) Management of intracranial hemorrhage
associated with anticoagulant therapy. Surg Neurol
44(5):438442 (discussion 443)
Kase CS, Robinson RK, Stein RW, DeWitt LD, Hier DB, Harp
DL, Williams JP, Caplan LR, Mohr JP (1985) Anticoagulantrelated intracerebral hemorrhage. Neurology 35(7):943948
Flaherty ML, Haverbusch M, Sekar P, Kissela BM, Kleindorfer
D, Moomaw CJ, Broderick JP, Woo D (2006) Location and
outcome of anticoagulant-associated intracerebral hemorrhage.
Neurocrit Care 5(3):197201
Hart RG (2000) What causes intracerebral hemorrhage during
warfarin therapy? Neurology 55(7):907908
Flaherty ML, Tao H, Haverbusch M, Sekar P, Kleindorfer D,
Kissela B, Khatri P, Stettler B, Adeoye O, Moomaw CJ, Broderick JP, Woo D (2008) Warfarin use leads to larger intracerebral hematomas. Neurology 71(14):10841089
Flibotte JJ, Hagan N, ODonnell J, Greenberg SM, Rosand J
(2004) Warfarin, hematoma expansion, and outcome of intracerebral hemorrhage. Neurology 63(6):10591064
Zubkov AY, Mandrekar JN, Claassen DO, Manno EM, Wijdicks
EF, Rabinstein AA (2008) Predictors of outcome in warfarinrelated intracerebral hemorrhage. Arch Neurol 65(10):13201325
Sjoblom L, Hardemark HG, Lindgren A, Norrving B, Fahlen M,
Samuelsson M, Stigendal L, Stockelberg D, Taghavi A, Wallrup
L, Wallvik J (2001) Management and prognostic features of
intracerebral hemorrhage during anticoagulant therapy: a
Swedish multicenter study. Stroke 32(11):25672574
Huttner HB, Schellinger PD, Hartmann M, Kohrmann M, Juettler E, Wikner J, Mueller S, Meyding-Lamade U, Strobl R,
Mansmann U, Schwab S, Steiner T (2006) Hematoma growth
and outcome in treated neurocritical care patients with intracerebral hemorrhage related to oral anticoagulant therapy: comparison of acute treatment strategies using vitamin K, fresh
frozen plasma, and prothrombin complex concentrates. Stroke
37(6):14651470
Oden A, Fahlen M (2002) Oral anticoagulation and risk of
death: a medical record linkage study. BMJ (Clinical research
ed) 325(7372):10731075
Koo S, Kucher N, Nguyen PL, Fanikos J, Marks PW, Goldhaber
SZ (2004) The effect of excessive anticoagulation on mortality
and morbidity in hospitalized patients with anticoagulant-related
major hemorrhage. Arch Intern Med 164(14):15571560
Aguilar MI, Hart RG, Kase CS, Freeman WD, Hoeben BJ,
Garcia RC, Ansell JE, Mayer SA, Norrving B, Rosand J, Steiner
T, Wijdicks EF, Yamaguchi T, Yasaka M (2007) Treatment of
warfarin-associated intracerebral hemorrhage: literature review
and expert opinion. Mayo Clin Proc 82(1):8292
Fredriksson K, Norrving B, Stromblad LG (1992) Emergency
reversal of anticoagulation after intracerebral hemorrhage.
Stroke 23(7):972977
Phan TG, Koh M, Wijdicks EF (2000) Safety of discontinuation
of anticoagulation in patients with intracranial hemorrhage at
high thromboembolic risk. Arch Neurol 57(12):17101713

J Neurol (2012) 259:212224


82. Wijdicks EF, Schievink WI, Brown RD, Mullany CJ (1998) The
dilemma of discontinuation of anticoagulation therapy for
patients with intracranial hemorrhage and mechanical heart
valves. Neurosurgery 42(4):769773
83. Dentali F, Ageno W, Crowther M (2006) Treatment of coumarin-associated coagulopathy: a systematic review and proposed
treatment algorithms. J Thromb Haemost 4(9):18531863
84. Goldstein JN, Thomas SH, Frontiero V, Joseph A, Engel C,
Snider R, Smith EE, Greenberg SM, Rosand J (2006) Timing of
fresh frozen plasma administration and rapid correction of
coagulopathy in warfarin-related intracerebral hemorrhage.
Stroke 37(1):151155
85. Warkentin TE, Crowther MA (2002) Reversing anticoagulants
both old and new. Can J Anaesth 49(6):S11S25
86. Leissinger CA, Blatt PM, Hoots WK, Ewenstein B (2008) Role
of prothrombin complex concentrates in reversing warfarin
anticoagulation: a review of the literature. Am J Hematol
83(2):137143
87. Preston FE, Laidlaw ST, Sampson B, Kitchen S (2002) Rapid
reversal of oral anticoagulation with warfarin by a prothrombin
complex concentrate (Beriplex): efficacy and safety in 42
patients. Br J Haematol 116(3):619624
88. van Aart L, Eijkhout HW, Kamphuis JS, Dam M, Schattenkerk
ME, Schouten TJ, Ploeger B, Strengers PF (2006) Individualized
dosing regimen for prothrombin complex concentrate more
effective than standard treatment in the reversal of oral anticoagulant therapy: an open, prospective randomized controlled
trial. Thromb Res 118(3):313320
89. Kuwashiro T, Yasaka M, Itabashi R, Nakagaki H, Miyashita F,
Naritomi H, Minematsu K (2011) Effect of prothrombin complex concentrate on hematoma enlargement and clinical outcome in patients with anticoagulant-associated intracerebral
hemorrhage. Cerebrovasc Dis 31(2):170176
90. Freeman WD, Brott TG, Barrett KM, Castillo PR, Deen HG Jr,
Czervionke LF, Meschia JF (2004) Recombinant factor VIIa for
rapid reversal of warfarin anticoagulation in acute intracranial
hemorrhage. Mayo Clin Proc 79(12):14951500
91. Lin J, Hanigan WC, Tarantino M, Wang J (2003) The use of
recombinant activated factor VII to reverse warfarin-induced
anticoagulation in patients with hemorrhages in the central
nervous system: preliminary findings. J Neurosurg 98(4):
737740
92. Brody DL, Aiyagari V, Shackleford AM, Diringer MN (2005)
Use of recombinant factor VIIa in patients with warfarin-associated intracranial hemorrhage. Neurocrit Care 2(3):263267
93. Lee SB, Manno EM, Layton KF, Wijdicks EF (2006) Progression of warfarin-associated intracerebral hemorrhage after INR
normalization with FFP. Neurology 67(7):12721274
94. Yasaka M, Sakata T, Minematsu K, Naritomi H (2002) Correction of INR by prothrombin complex concentrate and vitamin
K in patients with warfarin related hemorrhagic complication.
Thromb Res 108(1):2530
95. Illanes S, Zhou W, Schwarting S, Heiland S, Veltkamp R (2011)
Comparative effectiveness of hemostatic therapy in experimental warfarin-associated intracerebral hemorrhage. Stroke
42(1):191195
96. Hanley JP (2004) Warfarin reversal. J Clin Pathol 57(11):
11321139
97. Bertram M, Bonsanto M, Hacke W, Schwab S (2000) Managing
the therapeutic dilemma: patients with spontaneous intracerebral
hemorrhage and urgent need for anticoagulation. J Neurol
247(3):209214
98. Lieberman A, Hass WK, Pinto R, Isom WO, Kupersmith M,
Bear G, Chase R (1978) Intracranial hemorrhage and infarction
in anticoagulated patients with prosthetic heart valves. Stroke
9(1):1824

223
99. Butler AC, Tait RC (1998) Management of oral anticoagulantinduced intracranial haemorrhage. Blood Rev 12(1):3544
100. Leker RR, Abramsky O (1998) Early anticoagulation in patients
with prosthetic heart valves and intracerebral hematoma. Neurology 50(5):14891491
101. Nakagawa T, Kubota T, Handa Y, Kawano H, Sato K (1995)
Intracranial hemorrhage due to long-term anticoagulant therapy
in patients with prosthetic heart valvesfour case reports. Neurol
Med Chir 35(3):156159
102. Tinker JH, Tarhan S (1978) Discontinuing anticoagulant therapy
in surgical patients with cardiac valve prostheses. Observations
in 180 operations. JAMA 239(8):738739
103. Chamorro A, Busse O, Obach V, Toni D, Sandercock P,
Reverter JC, Cervera A, Torres F, Davalos A (2005) The rapid
anticoagulation prevents ischemic damage study in acute
strokefinal results from the writing committee. Cerebrovasc
Dis 19(6):402404. doi:10.1159/000086100
104. Pessin MS, Estol CJ, Lafranchise F, Caplan LR (1993) Safety of
anticoagulation after hemorrhagic infarction. Neurology
43(7):12981303
105. Chamorro A, Vila N, Saiz A, Alday M, Tolosa E (1995) Early
anticoagulation after large cerebral embolic infarction: a safety
study. Neurology 45(5):861865
106. Einhaupl KM, Villringer A, Meister W, Mehraein S, Garner C,
Pellkofer M, Haberl RL, Pfister HW, Schmiedek P (1991)
Heparin treatment in sinus venous thrombosis. Lancet
338(8767):597600
107. Stam J, De Bruijn SF, DeVeber G (2002) Anticoagulation for
cerebral sinus thrombosis. Cochrane database of systematic
reviews (Online) 4:CD002005
108. Chamorro A (2001) Immediate anticoagulation in acute focal
brain ischemia revisited: gathering the evidence. Stroke
32(2):577578
109. Knudsen KA, Rosand J, Karluk D, Greenberg SM (2001)
Clinical diagnosis of cerebral amyloid angiopathy: validation of
the Boston criteria. Neurology 56(4):537539
110. PROGRESS Collaborative Group (2001) Randomised trial of a
perindopril-based blood-pressure-lowering regimen among
6,105 individuals with previous stroke or transient ischaemic
attack. Lancet 358(9287):10331041
111. ODonnell HC, Rosand J, Knudsen KA, Furie KL, Segal AZ,
Chiu RI, Ikeda D, Greenberg SM (2000) Apolipoprotein E
genotype and the risk of recurrent lobar intracerebral hemorrhage. N Engl J Med 342(4):240245
112. Eckman MH, Rosand J, Knudsen KA, Singer DE, Greenberg
SM (2003) Can patients be anticoagulated after intracerebral
hemorrhage? A decision analysis. Stroke 34(7):17101716
113. Stangier J (2008) Clinical pharmacokinetics and pharmacodynamics of the oral direct thrombin inhibitor dabigatran etexilate.
Clin Pharmacokinet 47(5):285295
114. Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J,
Parekh A, Pogue J, Reilly PA, Themeles E, Varrone J, Wang S,
Alings M, Xavier D, Zhu J, Diaz R, Lewis BS, Darius H, Diener
HC, Joyner CD, Wallentin L (2009) Dabigatran versus warfarin in
patients with atrial fibrillation. N Engl J Med 361(12):11391151
115. Gage BF (2009) Can we rely on RE-LY? N Engl J Med
361(12):12001202
116. van Ryn J, Stangier J, Haertter S, Liesenfeld KH, Wienen W,
Feuring M, Clemens A (2010) Dabigatran etexilatea novel,
reversible, oral direct thrombin inhibitor: interpretation of
coagulation assays and reversal of anticoagulant activity.
Thromb Haemost 103(6):11161127
117. Hylek EM (2010) Therapeutic potential of oral factor Xa
inhibitors. N Engl J Med 363(26):25592561
118. Connolly SJ, Eikelboom J, Joyner C, Diener HC, Hart R, Golitsyn S, Flaker G, Avezum A, Hohnloser SH, Diaz R, Talajic M,

123

224

119.

120.

121.

122.

J Neurol (2012) 259:212224


Zhu J, Pais P, Budaj A, Parkhomenko A, Jansky P, Commerford
P, Tan RS, Sim KH, Lewis BS, Van Mieghem W, Lip GY, Kim
JH, Lanas-Zanetti F, Gonzalez-Hermosillo A, Dans AL,
Munawar M, ODonnell M, Lawrence J, Lewis G, Afzal R,
Yusuf S (2011) Apixaban in patients with atrial fibrillation.
N Engl J Med 364(9):806817
Berwaerts J, Dijkhuizen RS, Robb OJ, Webster J (2000) Prediction of functional outcome and in-hospital mortality after
admission with oral anticoagulantrelated intracerebral hemorrhage. Stroke 31:25582562
Baker RI, Coughlin PB, Gallus AS, Harper PL, Salem HH,
Wood EM; Warfarin Reversal Consensus Group (2004) Warfarin reversal: consensus guidelines, on behalf of the Australasian Society of Thrombosis and Haemostasis. Med J Aust
181(9):492497
Baglin TP, Keeling DM, Watson HG; British Committee for
Standards in Haematology (2006) Guidelines on oral anticoagulation (warfarin): third edition2005 update. Br J Haematol
132(3):277285
Steiner T, Kaste M, Forsting M, Mendelow D, Kwiecinski H,
Szikora I, Juvela S, Marchel A, Chapot R, Cognard C, Unterberg
A, Hacke W (2006) Recommendations for the management of

123

intracranial haemorrhagepart I: spontaneous intracerebral


haemorrhage. The European Stroke Initiative Writing Committee and the Writing Committee for the EUSI Executive Committee. Cerebrovasc Dis 22(4):294316
123. Broderick J, Connolly S, Feldmann E, Hanley D, Kase C,
Krieger D, Mayberg M, Morgenstern L, Ogilvy CS, Vespa P,
Zuccarello M; American Heart Association; American Stroke
Association Stroke Council; High Blood Pressure Research
Council; Quality of Care and Outcomes in Research Interdisciplinary Working Group (2007) Guidelines for the management
of spontaneous intracerebral hemorrhage in adults: 2007 update:
a guideline from the American Heart Association/American
Stroke Association Stroke Council, High Blood Pressure
Research Council, and the Quality of Care and Outcomes in
Research Interdisciplinary Working Group. Stroke 38(6):2001
2023
124. Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G;
American College of Chest Physicians (2008) Pharmacology
and management of the vitamin K antagonists: American College of Chest Physicians Evidence-Based Clinical Practice
Guidelines (8th Edition). Chest 133(6 Suppl):160S198S

Das könnte Ihnen auch gefallen