Sie sind auf Seite 1von 12

Journal of Clinical Anesthesia (2016) 32, 224235

Review

Use of direct oral anticoagulants with regional


anesthesia in orthopedic patients
Gianluca Cappelleri MD a,, Andrea Fanelli MD b
a

Anaesthesia and Intensive Care Unit, Azienda Ospedaliera Istituto Ortopedico Gaetano Pini, 20122, Milan, Italy
Anaesthesia and Intensive Care Unit, Policlinico S. Orsola-Malpighi, 40138, Bologna, Italy

Received 28 May 2015; revised 5 January 2016; accepted 22 February 2016

Keywords:
Direct oral anticoagulant;
Major orthopedic surgery;
Neuraxial anesthesia;
Perioperative management;
Postoperative management

Abstract The use of direct oral anticoagulants including apixaban, rivaroxaban, and dabigatran, which are
approved for several therapeutic indications, can simplify perioperative and postoperative management
of anticoagulation. Utilization of regional neuraxial anesthesia in patients receiving anticoagulants carries
a relatively small risk of hematoma, the serious complications of which must be acknowledged. Given
the extensive use of regional anesthesia in surgery and the increasing number of patients receiving direct oral
anticoagulants, it is crucial to understand the current clinical data on the risk of hemorrhagic complications in
this setting, particularly for anesthesiologists. We discuss current data, guideline recommendations, and best
practice advice on effective management of the direct oral anticoagulants and regional anesthesia, including
in specic clinical situations, such as patients undergoing major orthopedic surgery at high risk of a thromboembolic
event, or patients with renal impairment at an increased risk of bleeding.
2016 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

1. Introduction
Venous thromboembolism (VTE), comprising deep vein
thrombosis (DVT) and pulmonary embolism (PE), and its
long-term complications cause signicant morbidity, mortality
and healthcare costs [13]. The risk of VTE is particularly
high in patients who undergo major orthopedic surgery, such

Conicts of interest: Editorial support was funded by Bayer HealthCare


Pharmaceuticals and Janssen Scientic Affairs, LLC. GC reports no conicts
of interest. AF has received speaker's honoraria from Bayer HealthCare
Pharmaceuticals.
Corresponding author at: Azienda Ospedaliera Istituto Ortopedico
Gaetano Pini, Piazza Cardinal Ferrari, 20122, Milan, Italy.
Tel.: +39 0258296297; fax: +39 0258296283.
E-mail addresses: gianluca.cappelleri@gpini.it (G. Cappelleri),
andre.fanelli@gmail.com (A. Fanelli).

as total knee or hip replacement surgery [4]; therefore, these


patients routinely receive anticoagulants for short-term perioperative and postoperative thromboprophylaxis [4,5].
Whether a patient is receiving long-term anticoagulation (for
the treatment or prevention of VTE or for stroke prevention
in patients with non-valvular atrial brillation [AF]) prior to
orthopedic surgery should also be considered because appropriate anticoagulant management based on the risk of thromboembolism and bleeding is crucial in these patients [5].
Additionally, the use of regional neuraxial anesthesia poses a
risk of hematoma in patients receiving anticoagulants, because
of the insertion and removal of the needle and catheter.
The direct oral anticoagulants (OACs) apixaban, rivaroxaban, and dabigatran are approved for the prevention of VTE
after elective knee or hip replacement surgery in the European
Union and/or United States, as well as for the prevention of

http://dx.doi.org/10.1016/j.jclinane.2016.02.028
0952-8180/ 2016 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/
licenses/by-nc-nd/4.0/).

Anticoagulation and anesthesia


stroke in patients with non-valvular AF, and the treatment of
VTE and prevention of recurrent VTE [611]. In Europe,
rivaroxaban is also indicated for co-administration with acetylsalicylic acid (ASA) alone or with ASA plus clopidogrel or
ticlopidine for the prevention of atherothrombotic events after
acute coronary syndrome with elevated cardiac biomarkers
[10]. Apixaban and rivaroxaban are direct inhibitors of Factor
Xa, whereas dabigatran is a direct inhibitor of thrombin.
All three agents have been found to exhibit predictable
pharmacokinetics and pharmacodynamics, and have a fast
onset of actionreaching maximum plasma concentration
rapidly after oral administration (apixaban, 3-4 hours; rivaroxaban, 2-4 hours; dabigatran, 0.5-2.0 hours) [6,8,10,1215].
The half-lives of apixaban, rivaroxaban, and dabigatran are
~ 12 hours, 5-13 hours, and 11-14 hours, respectively
[6,8,10]. Furthermore, the pharmacokinetic and pharmacodynamic characteristics are not inuenced to a clinically meaningful extent by body weight, age, or gender, which
collectively indicate that these direct OACs can be given as
xed dosing regimens without the need for routine coagulation
monitoring [1620]. The routes of drug elimination differ between the 2 drug classes: apixaban and rivaroxaban have multiple pathways including renal excretion (~ 27% and 66%
[33% as active drug and 33% after metabolic degradation], respectively), whereas elimination of dabigatran is predominantly via the renal system (85%) [6,8,10,21]. In patients receiving
dabigatran for the prevention of VTE after knee or hip replacement surgery, a dose reduction is required in those with moderate renal impairment (creatinine clearance 30-50 mL min1),
those aged 75 years or over, and those taking concomitant
mild-to-moderate P-glycoprotein inhibitors [8]. Owing to their
mode of action, all anticoagulants can increase the risk of
bleeding events. However, apixaban, rivaroxaban, and dabigatran did not pose any signicantly increased risk of major
bleeding events compared with enoxaparin in phase III clinical
trials in the prevention of VTE after knee or hip replacement
surgery [2230]. Despite the clinical evidence on the safety
and efcacy prole of direct OACs demonstrated in clinical trials,
physiciansparticularly anesthesiologistsneed to be aware of
how to minimize the risks of bleeding complications when treating patients who are taking an anticoagulant and are due to undergo surgery with regional anesthesia.
Regional anesthesia techniques have become increasingly popular because they offer several benets over general anesthesia and
traditional systemic analgesia, including reduced risk of mortality
and less postoperative pain, thus facilitating early postoperative
mobilization of the limbs and consequently reducing the chances
of postoperative VTE [31,32]. Regional anesthesia is used in
millions of operations each year and, at a time when the use
of the direct OACs is growing, bleeding-related complications
could arise if appropriate guidance is not followed [33]. This
review aims to provide anesthesiologists with an overview of
current data on the risk of hemorrhagic complications with
direct OACs when using regional anesthesia, particularly
in patients at a high risk of VTE, such as those undergoing
major orthopedic surgery. Moreover, current guideline

225
recommendations will be discussed and best practice in specic
clinical situations, such as in patients with renal impairment who
are at an increased risk of bleeding, will be highlighted.

2. Regional anesthesia and complications


The most common complications of regional anesthesia
include postdural puncture headache (occurs in 0.1%-36% of
patients but rarely lasts longer than 7 days) [34], backache (affects up to 11% of patients in the 5 days after the procedure)
[35], and peripheral nerve damage (eg, transient paresthesia
is estimated in up to 8%-10% of patients in the immediate days
after regional anesthesia) [36]. Serious complications, such as
spinal or epidural hematoma and epidural abscess, are rare and
therefore difcult to study in detail [37]. For peripheral nerve
block, neurological complications are extremely rare, ranging
between 0.02% and 0.04% [38,39].

2.1. Risk factors associated with regional anesthesia


In a systematic review of 141 studies (n = 9559),
neuraxial anesthesia was shown to be associated with lower
rates of mortality (2.1% vs 3.1%; odds ratio [OR] 0.7; 95%
condence interval [CI] 0.54-0.90; P = .006), occurrence of
DVT (odds reduction of 44%; OR 0.56; 95% CI 0.43-0.72;
P b .001) and PE (odds reduction of 55%; OR 0.45; 95% CI
0.29-0.69; P b .001), among other outcomes [31]. A more recent observational study showed that neuraxial anesthesia was
associated with a signicantly reduced need for blood transfusions compared with patients receiving general anesthesia
(28.5% vs 44.7%; OR 0.52; 95% CI 0.45-0.61; P b .0001) after simultaneous bilateral total knee replacement surgery [40].

2.2. Risk factors for the development of


spinal hematoma
The overall incidences of hematoma in patients receiving
neuraxial anesthesia (epidural and spinal) have been estimated
to be as low as 1 in 220 000 and 1 in 320 000, respectively, in
the absence of traumatic puncture and without the use of heparin or ASA [41]. However, the incidence of spinal hematoma
increases with the presence of any risk factors, listed in Table 1,
and more so if heparin anticoagulation is accompanied by traumatic puncture (increasing incidence rates to 1 in 2000 and 1
in 2900 for epidural and spinal anesthesia, respectively). For
ASA therapy the risk is 1 in 8500 and 1 in 12 000 for epidural
and spinal anesthesia, respectively [41,42]. An incidence of
between 1 in 1000 and 1 in 10 000 has been estimated for neuraxial hematoma after central neural blockade in patients undergoing orthopedic procedures and receiving preoperative
thromboprophylaxis with enoxaparin [43]. A recent review
assessing spinal/epidural anesthesia complications in Finland
from 2000 to 2009 showed the risk of developing a serious
complication (dened as a potentially fatal event or an event
of over 1 year in duration, including epidural hematoma) after

226

G. Cappelleri, A. Fanelli

Table 1 Risk factors associated with spinal epidural hematoma


during central neuraxial block [33]
Patient related
Elderly
Female sex
Inherited coagulopathy
Acquired coagulopathies (liver/renal failure, malignancy,
HELLP syndrome, DIC etc)
Thrombocytopenia
Spinal abnormalities (spinal bida/stenosis, spinal tumors,
ankylosing spondylitis, and osteoporosis)
Drug related
Anticoagulation/antiplatelet/brinolytic
Immediate (pre- and post-CNB) anticoagulant administration
Dual anticoagulant/antiplatelet therapies
Procedure related
Catheter insertion/removal
Traumatic procedure (multiple attempts)
Presence of blood in the catheter during insertion/removal
Indwelling epidural catheter N single-shot epidural block N
single-shot spinal block
CNB = central neuraxial block; DIC = disseminated intravascular coagulation;
HELLP = hemolysis, elevated liver enzymes, low platelet count.

central neuraxial block to be approximately 1 in 35 000


patients, with 1 in 53 000 patients presenting with irreversible
damage [44]. Of the 13 cases of neuraxial hematoma reported
from a total of 1.4 million procedures involving neuraxial
central blocks, 10 were associated with thromboprophylaxis
(enoxaparin, dalteparin, or fondaparinux); however, for
6 of 10 of these occurrences, the time elapsed between
cessation/initiation of thromboprophylaxis and puncture may
not have been sufcient to prevent a bleeding event [44]. A
recent retrospective study on the safety of epidural and spinal
neuraxial block showed that the incidence of perioperative
neurological complications in orthopedic patients was relatively low (0.96/1000; 95% CI 0.77-1.16/1000), but was
higher than previously reported in other non-obstetric populations [45]. Risk factors associated with an increased risk of
Table 2

clinically signicant bleeding causing spinal hematoma can


be divided into patient-, drug-, and procedure-related categories (Table 1).

2.3. Patient-related factors increasing the risk of


spinal hematoma
Regarding patient-related factors, clinicians must exercise
particular care with elderly patients requiring surgical interventionthese patients are at increased risk of thromboembolic disorders [46,47] and are, therefore, more likely to
be receiving anticoagulants. Furthermore, the bleeding risk in
elderly patients receiving anticoagulants is augmented
[48,49]. Elderly patients are also more likely to have renal or hepatic impairment, causing increased drug plasma concentrations,
and consequently drug elimination may be slower in these
patients [50]. Real-world data from the RIETE (Registro Informatizado de Pacientes con Enfermedad TromboEmblica)
registry have shown major and fatal bleeding to be more common in patients receiving anticoagulation therapy who were
aged 80 years compared with those aged b 80 years (3.4%
vs 2.1% and 0.8% vs 0.4%, respectively) [51]. Therefore,
knowledge about the metabolism of the direct OACs approved
for VTE prevention after major orthopedic surgery is essential
when deciding which anticoagulant to prescribe to a particular
patient. Whereas the metabolism of dabigatran is mostly
dependent on renal clearance, the metabolism of apixaban
and rivaroxaban involves multiple pathways and is less
dependent on renal function (Table 2). The high frequency of
co-medication use in elderly patients also increases both the
potential for drug-drug interactions and the risk of bleeding [52].

2.4. Drug-related factors increasing the risk of


spinal hematoma
Some drug interactions of anticoagulants might increase the
risk of bleeding complications, including the risk of spinal hematoma. The direct OACs have fewer drug-drug and drug-food

Pharmacokinetic characteristics of direct oral anticoagulants as per their Summary of Product Characteristics [6,8,10]
Apixaban

Rivaroxaban

Dabigatran

Bioavailability

~ 50% for doses up to 10 mg

~ 7%

Time to peak activity


Half-life

3-4 h
~ 12 h

Renal elimination

~ 27%

Pharmacokinetic interactions

Strong inhibitors/inducers
of both CYP3A4 and P-gp

80%-100%
(for 10 mg dose and for 15 mg
and 20 mg doses taken with food)
2-4 h
5-9 h (young individuals)
to 11-13 h (elderly individuals)
33% as active drug
(direct renal excretion) and 33%
after metabolic degradation
Strong inhibitors of both CYP3A4 and P-gp;
strong CYP3A4 inducers

CYP3A4 = cytochrome P450 3A4; P-gp = P-glycoprotein.

0.5-2.0 h
11-14 h
85%

Strong P-gp
inhibitors and
inducers

Anticoagulation and anesthesia


interactions compared with vitamin K antagonists. The most
signicant interactions are those of apixaban and rivaroxaban
with systemic co-administration of strong inhibitors of both cytochrome P450 3A4 and P-glycoprotein (such as the azoleantimycotics ketoconazole and itraconazole and the HIV protease
inhibitor ritonavir), which is not recommended [6,10], and
dabigatran with amiodarone and verapamil that requires a
reduction in the daily dose [8]. For all 3 direct OACs, care
should be taken when co-administering with non-steroidal
anti-inammatory drugs, ASA, or platelet aggregation inhibitors, which may increase bleeding risk [6,10].

2.5. Procedural-related factors increasing the risk of


spinal hematoma
The risk of spinal hematoma has also been associated with
procedural factors, including unintentional needle/catheterrelated mechanical damage of neural or vascular components
of the spine due to a difcult or traumatic spinal procedure
[5355]. The patient characteristics also have an inuence
on procedure-related risk factors; for example, aging is associated with skeletal degeneration that may make the anesthesia
procedure technically more difcult [56].

3. Inuence of anesthesia in patients receiving


direct oral anticoagulants: evidence from
phase III clinical studies with a focus on
major orthopedic surgery
Given the increasing utilization of direct OACs for the
treatment and prevention of thromboembolic disorders, it is
crucial to evaluate the risk of compressive hematoma and the
impact on clinical outcomes when regional anesthesia is used.
To discuss the effect of regional anesthesia on the risk of compressive hematoma and bleeding risk, we searched PubMed
with no date restrictions for phase III clinical trials comparing
apixaban, rivaroxaban, or dabigatran with standard of
care/low molecular weight heparin for VTE prevention/
thromboprophylaxis after total knee or hip replacement
surgery/knee or hip arthroplasty and for studies investigating
spinal hematoma after regional anesthesia.
It is worth noting the risk of spinal hematoma with regional
anesthesia when receiving traditional anticoagulants. Vitamin
K antagonist therapy is a contraindication for neuraxial
anesthesia, owing to the high risk of bleeding associated with
therapeutic doses [57]. The risk of spinal hematoma in patients
receiving low molecular weight heparin (LMWH) concomitantly with epidural anesthesia is reported to be 1:6600 for
single-shot epidural and 1:3100 with an indwelling catheter
for the LMWH dosage of 30 mg twice daily and 1:18 000
for a 40 mg once-daily dosage [57,58]. There are fewer data
for ASA, but it has been demonstrated that medical patients
receiving daily doses above 100 mg are at increased risk of
hemorrhagic complications [59].

227
Central nerve blocks have been classied as a high bleeding risk procedure owing to their rare but severe complication,
eg, spinal-epidural hematoma [60,61]. Because of the absence
of evidence-based data, when neuraxial procedures are
performed in these conditions, a systematic follow-up of these
patients and a heightened awareness of bleeding complications
should be observed.

3.1. Apixaban
Studies specically designed to investigate the relationship
between anesthesia and spinal hematoma risk in patients
receiving apixaban are lacking. However, phase III clinical
trials comparing apixaban (2.5 mg twice daily) with subcutaneous enoxaparin (30 mg twice daily or 40 mg once daily)
for VTE prevention after total knee or hip replacement surgery
found that apixaban was as effective as or better than enoxaparin at reducing the risk of the composite of VTE or death
with similar or lower rates of bleeding [2224]. In these 3
trials, 15.8% of patients randomized to apixaban had regional
anesthesia. Experience of apixaban use in the presence of
neuraxial blockade is limited and, according to its product label
and guidance from the European Society of Anaesthesiology
(ESA), should only be done with extreme caution [6,57].

3.2. Rivaroxaban
The incidences of intraspinal bleeding or hemorrhagic
puncture were included under the denition for major bleeding used in the RECORD (REgulation of Coagulation in
ORthopaedic surgery to prevent Deep vein thrombosis and
pulmonary embolism) clinical trial program of rivaroxaban
[27,62]. A post hoc analysis assessed 12 729 randomized
patients in the pooled RECORD1-4 program undergoing elective total knee or hip replacement surgery for events related to
neuraxial anesthesia, such as neuraxial hematoma [62].
Thromboprophylactic therapy was provided to patients as
either oral rivaroxaban (10 mg once daily, initiated 6-8 hours
after surgery) or subcutaneous enoxaparin (40 mg once daily,
started 12 hours prior to surgery, or 30 mg twice daily, started
12-24 hours after surgery) [62]. Although 66% of patients
(safety population, n = 12 383) received neuraxial anesthesia
(9% of patients also received general anesthesia), only 1
compressive hematoma after epidural catheter removal was
recorded. This event occurred after elective total knee replacement surgery in a patient treated with enoxaparin, who was at
increased risk of bleeding as a result of advanced age and renal
insufciency. The patient was adequately managed with a
laminectomy without further bleeding or neurological
complications [62]. The type of anesthesia (spinal with or
without general anesthesia, epidural, epidural with indwelling
catheter, general or spinal with femoral block, or general
alone) had no inuence on the rates of total venous thromboembolic events, which were lower with rivaroxaban than with
standard of care [62].

228
In the RECORD post hoc subanalysis, the incidence of total
venous thromboembolic events (the composite of any DVT,
non-fatal PE, and all-cause mortality) and safety proles were
similar irrespective of the type of anesthesia used. Neuraxial
anesthesia was associated with a slightly lower rate of
symptomatic thromboembolic events compared with general
anesthesia in patients undergoing elective total knee or hip
replacement surgery [62]. However, available experience
using rivaroxaban with neuraxial blockade remains limited,
and real risk is difcult to evaluate owing to the low incidences
of spinal and epidural hematoma in this setting. The ESA
guidelines, published before the completion of the relevant
RECORD subanalysis, recommend extreme caution when
using rivaroxaban with neuraxial blockade [57]. The
American Society of Regional Anesthesia and Pain Medicine
(ASRA) guidelines for dabigatran, as well as rivaroxaban,
are still being evaluated. These guidelines do not provide
detailed information for the management of anticoagulation
with direct OACs owing to a lack of available information
on block performance [41].

3.3. Dabigatran
An analysis of the pooled population of patients from the
phase III RE-MODEL, RE-NOVATE, and RE-MOBILIZE
studies who underwent major hip or knee replacement surgery
and received thromboprophylaxis with dabigatran (220 mg or
150 mg once daily) or enoxaparin showed a higher use of neuraxial anesthesia (n = 4212; 52%) compared with general anesthesia (n = 2311; 29%) or combination anesthesia, namely,
neuraxial or general anesthesia combined with a peripheral
nerve block (n = 1539; 19%) [63]. The use of neuraxial anesthesia was not associated with neuraxial hematomano cases
were reported [63]. The efcacy and safety outcomes of the
dabigatran groups did not differ from those of the standard
therapy groups, and were not signicantly inuenced by the
type of anesthesia used [63]. A reduced incidence of major
VTE and VTE-related mortality was observed with neuraxial
anesthesia compared with general anesthesia (4.2% vs 3.1%;
OR 1.40; 95% CI 1.03-1.90; P = .035) in the overall population,
but no signicant differences or clear trends were recorded
regarding the safety outcomes of major bleeding or major
and non-major clinically relevant bleeding between the types
of anesthesia [63]. Experience with combined use of neuraxial
blockade and dabigatran is restricted by its manufacturer's
advice against this, as stated by the ESA guidelines [8,57].

4. Management of direct oral anticoagulants and


neuraxial anesthesia in major orthopedic surgery
Management strategies must balance the surgical risk of
hematoma in the anesthetized patient with effective thromboprophylaxis. Understanding the pharmacodynamic and
pharmacokinetic properties of an anticoagulant and the inuence of patient age and hepatic and renal function is essential.

G. Cappelleri, A. Fanelli
Providing neuraxial anesthesia and the timing of needle/catheter
insertion/removal in patients who are receiving or are to receive
anticoagulant therapy involves individual assessment of the
risks and benets of such a procedure, together with rigorous
management of anticoagulation activity and monitoring for
signs of neurological dysfunction. It is of the utmost importance
to ensure that the anticoagulation effect is adequately reduced
before proceeding with a neuraxial block. It is possible to
measure the effect of the direct OACs either qualitatively or
quantitatively, if required.

4.1. Perioperative management of direct


oral anticoagulants
Adequate protocols for interrupting and resuming thromboprophylaxis based on the pharmacokinetic properties of the direct
OACs are essential. Clinical practice guidelines, such as those of
the ESA [57]), ASRA [41], and the Scandinavian Society of
Anaesthesiology and Intensive Care Medicine (SSAI) [64], provide recommendations for the regional anesthetic management
of patients undergoing surgery and requiring anticoagulation,
including with direct OACs (Table 3). These recommendations, based on data from clinical trials, the pharmacokinetic
prole of each anticoagulant, and from clinical experience, advise a waiting time between interrupting anticoagulation and
performing neuraxial blockade that depends on the elimination
half-life of the anticoagulant agent [57]. The percentages of
drug remaining in the circulation after 1-6 half-lives are
50%, 25%, 12.5%, 6.3%, 3.1%, and 1.6%, respectively [65].
A recommendation for a waiting period of 2 half-lives, to reduce the bioavailability of an anticoagulant to less than 25%,
does not take into consideration how the half-life elimination
of a drug is inuenced by patient characteristics. Patients
who undergo joint replacement surgery are typically elderly,
often with renal impairment and receiving antiplatelet therapy;
for these patients, a waiting time of 3-5 half-lives could be
more appropriate if hemostatic safety is not assured (Table 4).
To prevent VTE after major orthopedic surgery, the direct
OACs have to be administered in the postoperative period
when adequate hemostasis has been established, as determined
by the treating physician.
Recently, the direct OACs have been approved for long-term
prevention of thromboembolic events in non-valvular AF, and
for the treatment and secondary prophylaxis of VTE. Every
year, 15%-20% of patients receiving therapeutic doses of anticoagulants require perioperative temporary interruption, and
the characteristics of both the patients and the procedures should
be considered when scheduling interruption and resumption.
4.1.1. Perioperative management of apixaban
4.1.1.1. Interruption of apixaban before surgery. The
apixaban Summary of Product Characteristics recommends
that discontinuation should occur 48 hours prior to elective
surgery associated with a moderate or high risk of bleeding,
although a longer waiting period may be advisable in elderly
patients (who show a slower elimination rate and are at

Anticoagulation and anesthesia

229

Table 3 Recommendations provided by each direct oral anticoagulants Summary of Product Characteristics (SPC), the European Society
of Anesthesiology (ESA), and the Scandinavian Society of Anaesthesiology and Intensive Care Medicine (SSAI) for managing anticoagulation
interruption/initiation and puncture or catheter insertion/removal [6,8,10,57,64].
Time between last dose of anticoagulant and
puncture or catheter insertion or removal

Time between puncture or catheter insertion


or removal and initiation of anticoagulation

SPC

ESA

SSAI a

SPC

ESA

SSAI

Apixaban
Rivaroxaban c

20-30 h
18 h

26-30 h
22-26 h

Data not available


18 h

4-6 h
4-6 h

6h
6h

Dabigatran d

Contraindicated
by the manufacturer

Contraindicated
by the manufacturer

Not included

5 h
6 h
(24 h in case
of traumatic puncture)
2 h

6h

Not included

a
Recommended minimum time intervals between the last dose of anticoagulant and central neuraxial block, and between central neuraxial block and rst
dose or re-initiation of anticoagulant; the same recommendations apply for manipulation or removal of a catheter.
b
For prophylaxis, 2.5 mg twice daily.
c
For prophylaxis, 10 mg once daily.
d
For prophylaxis, 150-220 mg once daily.

increased risk of bleeding) [6]. There is no clinical experience


with the use of apixaban with indwelling intrathecal or epidural catheters [6]. However, if required, the recommendation
provided by the Summary of Product Characteristics is to wait
20-30 hours (considering 2 times its elimination half-life) between interruption of anticoagulation therapy and neuraxial
puncture/catheter removal, with at least 1 dose being omitted
[6,66]. The ESA recommends 26-30 hours between stopping
anticoagulation and conducting a neuraxial anesthesia procedure for management of anticoagulation with apixaban in the
context of elective orthopedic surgery [57].
4.1.1.2. Resumption of apixaban after surgery. The Summary of Product Characteristics of apixaban recommends starting thromboprophylaxis 12-24 hours after the end of surgery,
provided hemostasis is achieved and allowing for a waiting
period of at least 5 hours to re-initiate treatment after catheter
removal [6]. The ESA recommends 4-6 hours between catheter removal and initiation of anticoagulation, for management
of anticoagulation with apixaban in the context of elective
orthopedic surgery [57]. No further recommendations were
provided by the ASRA for management of anticoagulation
with apixaban in the context of elective orthopedic surgery
[41]. The SSAI guidelines state that the lack of data prevents
any recommendation being made regarding the use of apixaban and central neuraxial block, although a period of 6 hours
should be observed between central neuraxial block or catheter
manipulation and rst dose of this anticoagulant [64].
4.1.2. Perioperative management of rivaroxaban
4.1.2.1. Interruption of rivaroxaban before surgery. The
Summary of Product Characteristics of rivaroxaban suggest
in patients with normal renal function a minimum of 24 hours
between the last therapeutic dose of the anticoagulant and the
start of the surgery [10]. For patients with severe renal impairment, many authors recommend an interval of up to 4-5 days
[60,67,68]. Imberti et al suggested a discontinuation of rivaroxaban at least 48 hours prior to procedures involving a

standard risk of bleeding, such as orthopedic surgery; for


procedures with a high risk of bleeding (neuraxial block),
rivaroxaban should be stopped 72 hours before the intervention
[69]. In the authors' opinion, patients with AF with standard
bleeding risk (HAS-BLED score b 3) could undergo neuraxial
anesthesia 48 hours after the last rivaroxaban intake, whereas
in patients with high bleeding risk, neuraxial anesthesia should
be delayed for 4 days, and this type of anesthesia should not be
considered in cases of severe renal impairment (creatinine
clearance b 30 mL min1) [69].
In patients receiving ongoing rivaroxaban, if a central block
with an indwelling catheter is used, the SSAI guidelines
recommend a minimum of 18 hours between the last dose of
rivaroxaban and removal of an indwelling catheter [64].
Because of the prolongation of the half-life in elderly patients,
the ESA guidelines recommend a longer interval (22-26 hours)
between the last dose of rivaroxaban and removal of an
indwelling catheter [57]. These 2 recommendations are in
agreement with those in the report by Rosencher et al, which
recommends 2 half-lives between rivaroxaban discontinuation
and epidural catheter removal [66]. Furthermore, these guidelines highlight the fact that patients receiving anti-hemostatic
drugs and with impaired drug elimination, such as those with
renal impairment, may require waiting times to be prolonged
[64]. The ASRA guidelines issued for rivaroxaban recommend a cautious approach owing to the lack of information
on the specics (needle placement or catheter management)
of block performance and the prolonged half-life in elderly patients resulting from deterioration of renal function, even in the
absence of any reported spinal hematoma [41].
4.1.2.2. Resumption of rivaroxaban after surgery. Rivaroxaban should be started 6-10 hours after the end of surgery,
provided hemostasis is achieved [10]. The SSAI guidelines
recommend a minimum of 6 hours between central neuraxial
block and initiation of rivaroxaban, or a delay of 24 hours if
a traumatic puncture occurred during the performance of
central nerve block [10,64]. The ESA recommends an interval

230
Table 4 Half-lives of direct oral anticoagulant drugs and recommended interval between discontinuation and surgery or neuraxial procedure, set as 2-3 half-lives or 4-5 half-lives depending
on the bleeding risk of the surgical procedure and the level of anticoagulant effect permitted at surgery [65,67,68].
Drug

Apixaban

Variation
of elimination
half-life values
(h) a [65]

Bleeding risk of surgical procedure


Low bleeding
risk surgery
(2-3 half-lives) [67]

Days
between
last dose
and surgery
[67]

High
bleeding
risk surgery
(4-5 half-lives)
[67]

Days
between
last dose
and surgery
[67]

Mild to
moderate
anticoagulant
effect (2-3 half-lives)
[68]

Days between No
last dose and or minimal
surgery [68]
anticoagulant
effect (4-5 half-lives)
[68]

15

CrCl N 50 mL min1 b
CrCl 30-50 mL min1
CrCl N 50 mL min1 c
CrCl 30-50 mL min1
CrCl 15-29.9 mL min1
CrCl N 50 mL min1 e
CrCl 30-50 mL min1

2d
3d
2d
2d
3d
2d
3d

CrCl N 50 mL min1
CrCl 30-50 mL min1
CrCl N 50 mL min1
CrCl 30-50 mL min1
CrCl 15-29.9 mL min1
CrCl N 50 mL min1
CrCl 30-50 mL min1

3d
4d
3d
3d
4d
3d
4-5 d

CrCl N 50 mL min1
CrCl 30-50 mL min1
CrCl N 50 mL min1
CrCl 30-50 mL min1

2d
3d
2d
3d

Rivaroxaban 5-13

Dabigatran

12-17;
28 d

Anticoagulant residual effect at surgery

CrCl N 50 mL min1 2 d
CrCl 30-50 mL min1 3 d

CrCl N 50 mL min1
CrCl 30-50 mL min1
CrCl N 50 mL min1
CrCl 30-50 mL min1

Days
between
last dose
and surgery
[68]
3d
4-5 d
3d
4-5 d

CrCl N 50 mL min1 3 d
CrCl 30-50 mL min1 4-5 d

CrCl = creatinine clearance.


a
Half-lives differ from those in the Summary of Product Characteristics of each agent.
b
Apixaban (5 mg twice daily) half-life for CrCl N 50 mL min1 and CrCl 30-50 mL min1 considered as 7-8 and 17-18 hours, respectively.
c
Rivaroxaban (20 mg once daily) half-life for CrCl N 50 mL min1, CrCl 30-50 mL min1 and CrCl 15-29 mL min1 considered as 8-9, 9 and 9-10.5 hours, respectively.
d
End-stage renal disease.
e
Dabigatran (150 mg twice daily) half-life for CrCl N 50 mL min1 and CrCl 30-50 mL min1 considered as 14-17 and 16-18 hours, respectively.

G. Cappelleri, A. Fanelli

Anticoagulation and anesthesia


of 4-6 hours between epidural catheter removal and the next
dose of rivaroxaban [57]. Baron et al suggested a waiting time
of 48 hours before resuming anticoagulant after procedures
with a high risk of bleeding [60].
4.1.3. Perioperative management of dabigatran
4.1.3.1. Interruption of dabigatran before surgery. The
dabigatran Summary of Product Characteristics recommends
discontinuation of the anticoagulant for 2 days before elective surgery that carries a high risk of bleeding or is considered
to be major surgery, depending on the patient's renal
function [8]. High-risk procedures, including cardiac surgery,
neurosurgery, abdominal surgery, or procedures requiring
spinal anesthesia, may require cessation of dabigatran 2-4 days
before surgery in patients with normal renal function and 4
days before surgery in patients with moderate renal impairment (creatinine clearance 30-50 mL min 1) [70]. Based on
the pharmacokinetic characteristics of dabigatran, a time interval of 36 hours (skipping 1 dose) between the last dose of dabigatran and puncture/catheter removal is suggested [66], but
this has not been studied or conrmed.
4.1.3.2. Resumption of dabigatran after surgery. Provided hemostasis is achieved, dabigatran should be started 1-4
hours after the end of surgery [8]. The ESA guidelines recommend against the use of dabigatran in the presence of neuraxial
blockade, as also advised in the dabigatran Summary of
Product Characteristics [8]. If neuraxial anesthesia is used,
the rst dose of dabigatran should not be administered within
2 hours of catheter removal, in accordance with the Summary
of Product Characteristics [8]. The ESA guidelines recommend a waiting time of 6 hours after catheter removal before
re-initiating anticoagulation with dabigatran [57]. However,
the ASRA guidelines state simply that caution must be taken
owing to the paucity of information on the performance of
neuraxial block and the prolonged half-life of this anticoagulant after multiple doses or in patients with renal impairment,
despite the absence of spinal hematomas reported up to the
publication of these guidelines [41]. The SSAI guidelines
make the sole recommendation of waiting for at least 6 hours
after a central neuraxial block or catheter removal before
the rst dose of dabigatran is taken [64]. However, based on
the pharmacokinetic characteristics of dabigatran, it may be
prudent to wait at least 12 hours after catheter removal before
administration of dabigatran [66].

4.2. Laboratory measurement of direct


oral anticoagulants
In the context of perioperative management of patients who
are receiving a direct OAC, the concentration of the direct
OAC can be determined if, for example, an emergency
surgical intervention is required by an anticoagulated patient.
However, it is important to take into account the timing of
the last dose in relation to blood sampling time when interpreting
the test results [71].

231
4.2.1. Laboratory measurement of apixaban
The quantitative measurement of plasma concentrations of
apixaban can be performed using anti-Factor Xa chromogenic
assays and adequate standard calibration curves [72].
Although apixaban prolongs prothrombin time (PT), it is not
suitable for measuring the anticoagulant effect of apixaban
and there is no known relationship with bleeding risk [71].
4.2.2. Laboratory measurement of rivaroxaban
The PT can be used to determine the relative anticoagulant
activity of rivaroxaban [71,73]. However, PT is not specic
and can be inuenced by factors such as cancer, hepatic
impairment, and vitamin K deciency [74,75]. Furthermore,
there is marked variability in the sensitivity of the reagents
available for obtaining PT results for rivaroxaban; only a
sensitive reagent, such as Neoplastin Plus (Diagnostica Stago,
Asnires-sur-Seine, France), should be used [76]. Importantly,
the international normalized ratio must not be used for rivaroxaban. The quantitative measurement of plasma concentrations
of rivaroxaban can be performed using anti-Factor Xa
chromogenic assays (eg, STA Rotachrom, Diagnostica Stago,
Asnires-sur-Seine, France) with validated calibrators [77].
When anti-Factor Xa assays are unavailable, a PT test with a
reagent sensitive to rivaroxaban may be used to conrm the
absence or presence of an anticoagulant effect (if the sampling
time after last tablet intake is known) [76,78,79].
4.2.3. Laboratory measurement of dabigatran
The activated partial thromboplastin time (aPTT) assay, for
example, provides a qualitative measure of the anticoagulant
effect of dabigatran [73]. Unfortunately, aPTT is inuenced
by the coagulometers and reagents used [75]. In the case of
aPTT results for dabigatran, there is reasonable agreement
regardless of the reagent composition, and results are within
10% of reference measurements [70]. The aPTT assay may be
useful in dabigatran-treated patients who require emergency
interventions to exclude any clinically relevant dabigatran
anticoagulant effect [71,80]. The ecarin clotting time (ECT)
assay provides a direct measurement of the activity of dabigatran, but is not readily available [71]. Calibrated ecarin chromogenic assays are also available (eg, Ecarin Chromogenic
Assay, Diagnostica Stago, Asnires-sur-Seine, France) and
may allow faster ECT measurements than the ECT assay
[71]. Direct thrombin inhibitor assays (such as the
HEMOCLOT assay, HYPHEN BioMed, Neuville-sur-Oise,
France) can be used for the quantitative measurement of
plasma concentrations of dabigatran [81]. In the absence of
this assay, an aPTT test can be used [82].

5. Neurological complications and


regional anesthesia
Development of neurological complications is a major concern in cases of regional anesthesia in patients receiving direct

232
OACs (eg, for stroke prevention). Assessment of neurological
function is the most reliable method for detecting neurological
injury, because standard laboratory tests may not provide adequate information about the coagulation status of a patient
[83,84]. Monitoring of neurological function during regional
anesthesia alongside anticoagulant therapy is essentialbut
may not be sufcientto improve neurological outcomes.
Based on the pharmacokinetic properties of the direct OACs
and their rapid onset of action (within a few hours), conning
the assessment of neurological function to the period between
catheter removal or neuraxial puncture and 4 hours after
the initiation of therapy may seem reasonable. However, a
longer period of time during which neurological function
should be assessed may be required, because the development
of neurological injury may be slow, with the onset of
neurological symptoms potentially taking as long as 3 or more
days [55].

6. Use of non-neuraxial regional techniques and


direct oral anticoagulants in clinical practice
Total knee and hip replacement surgery causes intense
postoperative pain, which may impair rehabilitation and mobilization, prolong hospitalization, and increase the risk of VTE
[85]. The use of continuous peripheral nerve blocks (femoral
and lumbar plexus blocks) for hip and knee surgery may offer
advantages as a rst strategy to balance the risk of hematoma
in anticoagulated patients compared with epidural analgesia.
The results from a systematic review of randomized controlled
trials support the use of peripheral nerve blocks as a rst
choice of analgesia after major orthopedic surgery; femoral
nerve block provided a similar analgesic prole to epidural
analgesia, with fewer side-effects after total knee replacement surgery [86]. Lumbar plexus block represents the rst analgesic
choice after total hip replacement surgery [87]. However, both
lumbar plexus and femoral nerve blocks are still associated
with loss of quadricep muscle strength [88,89]. A study has
suggested that an adductor canal block after knee surgery
may provide a similar analgesic prole but without quadricep
weakness after total knee replacement surgery [88]. Although
no difference was observed between the 2 anesthesia techniques in terms of morphine use or level of pain, quadricep
strength was signicantly better preserved with an adductor
canal block than with a femoral nerve block (52% median of
baseline vs 18%, P = .004) [88]. Peripheral nerve blocks have
been associated with a potential for wound hematoma with
administration of heparin in some studies [57,9093] but not
others [94]. The potential severity of a hematoma in a noncompressible site and the lack of evidence on which to base
recommendations for the particular use of continuous peripheral nerve blocks have led the ESA and ASRA guidelines to
suggest expanding the clinical recommendations for neuraxial
anesthesia to deeper peripheral nerve blocks, such as lumbar
plexus [41,57].

G. Cappelleri, A. Fanelli
Studies specically investigating the use of apixaban after
non-neuraxial blocks in orthopedic surgery seem to be lacking.
However, there are some available data for rivaroxaban
and dabigatran. In a prospective observational study of 504
patients undergoing total knee replacement surgery who had
continuous femoral nerve block and whose femoral catheter
was removed 20 hours after starting rivaroxaban (10 mg once
daily), no incidences of hematoma causing neurovascular
compromise at the femoral catheter site or groin area were
reported [95]. Neither was rivaroxaban associated with an
increase in major bleeding in an observational analysis of
766 patients undergoing total knee or hip replacement surgery
and femoral, sciatic, and lumbar plexus blocks, although 3
hematomas in the operated knee requiring surgical evacuation
were noted during rivaroxaban administration [96].
In a post hoc pooled analysis of 3 phase III clinical trials of
dabigatran (220 mg or 150 mg once daily) versus enoxaparin
for VTE prevention after knee or hip replacement surgery, in
which 19% (n = 1539) received a combination of general anesthesia or neuraxial anesthesia plus peripheral nerve block, rates
of major VTE and VTE-related mortality were slightly higher
with general anesthesia compared with neuraxial or combination anesthesia, although not signicantly so, and neither were
there signicant differences in efcacy or safety outcomes
between dabigatran and enoxaparin [63].

7. Conclusions
Patients undergoing elective major orthopedic surgery
receive thromboprophylaxis because of their increased risk
of developing VTE. With the overall aging of the population,
more patients undergoing major orthopedic surgery are likely
to be receiving anticoagulation therapy for thromboembolic
disorders prior to their surgical procedure. Several direct
OACs are currently approved for different therapeutic indications, including prevention of VTE after elective knee or hip
replacement surgery, and their use in clinical practice has been
growing. Effective management of these direct OACs and
regional anesthesia is essential to avoid bleeding complications and is, therefore, a topic of crucial interest for anesthesiologists. Thromboprophylaxis with all direct OACs may start
between 1 and 24 hours after surgery [6,8,10], provided that
hemostasis has been achieved, and thus should present a lower
theoretical risk of bleeding or neuraxial hematoma in case of
central blocks, in comparison with preoperatively initiated
LMWH. After surgery, restarting direct OACs can be delayed
by up to 48 hours in cases of bleeding or a difcult neuraxial
block, and protection against thromboembolic events is
achieved within hours after the rst dose of these agents and
without the need for bridging therapy. The effect of the type
of anesthesia on clinical outcomes of the direct OACs is still
unclear. Subanalyses of RE-MODEL, RE-NOVATE, and
RE-MOBILIZE data suggest that, irrespective of the anesthesia used, neuraxial anesthesia was associated with a lower risk

Anticoagulation and anesthesia


of thromboembolic events and VTE-related mortality than
general anesthesia [63]. Conversely, a subanalysis of pooled
RECORD data found no signicant difference in the risk of
venous thromboembolic events between these types of
anesthesia used [62]. A growing body of clinical study and
real-world data, particularly with rivaroxaban, suggests that
neuraxial anesthesia can be undertaken in patients undergoing
major orthopedic surgery without increasing the risk of
compressive hematoma, provided that guideline recommendations are followed and bleeding risk is evaluated on an individual basis.

Acknowledgements
The authors would like to acknowledge Li Wan for
editorial support, with funding from Bayer HealthCare
Pharmaceuticals and Janssen Scientic Affairs, LLC.

References
[1] Deitelzweig SB, Johnson BH, Lin J, Schulman KL. Prevalence of
clinical venous thromboembolism in the USA: current trends and future
projections. Am J Hematol 2011;86:217-20.
[2] MacDougall DA, Feliu AL, Boccuzzi SJ, Lin J. Economic burden of
deep-vein thrombosis, pulmonary embolism, and post-thrombotic
syndrome. Am J Health Syst Pharm 2006;63:S5-S15.
[3] Spyropoulos A, Lin J. Direct medical costs of venous thromboembolism
and subsequent hospital readmission rates: an administrative claims
analysis from 30 managed care organizations. J Manag Care Pharm
2007;13:475-86.
[4] Falck-Ytter Y, Francis CW, Johanson NA, Curley C, Dhal OE,
Schulman S, et al. Prevention of VTE in orthopedic surgery patients:
antithrombotic therapy and prevention of thrombosis, 9th ed: American
College of Chest Physicians evidence-based clinical practice guidelines.
Chest 2012;141:e278S-325S.
[5] Douketis JD, Spyropoulos AC, Spencer FA, Mayr M, Jaffer AK,
Eckman MH, et al. Perioperative management of antithrombotic therapy:
antithrombotic therapy and prevention of thrombosis, 9th ed: American
College of Chest Physicians evidence-based clinical practice guidelines.
Chest 2012;141:e326S-50S.
[6] Bristol-Myers Squibb, Pzer. Eliquis Summary of Product Characteristics.
http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_Information/human/002148/WC500107728.pdf [accessed 26/11/2015].
[7] Bristol-Myers Squibb Company, Pzer Inc. Eliquis Prescribing
Information. http://packageinserts.bms.com/pi/pi_eliquis.pdf [accessed
16/10/2015].
[8] Boehringer Ingelheim International GmbH. Pradaxa Summary of Product Characteristics. http://www.ema.europa.eu/docs/en_GB/document_
library/EPAR_-_Product_Information/human/000829/WC500041059.
pdf [accessed 26/11/2015].
[9] Boehringer Ingelheim Pharmaceuticals Inc. Pradaxa Prescribing
Information. http://bidocs.boehringer-ingelheim.com/BIWebAccess/
ViewServlet.ser?docBase=renetnt&folderPath=/Prescribing%20Information/PIs/Pradaxa/Pradaxa.pdf [accessed 16/10/2015].
[10] Bayer Pharma AG. Xarelto Summary of Product Characteristics. http://
www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Product_
Information/human/000944/WC500057108.pdf [accessed 25/11/2015].

233
[11] Janssen Pharmaceuticals Inc. Xarelto Prescribing Information. http://
www.xareltohcp.com/sites/default/les/pdf/xarelto_0.pdf [accessed 16/
10/2015].
[12] Frost C, Wang J, Nepal S, Schuster A, Barret YC, Mosqueda-Garcia R,
et al. Apixaban, an oral, direct factor Xa inhibitor: single-dose safety,
pharmacokinetics, pharmacodynamics and food effect in healthy subjects. Br J Clin Pharmacol 2013;75:476-87.
[13] Kubitza D, Becka M, Voith B, Zuehlsdorf M, Wensing G. Safety, pharmacodynamics, and pharmacokinetics of single doses of BAY 59-7939,
an oral, direct factor Xa inhibitor. Clin Pharmacol Ther 2005;78:412-21.
[14] Kubitza D, Becka M, Wensing G, Voith B, Zuehlsdorf M. Safety,
pharmacodynamics, and pharmacokinetics of BAY 59-7939 an oral,
direct factor Xa inhibitor after multiple dosing in healthy male subjects.
Eur J Clin Pharmacol 2005;61:873-80.
[15] Blech S, Ebner T, Ludwig-Schwellinger E, Stangier J, Roth W.
The metabolism and disposition of the oral direct thrombin inhibitor,
dabigatran, in humans. Drug Metab Dispos 2008;36:386-99.
[16] Upreti W, Wang J, Barrett YC, Byon W, Boyd RA, Pursley J, et al. Effect
of extremes of body weight on the pharmacokinetics, pharmacodynamics, safety and tolerability of apixaban in healthy subjects. Br J Clin Pharmacol 2013;76:908-16.
[17] Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Body weight has limited
inuence on the safety, tolerability, pharmacokinetics, or pharmacodynamics
of rivaroxaban (BAY 59-7939) in healthy subjects. J Clin Pharmacol 2007;
47:218-26.
[18] Kubitza D, Becka M, Roth A, Mueck W. The inuence of age and gender
on the pharmacokinetics and pharmacodynamics of rivaroxaban an oral,
direct factor Xa inhibitor. J Clin Pharmacol 2013;53:249-55.
[19] Liesenfeld KH, Lehr T, Dansirikul C, Reilly PA, Connolly SJ, Ezekowitz
MD, et al. Population pharmacokinetic analysis of the oral thrombin inhibitor dabigatran etexilate in patients with non-valvular atrial brillation
from the RE-LY trial. J Thromb Haemost 2011;9:2168-75.
[20] Frost CE, Song Y, Shenker A, Wang J, Barrett YC, Schuster A, et al. Effects of age and sex on the single-dose pharmacokinetics and pharmacodynamics of apixaban. Clin Pharmacokinet 2015;54:651-62.
[21] Mueck W, Stampfuss J, Kubitza D, Becka M. Clinical pharmacokinetic
and pharmacodynamic prole of rivaroxaban. Clin Pharmacokinet
2014;53:1-16.
[22] Lassen MR, Raskob GE, Gallus A, Pineo G, Chen D, Portman RJ.
Apixaban or enoxaparin for thromboprophylaxis after knee replacement.
N Engl J Med 2009;361:594-604.
[23] Lassen MR, Raskob GE, Gallus A, Pineo G, Chen D, Hornick P, et al.
Apixaban versus enoxaparin for thromboprophylaxis after knee replacement (ADVANCE-2): a randomised double-blind trial. Lancet 2010;375:
807-15.
[24] Lassen MR, Gallus A, Raskob GE, Pineo G, Chen D, Ramirez LM, et al.
Apixaban versus enoxaparin for thromboprophylaxis after hip replacement. N Engl J Med 2010;363:2487-98.
[25] Cohen A, Drost P, Marchant N, Mitchell S, Orme M, Rublee D, et al. The
efcacy and safety of pharmacological prophylaxis of venous thromboembolism following elective knee or hip replacement: systematic review
and network meta-analysis. Clin Appl Thromb Hemost 2012;18:611-27.
[26] Wolowacz SE, Roskell NS, Plumb JM, Caprini JA, Eriksson BI. Efcacy
and safety of dabigatran etexilate for the prevention of venous thromboembolism following total hip or knee arthroplasty. A meta-analysis.
Thromb Haemost 2009;101:77-85.
[27] Turpie AG, Lassen MR, Eriksson BI, Gent M, Berkowitz SD, Misselwitz
F, et al. Rivaroxaban for the prevention of venous thromboembolism after hip or knee arthroplasty. Pooled analysis of four studies. Thromb Haemost 2011;105:444-53.
[28] Ageno W, Gallus AS, Wittkowsky A, Crowther M, Hylek EM, Palareti
G. Oral anticoagulant therapy: antithrombotic therapy and prevention
of thrombosis, 9th ed: American College of Chest Physicians evidencebased clinical practice guidelines. Chest 2012;141:e44S-88S.
[29] Rudd KM, Phillips EL. New oral anticoagulants in the treatment of
pulmonary embolism: efcacy, bleeding risk, and monitoring. Thrombosis 2013;2013:973710.

234
[30] Garcia D, Libby E, Crowther MA. The new oral anticoagulants. Blood
2010;115:15-20.
[31] Rodgers A, Walker N, Schug S, McKee A, Kehlet H, van Zundert A,
et al. Reduction of postoperative mortality and morbidity with epidural
or spinal anaesthesia: results from overview of randomised trials. Br
Med J 2000;321:1493.
[32] Allen DJ, Chae-Kim SH, Trousdale DM. Risks and complications of
neuraxial anesthesia and the use of anticoagulation in the surgical patient.
Proc (Baylor Univ Med Cent) 2002;15:369-73.
[33] Green L, Machin SJ. Managing anticoagulated patients during neuraxial
anaesthesia. Br J Haematol 2010;149:195-208.
[34] Jabbari A, Alijanpour E, Mir M, Bani HN, Rabiea SM, Rupani MA. Post
spinal puncture headache, an old problem and new concepts: review of articles about predisposing factors. Caspian J Intern Med 2013;4:595-602.
[35] Schwabe K, Hopf HB. Persistent back pain after spinal anaesthesia in the
non-obstetric setting: incidence and predisposing factors. Br J Anaesth
2001;86:535-9.
[36] Jeng CL, Torrillo TM, Rosenblatt MA. Complications of peripheral
nerve blocks. Br J Anaesth 2010;105(Suppl. 1):i97-107.
[37] Agarwal A, Kishore K. Complications and controversies of regional anaesthesia: a review. Indian J Anaesth 2009;53:543-53.
[38] Auroy Y, Benhamou D, Bargues L, Ecoffey C, Falissard B, Mercier FJ,
et al. Major complications of regional anesthesia in France: the SOS Regional Anesthesia Hotline Service. Anesthesiology 2002;97:1274-80.
[39] Barrington MJ, Watts SA, Gledhill SR, Thomas RD, Said SA, Snyder
GL, et al. Preliminary results of the Australasian Regional Anaesthesia
Collaboration: a prospective audit of more than 7000 peripheral nerve
and plexus blocks for neurologic and other complications. Reg Anesth
Pain Med 2009;34:534-41.
[40] Stundner O, Chiu YL, Sun X, Mazumdar M, Fleischut P, Poultsides L,
et al. Comparative perioperative outcomes associated with neuraxial versus general anesthesia for simultaneous bilateral total knee arthroplasty.
Reg Anesth Pain Med 2012;37:638-44.
[41] Horlocker TT, Wedel DJ, Rowlingson JC, Enneking FK, Kopp SL, Benzon HT, et al. Regional anesthesia in the patient receiving antithrombotic
or thrombolytic therapy: American Society of Regional Anesthesia and
Pain Medicine evidence-based guidelines (third edition). Reg Anesth
Pain Med 2010;35:64-101.
[42] Stafford-Smith M. Impaired haemostasis and regional anaesthesia. Can J
Anaesth 1996;43:R129-41.
[43] Horlocker TT, Heit JA. Low molecular weight heparin: biochemistry,
pharmacology, perioperative prophylaxis regimens, and guidelines for
regional anesthetic management. Anesth Analg 1997;85:874-85.
[44] Pitkanen MT, Aromaa U, Cozanitis DA, Forster JG. Serious complications associated with spinal and epidural anaesthesia in Finland from
2000 to 2009. Acta Anaesthesiol Scand 2013;57:553-64.
[45] Pumberger M, Memtsoudis SG, Stundner O, Herzog R, Boettner F,
Gausden E, et al. An analysis of the safety of epidural and spinal neuraxial anesthesia in more than 100,000 consecutive major lower extremity
joint replacements. Reg Anesth Pain Med 2013;38:515-9.
[46] Oger E, Bressollette L, Nonent M, Lacut K, Guias B, Couturaud F, et al.
High prevalence of asymptomatic deep vein thrombosis on admission in
a medical unit among elderly patients. Thromb Haemost 2002;88:592-7.
[47] Heit JA, Silverstein MD, Mohr DN, Petterson TM, Lohse CM, O'Fallon
WM, et al. The epidemiology of venous thromboembolism in the community. Thromb Haemost 2001;86:452-63.
[48] Levine MN, Raskob G, Landefeld S, Kearon C. Hemorrhagic complications of anticoagulant treatment. Chest 2001;119:108S-21S.
[49] Landefeld CS, Beyth RJ. Anticoagulant-related bleeding: clinical epidemiology, prediction, and prevention. Am J Med 1993;95:315-28.
[50] Mangoni AA, Jackson SH. Age-related changes in pharmacokinetics and
pharmacodynamics: basic principles and practical applications. Br J Clin
Pharmacol 2004;57:6-14.
[51] Lopez-Jimenez L, Montero M, Gonzalez-Fajardo JA, Arcelus JI, Surez
C, Lobo JL, et al. Venous thromboembolism in very elderly patients:
ndings from a prospective registry (RIETE). Haematologica 2006;91:
1046-51.

G. Cappelleri, A. Fanelli
[52] Lacut K, Le Gal G, Mottier D. Primary prevention of venous thromboembolism in elderly medical patients. Clin Interv Aging 2008;3:399-411.
[53] Vandermeulen EP, Van Aken H, Vermylen J. Anticoagulants and spinalepidural anesthesia. Anesth Analg 1994;79:1165-77.
[54] Ruff RL, Dougherty JH Jr. Complications of lumbar puncture followed
by anticoagulation. Stroke 1981;12:879-81.
[55] Horlocker TT, Wedel DJ. Neuraxial block and low-molecular-weight
heparin: balancing perioperative analgesia and thromboprophylaxis.
Reg Anesth Pain Med 1998;23:164-77.
[56] Bettelli G. Anaesthesia for the elderly outpatient: preoperative assessment and evaluation, anaesthetic technique and postoperative pain management. Curr Opin Anaesthesiol 2010;23:726-31.
[57] Gogarten W, Vandermeulen E, Van Aken H, Kozek S, Llau JV, Samama
CM. Regional anaesthesia and antithrombotic agents: recommendations
of the European Society of Anaesthesiology. Eur J Anaesthesiol 2010;
27:999-1015.
[58] Moen V, Dahlgren N, Irestedt L. Severe neurological complications after
central neuraxial blockades in Sweden 1990-1999. Anesthesiology 2004;
101:950-9.
[59] Serebruany VL, Steinhubl SR, Berger PB, Malinin AI, Bhatt DL, Topol
EJ. Variability in platelet responsiveness to clopidogrel among 544 individuals. J Am Coll Cardiol 2005;45:246-51.
[60] Baron TH, Kamath PS, McBane RD. Management of antithrombotic
therapy in patients undergoing invasive procedures. N Engl J Med
2013;368:2113-24.
[61] Heidbuchel H, Verhamme P, Alings M, Antz M, Hacke W, Oldgren J,
et al. European Heart Rhythm Association practical guide on the use of
new oral anticoagulants in patients with non-valvular atrial brillation.
Europace 2013;15:625-51.
[62] Rosencher N, Llau JV, Mueck W, Loewe A, Berkowitz SD, Homering
M. Incidence of neuraxial haematoma after total hip or knee surgery:
RECORD programme (rivaroxaban vs. enoxaparin). Acta Anaesthesiol
Scand 2013;57:565-72.
[63] Rosencher N, Noack H, Feuring M, Clemens A, Friedman RJ, Eriksson
BI. Type of anaesthesia and the safety and efcacy of thromboprophylaxis with enoxaparin or dabigatran etexilate in major orthopaedic surgery: pooled analysis of three randomized controlled trials. Thromb J
2012;10:9.
[64] Breivik H, Bang U, Jalonen J, Vigfsson G, Alahuhta S, Lagerkranser M.
Nordic guidelines for neuraxial blocks in disturbed haemostasis from the
Scandinavian Society of Anaesthesiology and Intensive Care Medicine.
Acta Anaesthesiol Scand 2010;54:16-41.
[65] Benzon HT, Avram MJ, Green D, Bonow RO. New oral anticoagulants
and regional anaesthesia. Br J Anaesth 2013;111(Suppl. 1):i96-113.
[66] Rosencher N, Bonnet MP, Sessler DI. Selected new antithrombotic
agents and neuraxial anaesthesia for major orthopaedic surgery: management strategies. Anaesthesia 2007;62:1154-60.
[67] Connolly G, Spyropoulos AC. Practical issues, limitations, and periprocedural management of the NOAC's. J Thromb Thrombolysis 2013;36:
212-22.
[68] Liew A, Douketis J. Perioperative management of patients who are receiving a novel oral anticoagulant. Intern Emerg Med 2013;8:477-84.
[69] Imberti D, Ambrosoli A, Cimminiello C, Compagnone C, Fanelli A, Tripodi A, et al. Periprocedural management of rivaroxaban-treated patients.
Expert Opin Pharmacother 2015;16:685-91.
[70] van Ryn J, Stangier J, Haertter S, Liesenfeld KH, Wienen W, Feuring M,
et al. Dabigatran etexilate - a novel, reversible, oral direct thrombin inhibitor: interpretation of coagulation assays and reversal of anticoagulant activity. Thromb Haemost 2010;103:1116-27.
[71] Heidbuchel H, Verhamme P, Alings M, Antz M, Diener HC, Hacke W,
et al. Updated European Heart Rhythm Association practical guide on the
use of non-vitamin K antagonist anticoagulants in patients with nonvalvular atrial brillation. Europace 2015;17:1467-507.
[72] Becker RC, Yang H, Barrett Y, Mohan P, Wang J, Wallentin L, et al.
Chromogenic laboratory assays to measure the factor Xa-inhibiting properties of apixaban - an oral, direct and selective factor Xa inhibitor. J
Thromb Thrombolysis 2011;32:183-7.

Anticoagulation and anesthesia


[73] Baglin T, Keeling D, Kitchen S. Effects on routine coagulation screens
and assessment of anticoagulant intensity in patients taking oral dabigatran or rivaroxaban: guidance from the British Committee for Standards
in Haematology. Br J Haematol 2012;159:427-9.
[74] Lindhoff-Last E, Samama MM, Ortel TL, Weitz JI, Spiro TE. Assays for
measuring rivaroxaban: their suitability and limitations. Ther Drug Monit
2010;32:673-9.
[75] Bates SM, Weitz JI. Coagulation assays. Circulation 2005;112:e53-60.
[76] Samama MM, Contant G, Spiro TE, Perzborn E, Le Flem L, Guinet C,
et al. Laboratory assessment of rivaroxaban: a review. Thromb J 2013;
11:11.
[77] Samama MM, Contant G, Spiro TE, Perzborn E, Guinet C, Gourmelin Y,
et al. 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.
[78] Haas S, Bode C, Norrving B, Turpie AGG. Practical guidance for using
rivaroxaban in patients with atrial brillation: balancing benet and risk.
Vasc Health Risk Manag 2014;10:101-14.
[79] Tripodi A, Chantarangkul V, Guinet C, Samama MM. The international
normalized ratio calibrated for rivaroxaban has the potential to normalize
prothrombin time results for rivaroxaban-treated patients: results of an
in vitro study. J Thromb Haemost 2011;9:226-8.
[80] Pfeilschifter W, Luger S, Brunkhorst R, Lindhoff-Last E, Foerch C. The
gap between trial data and clinical practice an analysis of case reports
on bleeding complications occurring under dabigatran and rivaroxaban
anticoagulation. Cerebrovasc Dis 2013;36:115-9.
[81] Stangier J, Feuring M. Using the HEMOCLOT direct thrombin inhibitor
assay to determine plasma concentrations of dabigatran. Blood Coagul
Fibrinolysis 2012;23:138-43.
[82] Tripodi A. The laboratory and the new oral anticoagulants. Clin Chem
2013;59:353-62.
[83] Kozek-Langenecker S. Management of massive operative blood loss.
Minerva Anestesiol 2007;73:401-15.
[84] Meybohm P, Zacharowski K, Weber CF. Point-of-care coagulation management in intensive care medicine. Crit Care 2013;17:218.
[85] Horlocker TT. Pain management in total joint arthroplasty: a historical review. Orthopedics 2010;33:14-9.

235
[86] Fowler SJ, Symons J, Sabato S, Myles PS. Epidural analgesia compared
with peripheral nerve blockade after major knee surgery: a systematic review and meta-analysis of randomized trials. Br J Anaesth 2008;100:
154-64.
[87] The PROSPECT Group. Procedure specic postoperative pain management (PROSPECT) nal recommendations; Overall Recommendations
for Total Hip Arthroplasty, 11. http://www.postoppain.org/sections/?
root_id=8861&section=3# [accessed 03/11/2015].
[88] Jaeger P, Zaric D, Fomsgaard JS, Hilsted KL, Bjerregaard J, Gyrn J, et al.
Adductor canal block versus femoral nerve block for analgesia after total
knee arthroplasty: a randomized, double-blind study. Reg Anesth Pain
Med 2013;38:526-32.
[89] Ilfeld BM, Duke KB, Donohue MC. The association between lower extremity continuous peripheral nerve blocks and patient falls after knee
and hip arthroplasty. Anesth Analg 2010;111:1552-4.
[90] Aveline C, Bonnet F. Delayed retroperitoneal haematoma after failed
lumbar plexus block. Br J Anaesth 2004;93:589-91.
[91] Bickler P, Brandes J, Lee M, Bozic K, Chesbro B, Claassen J. Bleeding
complications from femoral and sciatic nerve catheters in patients receiving low molecular weight heparin. Anesth Analg 2006;103:1036-7.
[92] Gogarten W. The inuence of new antithrombotic drugs on regional anesthesia. Curr Opin Anaesthesiol 2006;19:545-50.
[93] Wiegel M, Gottschaldt U, Hennebach R, Hirschberg T, Reske A. Complications and adverse effects associated with continuous peripheral
nerve blocks in orthopedic patients. Anesth Analg 2007;104:1578-82
[table].
[94] Chelly JE, Schilling D. Thromboprophylaxis and peripheral nerve
blocks in patients undergoing joint arthroplasty. J Arthroplast 2008;23:
350-4.
[95] Idestrup C, Sawhney M, Nix C, Kiss A. The incidence of hematoma
formation in patients with continuous femoral catheters following
total knee arthroplasty while receiving rivaroxaban as thromboprophylaxis: an observational study. Reg Anesth Pain Med 2014;39:
414-7.
[96] Chelly JE, Metais B, Schilling D, Luke C, Taormina D. Combination of
supercial and deep blocks with rivaroxaban. Pain Med 2015;16:
2024-30.

Das könnte Ihnen auch gefallen