You are on page 1of 24

Europace (2011) 13, 723–746 EHRA POSITION PAPER

doi:10.1093/europace/eur126

Bleeding risk assessment and management


in atrial fibrillation patients: a position document
from the European Heart Rhythm Association,
endorsed by the European Society of Cardiology

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


Working Group on Thrombosis
Gregory Y.H. Lip (Chair) 1*†, Felicita Andreotti 2†‡, Laurent Fauchier 3†, Kurt Huber 4*†,
Elaine Hylek 5†, Eve Knight 6†, Deirdre A. Lane 1†, Marcel Levi 7†, Francisco Marin 8†,
Gualtiero Palareti9†, and Paulus Kirchhof (Co-chair)10†
Document reviewers: Jean-Philippe Collet 11, Andrea Rubboli 12, Daniela Poli 13, and John Camm 14

1
University of Birmingham Centre for Cardiovascular Sciences, City Hospital, Birmingham B18 7QH, UK; 2Department of Cardiovascular Medicine, ‘A. Gemelli’ University Hospital,
Rome, Italy; 3Cardiologie B, Centre Hospitalier Universitaire Trousseau et Université François Rabelais, Tours, France; 43rd Department of Medicine, Cardiology and Emergency
Medicine, Wilhelminenhospital, A-1160 Vienna, Austria; 5Department of Medicine, Research Unit-Section of General Internal Medicine, Boston University Medical Center, Boston, MA
02118, USA; 6AntiCoagulation, Europe; 7Department of Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands; 8Department of Cardiology,
Hospital Universitario Virgen de la Arrixaca, Murcia, Spain; 9Department of Angiology and Blood Coagulation, University Hospital S. Orsola-Malpighi, Bologna, Italy; 10Department of
Cardiology and Angiology, Universitätsklinikum Münster, D-48149 Münster, Germany; 11Groupe Hopitalier Pitie-Salpetriere, Paris, France; 12Maggiore Hospital, Bologna, Italy;
13
Department of Heart and Vessels, Thrombosis Center, Florence, Italy; and 14Division of Cardiac and Vascular Sciences, St.George’s University of London, London, UK

Despite the clear net clinical benefit of oral anticoagulation (OAC) in atrial fibrillation (AF) patients at risk for stroke, major bleeding events
(especially intra-cranial bleeds) may be devastating events when they do occur. The decision for OAC is often based on a careful assessment
of both stroke risk and bleeding risk, but clinical scores for bleeding risk estimation are much less well validated than stroke risk scales. Also,
the estimation of bleeding risk is rendered difficult since many of the known factors that increase bleeding risk overlap with stroke risk
factors. As well as this, many factors that increase bleeding risk are transient, such as variable international normalized ratio values, oper-
ations, vascular procedures, or drug –drug and food –drug interactions.
In this Position Document, we comprehensively review the published evidence and propose a consensus on bleeding risk assessments
in AF patients, with a view to summarizing ‘best practice’ when approaching antithrombotic therapy in AF patients. We address the epide-
miology and size of the problem of bleeding risk in AF and review established bleeding risk factors. We also summarize definitions of bleeding
in the published literature. Patient values and preferences balancing the risk of bleeding against thrombo-embolism is reviewed, and the prog-
nostic implications of bleeding are discussed. We also review bleeding risk stratification and currently published bleeding risk schema. A brief
discussion of special situations [e.g. peri-ablation, peri-devices (implantable cardioverter-defibrillator, pacemakers) and presentation with
acute coronary syndromes and/or requiring percutaneous coronary interventions/stents and bridging therapy], as well as a discussion of pre-
vention of bleeds and managing bleeding complications, is made. Finally, this document also puts forwards consensus statements that may
help to define evidence gaps and assist in everyday clinical practice.
Bleeding risk is almost inevitably lower than stroke risk in patients with atrial fibrillation. Nonetheless, identification of patients at high risk of bleed-
ing and delineation of conditions and situations associated with bleeding risk can help to refine antithrombotic therapy to minimize bleeding risk.
-----------------------------------------------------------------------------------------------------------------------------------------------------------
Keywords Bleeding † Oral anticoagulation † Atrial fibrillation † Risk assessment † Stroke prevention † Antithrombotic
therapy † Triple therapy

* Corresponding author. Tel: +44 121 5075080; Fax: +44 121 507 4907; Email: g.y.h.lip@bham.ac.uk

Task Force Members.

Representing the ESC Working Group on Thrombosis.
Published on behalf of the European Society of Cardiology. All rights reserved. & The Author 2011. For permissions please email: journals.permissions@oup.com.
724 G.Y.H. Lip et al.

aimed to review the published evidence and to propose a consensus


Introduction and scope on bleeding risk assessment in AF patients, with a view to
Prevention of stroke and thrombo-embolism is one of the main summarizing ‘best practice’. The present document summarizes
therapeutic goals in atrial fibrillation (AF).1,2 Oral anticoagulation the available evidence and put forwards consensus statements that
(OAC) is highly effective in preventing ischaemic strokes in patients may help to define evidence gaps and assist in everyday clinical
with AF and conveys a clear net clinical benefit despite a potential practice.
risk for major bleeding events.3 The ultimate judgement regarding care of a particular patient
Currently, anticoagulation therapy manily consists of vitamin K must be made by the healthcare provider and the patient in light
antagonists (VKAs), often warfarin or acenocoumarol or phenpro- of all of the circumstances presented by that patient.
coumon, which are dose adjusted to achieve an international nor- Literature searches were conducted in the following databases:
malized ratio (INR) of 2.0 –3.0.2 The VKAs have many limitations, PubMed/MEDLINE and the Cochrane Library (including the
including a significant inter- and intra-patient variability of effective Cochrane Database of Systematic Reviews and the Cochrane Con-
dose, and various food and drug interactions. Thus, regular antic- trolled Trials Registry). Searches focused on English-language

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


oagulation monitoring is required in all patients to keep the INR sources and studies in human subjects. Articles related to animal
within the narrow therapeutic range of 2.0 –3.0. Furthermore, experimentation were only cited when the information was impor-
this variability causes many patients to spend significant amounts tant for understanding pathophysiological concepts pertinent to
of time outside the therapeutic INR window, and an important patient management and comparable data were not available
proportion of patients discontinue OAC therapy.4 New OACs, from human studies. Additional information was requested from
broadly divided into two categories, the oral direct thrombin the authors where necessary.
inhibitors and the oral direct factor Xa inhibitors, are in advanced
clinical development, and may offer alternative therapies to
patients who suffer from the limitations and dis-utility associated
with VKAs.5 Indeed, indirect comparisons show how well these
Systematic review of evidence/
new OACs may perform relative to VKA, aspirin–clopidogrel data
combination therapy, aspirin monotherapy or placebo.6
Anticoagulant therapy with VKAs carries a risk for bleeding, Epidemiology and size of the problem
including severe bleeding events, but the clinical benefit of OAC of bleeding risk in atrial fibrillation
clearly outweighs the risk of OAC therapy, especially in patients The risk of stroke attributable to AF increases with age. For
at high risk for stroke: bleeding events are five to eight times example, among individuals aged 50–59 years old, 1.5% of
less likely than ischaemic strokes reported among AF patients strokes are attributable to AF compared with 23.5% of strokes
from trials and registry data.7 among individuals aged 80–89 years.12 Oral anticoagulation
Estimation of the stroke risk in an individual patient with AF can therapy greatly reduces the risk of stroke in AF, and the clinical
be achieved using easily applicable clinical stroke risk estimators dilemma faced by physicians and patients is anticoagulant-related
such as the CHADS2 score8 and an increasingly more refined haemorrhage, which also increases with age. It is of note that the
score that considers additional stroke risk factors, the rate of intra-cranial haemorrhage has quintupled in recent years,
CHA2DS2 –VASC [congestive heart failure, hypertension, age due to the expanded use of anticoagulants and antiplatelet
≥75 (doubled), diabetes, previous stroke (doubled)–vascular therapy in older adults.13 Perceived bleeding risk and older age
disease, age 65– 74, sex category] score.2,9 Patients at moder- are potent negative predictors of receiving warfarin and partly
ate–high risk for stroke are increasingly being considered explain the reported low rates of warfarin use in clinical prac-
candidates for OAC based on both an old and a new evidence tice.14,15 It is worth remembering that the risk of both haemorrhage
base.2,10 and stroke are highest when AF is newly diagnosed and during the
Despite the clear net clinical benefit of OAC to AF patients at risk initiation of anticoagulant medication.16 Nonetheless, one recent
for stroke, major bleeding events, especially intra-cranial bleeds, may study does not suggest that OAC naı̈ve status conferred a disadvan-
be devastating when they do occur.3,11 The decision for OAC should tage in relation to efficacy and bleeding endpoints.17,18
therefore be based on a careful assessment of both stroke risk and Reported rates of major bleeding among individuals with AF
bleeding risk. Unfortunately, clinical scores for bleeding risk estimation taking oral VKAs vary widely ranging from 1.3 to 7.2% per year
are much less well validated than stroke risk scales. Moreover, the (Table 1). These disparate rates reflect the variability in patient
estimation of bleeding risk is rendered difficult by other factors: population characteristics studied and the methodology employed.
The early trials in AF that established the efficacy of warfarin
(i) many of the known factors that increase bleeding risk overlap
excluded almost 90% of individuals screened. To establish drug
with stroke risk factors;
efficacy, efforts to minimize trial drop-out and cross-over are
(ii) many factors that increase bleeding risk are transient, such as
imperative. Trial participants, therefore, are often selected based
variable INR values, operations, vascular procedures, and
on a lower bleeding risk profile and higher likelihood of adherence.
drug –drug or food –drug interactions.
For these reasons, bleeding rates reported from randomized trials
In recognizing this problem, the European Heart Rhythm Associ- will often be lower than in clinical practice. Randomized controlled
ation (EHRA) and the European Society of Cardiology (ESC) trials designed to evaluate the safety and efficacy of new
Working Group on Thrombosis convened a Task Force, which anti-thrombotic agents should also include substantial numbers
Bleeding risk assessment and management in atrial fibrillation patients 725

Table 1 Annual rates of major haemorrhage among patients taking warfarin

Study Year published Population (n) Major haemorrhage, ICH % per year New to Age, mean
% per year warfarin, %
...............................................................................................................................................................................
Randomised trials
AFI18 1994 AF (n ¼ 3691) 1.3 0.3 100 69
SPAF II19 (2 age strata) 1994 AF (n ¼ 715) 1.7 0.5 100 NR
AF (n ¼ 385) 4.2 1.8 100 80
AFFIRM20 2002 AF (n ¼ 4060) 2.0 0.6 NR 70
SPORTIF III21 2003 AF (n ¼ 3407) 2.2 0.4 27 70
SPORTIF V22 2005 AF (n ¼ 3422) 3.4 0.1 15 72
ACTIVE W23 2006 AF (n ¼ 6706) 2.2 NR 23 71

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


RE-LY24 2009 AF (n ¼ 18006) 3.4 0.74 51 72
ROCKET-AF25 Presented 2010 AF (n ¼ 14264) 3.5 0.7 37 73
Inception cohort
Landefeld and Goldman26 1989 All (n ¼ 565) 7.4 1.3 100 61
Steffensen et al. 27 1997 All (n ¼ 682) 6.0 1.3 100 59F/66M
Beyth et al. 28 1998 All (n ¼ 264) 5.0 0.9 100 60
Pengo et al. 29 2001 AF (n ¼ 433) Age ≥ 75: 5.1 NA 100 68
Age , 75: 1.0
Hylek et al. 30 2007 AF (n ¼ 472) 7.2 2.5 100 77
Non-inception cohort (prevalent warfarin use)
Van der Meeret al. 31 1993 All (n ¼ 6814) 2.7 1.3 NR 66
Fihn et al. 32 1996 All (n ¼ 928) 1.0 1.3 NR 58
ATRIA33 2003 AF (n ¼ 6320) 1.52 0.46 NR 71
Poli et al. 34 2009 AF (n ¼ 783) 1.4 2.5 NR 75
Rose et al. 35 2009 AF (n ¼ 3396) 1.9 NA 5 74

of patients without prior exposure to anticoagulation since these quality of monitoring seem to have the most important roles in
individuals are at the highest risk for bleeding and explaining such differences. At least in part, the difference in
thrombo-embolism.17 Observational studies are also subject to reported rates, however, can also be attributed to the diverse
selection bias and methodological differences. classification of bleeding events (major, life-threatening, and
Thus, to more fully interpret bleeding rates from real-world minor) adopted in each study. For example, large differences are
observational cohorts of AF patients, it is important to know the found in the various definitions as regards the decrease in haemo-
proportion of patients within the defined AF population taking globin level required for a bleed to be considered as ‘major’.40
warfarin. This proportion reflects individual physician judgement The various definitions appear to have different validities
of VKA candidacy or eligibility, which is often subjective. Prospec- depending on the clinical situation in which the antithrombotic
tive registries that require written informed consent for partici- drug is being used, complicating the formulation of a single univer-
pation are less likely to enroll the more acutely ill, medically sal bleeding definition. This is particularly true now that several
complex, or frail individuals, and thus will also underestimate the trials have incorporated the rate of major bleeding as a component
bleeding that occurs in routine care. Indeed, rates reported from of the primary study endpoints.
these studies are significantly lower than rates of haemorrhage Heterogeneous definitions are frequently observed in the trials
from recent studies that have enrolled older individuals and assessing the benefits of antithrombotic drugs in acute coronary
more first-time takers of warfarin (warfarin naı̈ve).34 – 38 syndromes (ACS), with the TIMI (thrombolysis in myocardial
Available data suggest that strokes in AF patients are more infarction) and GUSTO being the two bleeding definitions most
severe than other types of stroke, and more often result in perma- commonly used in trials on ACS.41,42 Different definitions are
nent disability or death than in non-AF stroke.39 Given the severity also used in studies on patients with other clinical conditions
of these presumably ischaemic strokes, determination of a bleeding (Table 2). The Academic Research Consortium has defined bleed-
risk threshold that would justify withholding anticoagulant therapy ing clinical endpoints in Coronary Stent Trials, as shown in
is difficult. Table 2.55 However, most studies focusing on bleeding events in
AF patients have used broadly similar definitions for major bleed-
Definitions of bleeding ing, as follows: fatal bleeding, bleeding requiring hospitalization or
The incidence of bleeding with OAC varies widely in published transfusion of ≥2 units of packed red blood cells, or bleeding
studies. Differences in study design, patient populations, and with involvement of a critical site (i.e. intra-cranial, retroperitoneal,
726 G.Y.H. Lip et al.

Table 2 Major bleeding definitions in clinical trials

Study or author, year Reference Definition of major bleeding


...............................................................................................................................................................................
(a) Acute coronary syndrome (ACS) trials
TIMI, 1987 41 Fatal, intra-cranial, bleeding associated with a decrease in Hb of at least 5 g/dL, transfusions are factored in 1
unit¼1 g/dL Hb, and cardiac tamponade
GUSTO, 1993 42 Intra-cerebral, bleeding associated with blood transfusion, or bleeding resulting in haemodynamic compromise
requiring treatment
ASSENT-3, 2001 43 Bleeding associated with blood transfusion and bleeding resulting in haemodynamic compromise
CURE, 2001 44 Major bleeding was subclassified as life threatening if it was fatal, resulted in a drop in haemoglobin
concentration of at least 5 g/dL, caused significant hypotension requiring intra-venous inotropes or surgical
intervention, resulted in symptomatic intra-cranial haemorrhage, or if four or more units of blood were
transfused

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


ACUITY, 2004 45 Intracranial or intra-ocular, drop in Hb of at least 4 g/dL without an overt source of bleeding, or of at least 3 g/dL
with an overt source of bleeding. Bleeding associated with blood transfusion. Haematoma ≥5 cm in
diameter, bleeding requiring re-operation, or access site haemorrhage requiring intervention
OASIS-5, 2006 46 Clinically overt bleeding that is fatal. Symptomatic intra-cranial, retroperitoneal, or intra-ocular. Overt bleeding
with a drop in haemoglobin of at least 3 g/dL. Overt bleeding associated with transfusion of 2 units of red
blood cells
PLATO, 2009 47 Fatal bleeding, intra-cranial bleeding, intra-peri-cardial bleeding with cardiac tamponade, hypovolemic shock or
severe hypotension due to bleeding, bleeding requiring pressors or surgery, bleeding associated with a drop
in haemoglobin of at least 5 g/dL or more, or associated with transfusion of 4 units of blood; or bleeding
either associated with a drop in haemoglobin of 3– 5 g/dL, bleeding associated with transfusion of 2– 3 units
of red cells. Intraocular bleeding with permanent vision loss
(b) Major bleeding definition in other conditions
Landefeld and Goldman, 26 Fatal, life threatening, potentially life threatening, leading to severe blood loss, to surgical treatment or to
1989 moderate blood loss (acute or subacute) not explained by trauma or surgery
Palareti, 1996 73 Fatal intra-cranial (documented by imaging); ocular (with blindness); articular; retroperitoneal; if surgery or
angiographic intervention are necessary to stop bleeding; haemoglobin drop of 2 g/dL or need for transfusion
of ≥2 blood units
Beyth et al., 1998 28 Overt bleeding leading to a drop in haemoglobin of 2 g/dL in 7 days or less, or life threatening
Kuijer et al., 1999 49 Clinically overt and associated with a decline in haemoglobin concentration of at least 2 g/dL, transfusion of 2
units or more of red blood cells, retroperitoneal or intra-cranial, warranted permanent discontinuation of
treatment
Wells et al., 2003 48 Loss of 2 units of blood in a 7-day period or bleeding otherwise life threatening
ISTH, 2005 58 Fall in haemoglobin of 2 g/dL or transfusion of 2 or more units of blood, bleeding that is symptomatic in a critical
organ (intra-cranial, intra-spinal, intra-ocular, retroperitoneal, intra-articular or peri-cardial, or intra-muscular
with compartment syndrome) or fatal
Aspinall et al., 2005 51 Haemodynamic instability, association with transfusion of blood, intra-cranial haemorrhage, or death (e.g. a
gastrointestinal bleed in a hypotensive patient, subdural haematoma)
Gage et al., 2006 52 International classification of disease-9th version codes for bleed in any location
Shireman et al., 2006 53 Hospitalization for ‘major acute bleeding’ (including gastrointestinal haemorrhage or intra-cranial haemorrhage
Ruiz-Gimenez et al., 54 Overt, requiring a transfusion of 2 or more units of blood, retroperitoneal, spinal, intra-cranial, fatal
2008
Poli et al., 2009 34 Fatal, intra-cranial (documented by imaging), ocular causing blindness, articular, or retroperitoneal; when
surgery or transfusion of more than two blood units were required or with haemoglobin drop of 2 g/dL or
more
(c) BARC (Bleeding Academic Research Consortium) bleeding definitions 55
Type 0 No bleeding
Type 1 Bleeding that is not actionable and does not cause the patient to seek unscheduled performance of studies, hospitalization, or
treatment by a health care professional
Type 2 (minor) Any overt sign of haemorrhage (e.g. more bleeding than would be expected for a clinical circumstance; including bleeding
found by imaging alone) that does not meet criteria for Type 3 BARC bleeding, Type 4 BARC bleeding (CABG-related), or
Type 5 BARC bleeding (fatal bleeding) that is actionable
Type 3 (major) a. BARC Type 3a bleeding
Intracranial haemorrhage (does not include microbleeds; does include intra-spinal). Subcategories; confirmed by autopsy or
imaging or LP
Intra-ocular compromising vision (even temporarily)
Overt bleeding plus haemoglobin drop .5 g/dL (provided haemoglobin drop is related to bleed)

Continued
Bleeding risk assessment and management in atrial fibrillation patients 727

Table 2 Continued

Study or author, year Reference Definition of major bleeding


...............................................................................................................................................................................
Tamponade
Bleeding requiring surgical or percutaneous intervention for control (exclude dental/nose/skin/haemorrhoids) or inotropes
b. BARC Type 3b bleeding
Any transfusion with overt bleeding
Overt bleeding plus haemoglobin drop 3 to 5 g/dL
Type 4 (CABG) BARC CABG-related bleeding definitions must include the same criteria for fatal bleeding, intra-cranial haemorrhage, need for
intervention to control bleeding and the number of transfusions as for BARC non-CABG related bleeding
Type 5 (Fatal) Fatal bleeding is bleeding that directly causes death with no other explainable cause. BARC fatal bleeding is categorized as
either definite or probable

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


CABG, coronary artery bypass graft surgery; PLATO, PLATelet inhibition and clinical Outcomes.

intra-spinal, intra-ocular, peri-cardial, or atraumatic intra-articular less relevant major bleeds would be less acute events, e.g. an
haemorrhage).56,57 asymptomatic haemoglobin drop and bleeding events that result
The subcommittee on control of anticoagulation of the Scientific in a temporary cessation of antithrombotic therapy. From a clini-
and Standardization Committee of the International Society of cal perspective, many bleeding events appear minor, as evidenced
Thrombosis and Haemostasis (ISTH) proposed a definition of by continuous antithrombotic therapy. Clinical decision making
bleeding complications in non-surgical patients that was recently may be made easier if such subgroups of major bleeding events
revisited.50,58 Most of the contemporary AF studies have been per- were reported in clinical trials.
formed according to this standardized definition. Although the
adoption of a single standardized definition would facilitate com-
parison of bleeding events across studies, it remains to be estab- Prognostic implications of bleeding
lished whether the proposed ISTH definition is more reliable Despite the varying clinical impact of different subtypes of ‘major’
than other existing definitions for identifying clinically relevant bleeds, major bleeding events are associated with a several-fold
bleeding episodes. Even the ISTH definition suffers from the risk of death for up to 1 year compared with non-bleeders, at
inclusion of a wide range of events with variable clinical relevance least in patients with acute coronary syndromes.59 – 66
to the patient, ranging from death or severely life-threatening Although extensive information regarding the prognostic
events to ‘laboratory indices of bleeding’ (2 g/dL Hb drop), impact of bleeding in patients with AF is lacking, the mechanisms
which may well be a normal variant, for example, in well-hydrated underlying the adverse prognosis associated with bleeding are
ACS patients undergoing percutaneous coronary interventions likely to be similar (at least in part) to those observed among
(PCI) with a small access site haematoma. ACS patients who develop a major bleed. Severe bleeding
events may be a marker of adverse outcomes, as well as a
How to interpret major bleeding rates in clinical practice cause of poor outcomes.
Clearly, there is a need to record major bleeds in order to assess The short-term adverse prognosis of patients with bleeding
the safety of a new antithrombotic therapy. Especially prior to events compared with patients without such events may stem
regulatory approval, counting major bleeds will allow to detect not only from the critical location of blood loss (intra-cranial, peri-
a signal for enhanced bleeding risk early during the course of cardial, and haemothorax) or the development of haemorrhagic
clinical development of a new antithrombotic regime. On the shock, but also from the negative impact of transfusions67 and
other hand, many of the events that are counted as major from the frequent discontinuation of antithrombotic therapy,
bleeds in large trials are clinically less significant: in addition to with the ensuing enhanced risk of thrombo-embolic events.68
life-threatening bleeds, events that cause permanent organ Additionally, in the short term, reduced tissue oxygenation
damage, and bleeds requiring acute intervention or operation through declining haemoglobin concentrations, increased cardiac
to stabilize the patient, major bleeds also comprise asymptomatic work, haemodynamic compromise, and activation of sympathetic,
haemoglobin drop of 2 or 3 g/dL combined with a small access vasoconstrictive and prothrombotic mechanisms may all concur
site or nasal bleed. Furthermore, modern management of gastro- to produce adverse outcomes (Table 3).
intestinal bleeds has helped to contain long-term consequences Importantly, both the short- and long-term prognoses of patients
of such bleeds for patients. For decision making in clinical prac- with bleeding events (even of those who develop minor bleeding
tice, it may therefore be helpful to distinguish major bleeding events)69 may relate to an unfavourable cluster of baseline charac-
events into clinically relevant major bleeds and clinically less rel- teristics, typical of ‘high-risk’ patients; indeed, patients with bleeding
evant major bleeds. The former would include life-threatening events are older and with more co-morbidities (e.g. renal failure,
events, symptomatic intra-cerebral bleeds and other bleeding hypertension, history of prior bleed, or stroke) compared with non-
events resulting in permanent organ damage and bleeds that bleeders.69 In fact, the CHADS2 score is a valid indicator not only
require an acute operation to stabilize the patients. Clinically for stroke risk, but also for bleeding risk, and sums the known
728 G.Y.H. Lip et al.

risk (Table 4). Drug compliance and maintenance of a therapeutic


Table 3 Factors by which bleeding may negatively INR range are other considerations.
impact short- and long-term outcome

Short-, mid- and long-term prognosis Age


† Accumulation of risk factors for cardiovascular events in patients Older age, in the majority of studies, has been shown to increase
with bleeding events the risk of major haemorrhage. Elderly patients have a two-fold
Short- and mid-term prognosis increased risk of bleeding36 and the relative risk of intra-cranial
† Critical location, e.g. intra-cranial, peri-cardial, haemothorax haemorrhage (in particular at higher INRs) was 2.5 (95% CI 2.3 –
† Impaired tissue perfusion, e.g. by hypotension, shock, hypoxemia 9.4) in patients .85 years old compared with patients 70– 74
† Withdrawal of antithrombotic agents, with resultant increased risk years old.37 In the AFFIRM trial (Atrial Fibrillation Follow-up Inves-
of ischaemic complications tigation of Rhythm Management), the risk of major bleeding
† Activation of sympathetic, vasoconstrictive, and prothrombotic increased by 5% per year of age.20
mechanisms

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


† Increased cardiac work through increased heart rate and output International normalized ratio range
† Negative impact of transfusions, especially of older blood The most important risk factor for haemorrhage is the intensity of
† Prolonged hospitalisation and bed rest, with increased risk of venous the anticoagulant effect (Table 4).38 Studies indicate that with a
thrombo-embolism
target INR of .3.0 the incidence of major bleeding is twofold
greater compared to those with a target INR of 2.0–3.0, at least in
some patient groups.72,73 In studies of patients with prosthetic
heart valves, a lower INR target range resulted in a lower frequency
of major bleeding and intra-cranial haemorrhage with a similar antith-
Table 4 Factors affecting bleeding risk when using oral
rombotic efficacy.74 One retrospective analysis of outpatients using
anticoagulant therapy
warfarin who presented with intra-cranial haemorrhage demon-
Intensity of anticoagulation strated that the risk of this complication doubled for each 1 unit
Management modality increment of the INR.75 Not only the target INR but also the
† Usual care vs. dedicated anticoagulation clinic or increased actual individual INR is strongly associated with the risk of bleeding.76
monitoring frequency or self management Time spent in the therapeutic range (TTR) is also a marker of
Patient characteristics risk. Among individuals randomized to warfarin in the SPORTIF
† Age (Stroke Prevention Using an Oral Direct Thrombin Inhibitor in
† Genetics (may also be assessed by the INR response in the initial Atrial Fibrillation) trials, those with TTR ,60% experienced
period of VKA therapy initiation) higher rates of major haemorrhage compared with those with
† Prior stroke TTR .75%, 3.85%/year vs. 1.58%/year, P , 0.01.77 A similar
† History of bleeding trend was found in the ACTIVE W (Atrial Fibrillation Clopidogrel
† Anaemia Trial with Irbesartan for Prevention of Vascular Events).78 It is likely
† Co-morbidity (hypertension, renal insufficiency, liver disease) that this association reflects the increased bleeding risk during
Use of concomitant medication or alcohol times spent with supra-therapeutic INR values.
† Antiplatelet agents It has been clearly shown that structured and well-organized
† NSAIDs management of anticoagulant treatment, e.g. by specialized antic-
† Medication that affects the intensity of anticoagulation oagulation clinics, results in higher proportions of patients in the
† Alcohol abuse therapeutic target range.79 Point of care testing as well as patient
self-management has also been shown to improve TTR compared
with conventional care in older studies, but not in a more recent
risk factors for cardiovascular events. Thus, the occurrence of any large trial.80 Careful INR monitoring by experienced personnel,
bleeding event identifies individuals as belonging to a high-risk e.g. in anticoagulation clinics, results in similar rates of bleeding
subset. Not surprisingly, the baseline features of patients who and thrombosis as self-monitoring.80,81
bleed largely overlap with those of individuals at high risk of
thrombo-embolic events,70 thereby denoting a general condition Genetic factors affecting vitamin K antagonists
of vascular frailty. Intra-cranial and spontaneous bleeds carry metabolism and their antithrombotic effect
worse prognosis than procedure-related or extracranial bleeds.71 Recently, genetic factors have been identified that may affect the risk
The Task Force recognizes that there is a need to better under- of bleeding. Common polymorphisms in the cytochrome P450 2C9
stand the causes of adverse outcomes in patients who bleed during gene were found to be associated with slow metabolism of VKAs
OAC (Table 4). This may be approached by analysing existing data- and (possibly) a higher risk of bleeding.82 Other genetic factors
bases and by prospective ‘real-world’ registries of patients who bleed. that may influence the requirement of VKAs are variants in the
vitamin K epoxide reductase complex subunit 1 gene (VKORC1).83
Established bleeding risk factors Indeed, the combined analysis of VKORC1, CYP2C9 SNPs, and
Intensity of anticoagulation, management modality, and patient age may account for .50% of the individual variability in the war-
characteristics constitute an important determinant of bleeding farin maintenance dosage, and based on this, prediction models of
Bleeding risk assessment and management in atrial fibrillation patients 729

warfarin maintenance dosage taking into account these individual studies. In six pivotal trials that demonstrated the superiority of
parameters have been developed.84 While available data from warfarin over placebo in the prevention of thrombo-embolic com-
trials suggest that patients carrying such variants require a lower plications in patients with AF, 28 787 patients were screened but
daily dose of VKAs to achieve therapeutic INR values, the clinical only 12.6% of these patients were included in the study.94 In a case-
relevance of these genetic polymorphisms is still controversial. control study in 993 patients on VKAs with major bleeding and 993
non-bleeding controls, ,70% of patients using VKAs would have
Co-morbidities including uncontrolled hypertension, been eligible for the clinical trials.94 In the group of patients that
hepatic, and renal insufficiency presented with haemorrhage, the proportion with any exclusion
History of bleeding and anaemia are risk factors for subsequent criteria was considerably greater (40%; 95% CI 37 –43%) than in
bleeding,56,57 being part of various bleeding risk prediction the control group (23%; 95% CI 21 –26%). Bleeding risk increased
models (see later). Also, anaemia is frequent in patients with sharply with the number of exclusion criteria: two vs. none
renal failure, who are at high risk of bleeding complications when increased the risk four-fold (odds ratio, 3.8; 95% CI 2.7 –5.2) and
anticoagulated.85 three or more vs. none increased the risk 15-fold (odds ratio,

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


Prior stroke is a potent risk factor for thrombo-embolic stroke 14.9; 95% CI 4.7 –46). The fact that many patients were not
in AF, but it is also a risk factor for intra-cerebral haemorrhage.56,57 included in the clinical trials may have a major impact on the exter-
Other co-morbidities, such as hypertension, renal, or hepatic insuf- nal validity of these trials, in particular regarding safety.94
ficiency, also significantly increase the risk of bleeding. A systolic Recent trials of stroke prevention in AF have reported rates of
blood pressure of 140 mmHg or greater has been shown to major haemorrhage of 3%/year among patients randomized to
increase the risk of both haemorrhagic and ischaemic stroke warfarin.22,24 Bleeding rates did not differ markedly between
among patients with AF56,57 A case-control study in 1986 patients open and double-blind trials.94 Rates as high as 7% have been
on VKAs showed that renal impairment and hepatic disease each reported in the first year of warfarin therapy among unselected
independently more than doubled the risk of bleeding.86 These patients with AF in routine practice.29 Hence, in patients not fulfill-
associations were confirmed in the AFFIRM study, in which ing the criteria for randomized controlled trials, a more individua-
hepatic or renal disease conferred a two-fold increase in risk lized approach may be necessary to weigh the expected risks and
(hazard ratio, 1.93; 95% CI, 1.27–2.93). benefits of VKA therapy. Furthermore, warfarin-naive patients may
The AFFIRM investigators also found that heart failure and dia- carry a higher risk for severe bleeding events than other patients.
betes increase bleeding risk, with hazard ratios of 1.43 and 1.44, Taken together, clear risk factors for bleeding in patients using
respectively.20 However, diabetes has been a less consistent risk VKAs can be identified. Systematic reviews56,57 have concluded
factor for bleeding in other overviews.56,57 that the following patient characteristics had supporting evidence
for being risk factors for anticoagulation-related bleeding compli-
Concomitant medications cations in AF patients: advanced age, uncontrolled hypertension,
Another critically important determinant of bleeding risk is the use history of myocardial infarction or ischaemic heart disease, cer-
of concomitant medications, especially antiplatelet drugs. Two ebrovascular disease, anaemia or a history of bleeding, and the
meta-analyses, comprising 6 trials with a total of 3874 patients concomitant use of other drugs such as antiplatelet agents. The
and 10 trials with a total of 5938 patients, found a relative risk presence of diabetes mellitus, controlled hypertension, and
of major bleeding when VKAs were combined with aspirin of 2.4 gender were not identified as significant risk factors.
(95% CI 1.2 –4.8) and 2.5 (95% CI 1.7– 3.7), respectively.87,88 Appreciation of bleeding risk factors will help to inform decision
A population-based case-control study confirmed the high risk of making and will also identify higher risk patients for whom manage-
upper gastro-intestinal bleeding in patients using VKAs in combi- ment strategies to mitigate bleeding risk should be implemented.
nation with aspirin and/or clopidogrel.89,90
Non-steroidal anti-inflammatory drugs (NSAIDs) and alcohol
abuse are also associated with an enhanced risk of gastro-intestinal Bleeding risk stratification and current
bleeding. The combined use of VKAs and NSAIDs may result in an published bleeding risk schema
11-fold higher risk of hospitalization for gastro-intestinal bleeding as Several distinctive clinical prediction rules have been proposed for
compared with the general population.90 – 92 This risk is not signifi- the assessment of the individual risk for bleeding during OAC in
cantly lower when using selective inhibitors of cyclo-oxygenase 2.91,92 patients with AF, based on a combination of treatment- and
Clearly, frailty is an important consideration and biological age person-associated factors.56,57 These prediction rules may help
rather than calendar age is perhaps a better marker of bleeding physicians stratify patients into categories of increasing risk of
risk. Falls may be overstated as a risk factor for bleeding, and bleeding so as to evaluate the individual risk/benefit ratio of an
based on a decision analysis model, a patient with AF would oral anticoagulant, either prior to starting or during treatment.
need to fall 295 times per year for the benefits of stroke preven- The characteristics of the available clinical prediction rules specifi-
tion with warfarin to be outweighed by the risk of intra-cranial cally addressing AF patients are reported in Table 5.
haemorrhage.93 The modified Outpatient Bleeding Risk Index (mOBRI)28 has
been prospectively derived and validated28,48 in patients with
Implications for clinical practice different indications for OAC, including AF patients.51 Another
The assessment of the absolute risk of bleeding in an individual strength of the mOBRI schema was the blinded outcome assess-
patient may be difficult when using data from published clinical ment of bleeding.28 In addition, the settings of the anticoagulation
730
Table 5 Published bleeding risk prediction schemas for patients with atrial fibrillation

Schema, acronym, Reference Population Designa, n b, mean Definition of major Calculation of risk score Bleeding risk classification Major bleeding events by Validated in
author (SD) age, % male; indication for bleeding event n (%) patients in each risk risk category in validation other cohorts
anticoagulationc; length of adjudication category cohort, n (%)
follow-upd
.............................................................................................................................................................................................................................................

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


mOBRI Beyth et al. 28 Prospective inception cohorta Overt bleeding that resulted Age ≥65 years, previous stroke, GI bleed Low: 0 Cumulative incidence of Aspinall
1998 in the loss of ≥2.0 units in in last 2 weeks, ≥1 of the following major bleeding (95% CI) at et al.200551
≤7 days, or was otherwise comorbidities (recent MI, haematocrit 3/12/48 months
life threatening ,30%, creatinine .1.5mg/dL, or
diabetes mellitus) with 1 point for
presence of each risk factor and 0 if
absent
Derivation-n ¼ 556, 61 (14); 46.6%b Intermediate: 1–2
High: ≥3
Derivation
Validation—n ¼ 264, 60 (16); 47.3% Blinded event adjudication, Low: 186 (33.5%) Low: 1 (0–4)/3 (0– 8)/3 (0 –8)
unaware of bleeding risk
factors
Derivation—Post-operative cardiac Intermediate: 336 (60.4%) Intermediate: 5 (1–8)/8 (3–
surgery, mitral valve disease, AF, 12)/12 (5–19)
stroke, TIA, PE, DVT, or other
thrombo-embolismc
Validation—VTE and cardiac surgery High: 34 (6.1) High: 6 (0 –17)/30 (0 –62)/53
(11 –97)
4 yearsd Validation
Low: 80 (30.3)
Intermediate: 166 (62.9)
High: 18 (6.8)
HEMORR2HAGES 52 Retrospective analysis of NRAF ICD-9-CM codes for major Hepatic or renal disease, ethanol abuse, Low: 0– 1 Low: 15 (2.1%)
Gage et al. 2006 cohorta bleeds except those malignancy, older (aged .75), reduced
unrelated to platelet count, re-bleeding risk,
antithrombotic therapye uncontrolled hypertension, anaemia,
genetic factors (CYP 2C9 single
nucleotide polymorphisms), excessive
fall risk, previous stroke/TIA, 1 point for
each risk factor present, and 2 points for
previous bleed
Validation—n ¼ 3971, 80.2e; 43%b Intermediate: 2–3
AFc High: ≥ 4 Intermediate: 35 (5.0%)
3138 patient yearsd Among those on warfarin High: 17 (8.8%)
(n ¼ 1604)
Bleeding risk factors for schema Low: 717 (44.7%)
identified by adaptation of 3
existing bleeding risk schema
(mOBRI, SBRPS, plus bleeding risk
factors identified in literature: Beyth
et al.28 Gage et al-Pub Med search)
Intermediate: 694 (43.3%)

G.Y.H. Lip et al.


High: 193 (12.0%)
Bleeding risk assessment and management in atrial fibrillation patients
Shireman et al. 2006 53 Retrospective chart review of NRAF Hospitalisation within 90 days (0.49×age ≥ 70) + (0.32×female Low: ≤1.07 Low: 35 (0.9%)
cohort and Medicare dataa of hospital discharge gender)+ (0.58×remote
following index AF for GI bleed)+(0.62×recent bleed)+
haemorrhage (0.71×alcohol/drug abuse)+
(diagnosis-related group (0.27×diabetes)+ (0.86×anaemia)+
code 174 or 175) or (0.32×antiplatelet) with 1 point for the
intra-cranial haemorrhage presence of each condition and 0 if

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


e
(ICD-9 430 –432) absent
Derivation—n ¼ 19 875, ≥65 years; Intermediate: .1.07 to Intermediate: 48 (2.0%)
47.5%b ,2.19
Validation—n ¼ 6511, ≥65 years; High: ≥2.19 High: 12 (5.4%)
46.9%
AFc Low: 3889 (59.7%)
3 monthsd Intermediate: 2400 (36.9%)
High: 222 (3.4%)
HAS-BLED Pisters 95 Retrospective analysis of Euro Heart Requiring hospitalisation and/ Hypertension (uncontrolled SBP.160 mm Low: 0– 1 Low: 22 (1.1%)
et al. 2010 Survey cohorta or causing Hb ≥2g/L, Hg), Abnormal renal and/or liver
need for blood transfusion function, Stroke, bleeding history, Labile
that was not a INR, elderly (age . 65 years), drugs
e
haemorrhagic stroke (antiplatelets/NSAIDS)/concomitant
alcohol (≥8 units/week), with 1 point
for the presence of each risk factor
(maximum of 9 points)
Derivation—n ¼ 3978b Intermediate: 2 Intermediate: 14 (1.9%)
Validation—n ¼ 3071, 66.8 (12.8); High: ≥3 High: 12 (4.9%)
59% male
AF patientsc Low: 2084 (67.9%)
12 monthsd Intermediate: 744 (24.2%)
Bleeding risk factors drawn from Euro High: 243 (7.9%)
Heart Survey cohort and ‘historic’
bleeding risk factors (OAC, alcohol
use and hypertension)
e
Fang et al. 2010 (in 96 Retrospective analysis of the ATRIA Anaemia, severe renal disease (GFR Low: 0 to 3 Low: 0.8%
abstract form cohorta ,30 mL/min or dialysis dependent),
only) age ≥75 years, previous bleed,
hypertension, with 1 point each for
presence of previous bleed and
hypertension, 2 points for age≥75, and
3 points each for presence of anaemia
and renal disease
9186b,e Moderate: 4 Moderate: 2.6%
AF patientsc High: 5 –10 High: 5.8%
ed

AF, atrial fibrillation; ATRIA, AnTicoagulation and Risk factors In Atrial fibrillation; DVT, deep vein thrombosis; GFR, glomerular filtration rate; GI, gastrointestinal; HAS-BLED, Hypertension, Abnormal renal &/or liver function, Stroke, Bleeding
history, Labile INR, Elderly (age . 65 years), Drugs (antiplatelets/NSAIDS)/concomitant alcohol (≥8 units/week); Hb haemoglobin; HEMORR2HAGES Hepatic or renal disease, Ethanol abuse, Malignancy, Older (aged.75), Reduced platelet
count, Re-bleeding risk, uncontrolled Hypertension, Anaemia, Genetic factors (CYP 2C9 single nucleotide polymorphisms), Excessive fall risk, previous Stroke/TIA; ICD-9-CM, international classification of disease-9th version, clinical
modification; ICU, intensive/critical care stay; INR, international normalised ratio; MI, myocardial infarction; mOBRI, modified Outpatient Bleeding Risk Index; NRAF, National Registry of Atrial Fibrillation; NSAIDs, non-steroidal
anti-inflammatory drugs; OAC, oral anticoagulation; OBRI, Outpatient Bleeding Risk Index; PE, pulmonary embolism; RCT, randomised controlled trial; SBP, systolic blood pressure; TIA, transient ischaemic attack; VTE, venous
thrombo-embolism.
e
Not reported.

731
732 G.Y.H. Lip et al.

control were the primary care physician28 or a pharmacist-run bleeding risk when only antiplatelet therapy (c-statistic 0.91) or
anticoagulation clinic,51 demonstrating derivation and validation in no antithrombotic therapy at all (c-statistic 0.85) were used. In
‘real-life’ AF patients. the SPORTIF III and V cohorts, the HAS-BLED score was a good
All the clinical prediction rules include age as a risk factor for predictor of bleeding events among warfarin-naı̈ve patients at base-
bleeding, but each schema employs a different cut-off. In the line (c-statistic 0.66) and those patients receiving warfarin plus
mOBRI, age ≥ 65 years scores one point. Given the advanced age aspirin (c-statistic 0.60).97 The HAS-BLED score has been incor-
of most AF patients, the majority of the evaluated patients would porated into the 2010 ESC guidelines for the management of
be assigned at least to the intermediate-risk classification (1–2 AF2 and the Canadian guidelines.98
points) using the mOBRI, which in the validation cohort28 was associ- Preliminary data on a bleeding score from the ATRIA study have
ated with an incidence of major bleeding of 5, 8, and 12% after 3, 12, been presented in abstract form.96 This is a weighted score con-
and 48 months of treatment, respectively. In the independent vali- taining elements already contained within the HAS-BLED score,
dation of the mOBRI among elderly AF patients by Aspinall et al.,51 and gives 3 points for anaemia, 3 points for severe renal disease,
the mOBRI discriminated significantly (P , 0.001) between those 2 points for age ≥ 75, and 1 point each for prior bleeding and

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


in the intermediate- and high-risk categories for major bleeding. hypertension; essentially including similar components as the
The remaining prediction schemas have a retrospective design, scores discussed above, but assigning a weight to the various risk
based on the review of data from the National Registry of Atrial factors (thus making it more complicated). The c-statistic for the
Fibrillation,52 the same Registry plus Medicare data,53 the Euro continuous risk score was 0.74, but on collapsing points into a
Heart Survey cohort,95 and the AnTicoagulation and Risk factors three-category risk index, the annual major haemorrhage rate
in Atrial fibrillation (ATRIA) study.96 The retrospective design was 0.8% in the low-risk group (0–3 points), 2.6% in medium
may represent a limitation of the validity of these schemas since risk (4 points), and 5.8% in high risk (5–10 points).
a potential loss of patients with complications in the early phase In summary, the four available published bleeding risk prediction
of treatment cannot be excluded. rules demonstrate wide variation in the proportions of patients
The HEMORR2HAGES (Hepatic or renal disease, Ethanol abuse, determined to be at low, intermediate, and high risk of bleeding,
Malignancy, Older (aged .75), Reduced platelet count, Re-bleeding due to differences in the risk factors comprising each schema.
risk, uncontrolled Hypertension, Anaemia, Genetic factors (CYP Further, the four published schemas do not share a common defi-
2C9 single nucleotide polymorphisms), Excessive fall risk, previous nition of major bleeding and the length of follow-up of the cohorts
Stroke/TIA) score,52 which considers age . 75 years as a condition differs between studies, which affects the event rate (but this can
at risk, includes factors such as CYP2C9 single nucleotide poly- be easily corrected for). Despite the limitations of the available
morphism, which is rarely investigated, and reduced platelet count bleeding risk schema, they do offer a starting point for physicians
and anaemia that are not easily available, whereas important to consider bleeding when initiating and/or continuing long-term
factors such as antiplatelet treatment or other co-medications are OAC in AF patients, and to think about potentially correctable
not included. The rate of patients at high risk of bleeding was risk factors, for example, in the case of the HAS-BLED score, by
12.0%. The cumulative incidence of major bleeding by risk category treating uncontrolled blood pressure, improving anticoagulation
in the validation cohort was 2.1, 5.0, and 8.8% patient-years in the control labile INRs (if on VKAs) or stopping concomitant aspirin
low, intermediate, and high risk categories, respectively. use.
The schema by Shireman et al.53 incorporates eight risk factors Bleeding risk stratification should be considered as an integral
for bleeding, including female gender and an antiplatelet treatment, part of anticoagulation treatment decision making, and this group
as well as age. There was a relatively short (90 days) follow-up recommends the use of the HAS-BLED score, in keeping with
period and the rate of patients at a high risk was very low international guideline recommendations.2,98
(3.4%). This schema requires a complex mathematical calculation
to derive the individual patient bleeding risk score, thereby limiting Patient values and preferences
its applicability. In general, some important factors related to Patients’ beliefs about their health, the medications and healthcare
bleeding risk, such as poor quality anticoagulation control and they receive are important determinants of whether or not they
the presence of NSAIDs as concomitant medication,56,57 have accept recommended treatments and adhere to therapy. Patients
not been included in four30,52,53,96 of the aforementioned often have perceptions about VKAs, including the inconvenience
schemas. Reliable ascertainment of aspirin therapy given its non- of dosing adjustments, the need for daily medication and regular
prescription status is problematic. Moreover, no blinded blood tests to monitor INR levels, reduction/abstinence from
outcome assessment was conducted and no indication of the alcohol, dietary restrictions, the risk of minor and major bleeding,
setting of anticoagulation control was provided in either the and under-appreciation or lack of knowledge regarding the risk of
HEMORR2HAGES52 or Shireman et al.53 studies. stroke that may influence their acceptance of warfarin and their
More recently, a new clinical prediction rule for bleeding risk ability to maintain good INR control.99 – 101 On the other hand,
evaluation in AF patients, HAS-BLED (Hypertension, Abnormal patients may feel protected by taking VKAs. Thus, patients’ prefer-
renal/liver function, Stroke, Bleeding history or pre-disposition, ences and their beliefs about their health are fundamental in deter-
Labile INR, Elderly (.65), Drugs/alcohol concomitantly), has mining whether anticoagulant treatment, particularly with warfarin,
been proposed by Pisters et al.56,57,95 The HAS-BLED score is adopted in the first place and maintained long term. Changes in
demonstrated a good predictive accuracy in the overall cohort health-care policy emphasize the need to achieve, and benefits of,
(c-statistic 0.72) but performed particularly well in predicting patient involvement in the management of their own health102 and
Bleeding risk assessment and management in atrial fibrillation patients 733

incorporation of patients’ preferences for anti-thrombotic therapy achieve this, although this may represent a lack of patient understand-
should be considered in the decision-making process. ing of the disability associated with major bleeding, particularly intra-
To date, 15 studies have examined patient preferences for antith- cranial haemorrhage. However, other studies suggest that the
rombotic therapy in AF patients103 – 113 and in patients at high risk of decision to accept OAC is attenuated by inclusion of information
developing AF,114 – 118 although one study is yet to report its on intra-cranial haemorrhage risk, indicating that some patients
results118 (Table 6). A variety of decision aids, such as audio- place a greater importance on avoiding an event caused by
booklets,108,112,116,117 decision boards105,108,109,114,115,117 (see anti-thrombotic therapy (i.e. major bleed) than a stroke caused by
Figure 1) and interactive videos/computer programs,103,104,106,113,117 choosing not to take such therapy.110,115 Presenting patients with
have been designed to enable patients to participate in the decision- decision aids makes them more likely to be able to make a decision
making process with regard to their antithrombotic therapy, to regarding antithrombotic therapy and improves their knowledge of
ensure that treatment choices are consistent with their personal AF and the need for such therapy.105,108,113,116,117 However, research
preferences, values, and beliefs. These decision aids provide suggests that the use of decision aids results in fewer patients choos-
written, visual, and verbal information on the likelihood of clinically ing to take OAC108 – 110,113,115,117 and that incorporating patient

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


important outcomes, such as stroke and major haemorrhage associ- preferences would lead to fewer patients choosing to take oral
ated with antithrombotic therapy, present the treatment options anticoagulants than the current guidelines would recommend.110,113
(currently warfarin, aspirin, or no antithrombotic therapy), and ask It is hard to draw firm conclusions from the available
patients to indicate their treatment choice. studies as the sample sizes are often small, typically ≤100
Patients appear to trade-off the risks associated with antithrombo- patients,103 – 106,109 – 111,114,115,117 and there is significant heterogen-
tic treatment in order to avoid death.117,119 Overall, these studies eity between the studies (methods employed to elicit preferences,
appear to suggest that patients place greater emphasis on avoidance patients’ education, risks employed; inclusion of AF patients and
of stroke and are willing to accept a higher risk of bleeding to those without AF, etc.). It is important to distinguish between

Table 6 Patients’ preferences for antithrombotic therapy among atrial fibrillation patients and those at high risk of
developing atrial fibrillation

Author, year, Study population. Study design Method of eliciting patient Outcomes
country Number participants, preferences
mean (SD) age, years
...............................................................................................................................................................................
Studies in patients with atrial fibrillation
Gage et al. 1995, n ¼ 57; 70a Cross-sectional, Interviews, computer-based TTO † High utility for daily aspirin (0.998) or
USA103 Markov decision warfarin (0.988)
model † Disutility associated with severe stroke
(0.39) or extra-cranial haemorrhage
(0.76)
Gage et al. 1996, n ¼ 70; 70.1 (7.3) Cross-sectional, Interviews, computer-based TTO † High utility for daily aspirin (1.0) or
USA104 longitudinal warfarin (0.997)
† Disutility associated with
moderate-to-severe stroke (0.07 and
0.0, respectively)
Man Son Hing et al. n ¼ 64; 68.9 (9.0) RCT Interviews, PTOT † 52% willing to take warfarin for
1996, USA105 absolute risk reduction ≤1/100
Gage 1998, USA106 n ¼ 69; 70a Cross-sectional, Interviews, computer-based TTO † Disutility associated with strokeb
Markov decision
model
Sudlow et al. 1998, n ¼ 176; ≥50 yearsa Cross-sectional Questionnaire and interview † 89% willing to take warfarin to prevent
UK107 stroke
Man Son Hing et al. n ¼ 287; control 67, RCT PTOT vs. usual care † Proportion choosing warfarin greater
1999, USA108 intervention 65a in control group
† PTOT  ability to make decision
choice
Howitt and n ¼ 56a Cross-sectional ‘Qualitative’ interview, PTOT † 20 choose not to take warfarin despite
Armstrong knowledge of stroke risk
1999, UK109
Protheroe et al. n ¼ 97; 77 (3.9) Observational, Individualised decision analysis † 61% preferred warfarin based on
2000, UK110 Markov decision individualised stroke risk
model

Continued
734 G.Y.H. Lip et al.

Table 6 Continued

Author, year, Study population. Study design Method of eliciting patient Outcomes
country Number participants, preferences
mean (SD) age, years
...............................................................................................................................................................................
Thomson et al. n ¼ 57; 73a Cross-sectional, Interview, standard gamble † High utility for daily warfarin (0.94)
2000, UK111 Markov decision † Disutility associated with severe stroke
model (0.19)
McAlister et al. n ¼ 434; 72a Cluster randomised Self-administered, PTOT † PTOT  patient ability to choose
2005, USA112 trial ‘appropriate’ antithrombotic therapy
in short-term only
Thomson et al. n ¼ 109; 73 (6) RCT Computerised decision aid vs. † Computerised decision aid led to
2007, UK113 guideline evidence significantly fewer patients choosing

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


warfarin
Studies in patients at high risk of developing AF but without AF
Devereaux et al. n ¼ 61; aged 40– 74 Prospective Interview, PTOT † 74% willing to take warfarin if just one
2001, Canada114 yearsa observational stroke in 100 patients were prevented
over 2 years
† 57% willing to accept 22 extra bleeds
in 100 patients over a 2-year period
on warfarin
† Most patients willing to take aspirin if it
prevented just 1 stroke in 100 patients
over 2 years
Fuller et al. 2004, n ¼ 81; 81a Cross-sectional Qualitative interview and † Avoidance of stroke paramount
UK115 questionnaire, PTOT † .50% would decline warfarin when
presented with stroke risk
information plus increasing ICH risk
† Need for daily tablets, regular blood
tests, and restrictions on alcohol,
reduced number willing to take
warfarin slightly
Man Son Hing et al. n ¼ 198; 71 (7) RCT Qualitative vs. quantitative, PTOT † Patients more likely to choose aspirin
2002, Canada116 † Patients at moderate-stroke risk more
likely to choose warfarin
† No significant difference in treatment
choice between groups
Holbrook et al. n ¼ 98; 73.6 (6.1) RCT Interview, decision board vs. † When treatment names were blinded,
2007, Canada117 decision booklet with 40% chose warfarin, 42% chose
audiotape vs. interactive aspirin and 18% no treatment
computer program † Unblinding of treatment led to fewer
people choosing warfarin or no
treatment
† Most people chose aspirin
Alonso-Coello n≥96; ≥60 years Cross-sectional Interview, PTOT and visual † Ongoing study data not yet available
et al. 2008, analogue scale
Spain118

AF, atrial fibrillation; SD, standard deviation; UK, United Kingdom; USA, United States of America; ICH, intra-cranial haemorrhage; PTOT, probability trade-off technique; RCT,
randomised controlled trial; TTO, time-trade off; , increased; , decreased.
a
Not reported.
b
Based on data from only 14 patients.

studies of patients with103 – 113 and without AF,114 – 118 as prefer- antithrombotic therapy. Patients tend to choose their current
ences may differ markedly between patients with AF who need to therapy over other treatment choices to prevent cognitive disso-
decide about lifelong therapy and those in a hypothetical situation nance (i.e. distress/conflict between preferences and actual treat-
who need to decide, if they had AF, which treatment they would ment choice).109,110,113,115 – 117 Further studies need to elicit
choose. Patients’ previous or current antithrombotic use and their patient preferences for antithrombotic treatment in warfarin-naı̈ve
experiences (satisfaction with the antithrombotic regimen, adverse AF patients, with and without previous stroke, to remove these
events, etc.) may dramatically influence choice, since many studies potential biases. In addition, perceptions of risk can be altered by
enrolled large proportions of people either currently taking warfarin the way in which information is presented—the effect of ‘framing’.
or aspirin103 – 106,108 – 110,112,113 or those with previous experience of Positive framing, i.e. the chances of survival, is more effective in
Bleeding risk assessment and management in atrial fibrillation patients 735

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


Figure 1 Example of a probability trade-off decision aid (reproduced from Man Son Hing et al. JAMA 1999;282:738, with permission. Copy-
right & (1999) American Medical Association. All rights reserved).

persuading someone to take a ‘risky’ option, such as treatment, than provided to the patient by the care provider (physician, nurse,
negative framing (i.e. the chances of death).120 and other healthcare professionals), their level of education and
There is also tremendous disparity in perceptions of the impact understanding of the consequences of such treatment, and their
of warfarin therapy on the lives of AF patients and this may influ- previous experiences of antithrombotic therapy. Stroke is the
ence the acceptance of such therapy, with many physicians tending most feared complication that patients wish to avoid, but bleeding
to underestimate the level of patients’ satisfaction,121 whereas risk with treatment attenuates the proportion of patients willing
most patients report that warfarin did not precipitate any signifi- to take antithrombotic therapy. Patients need clear, simple, and indi-
cant changes in their day-to-day lives other than minor inconve- vidualized information (presented visually, verbally, and in writing)
niences (such as regular blood tests, adjusting warfarin dose, and on their need for antithrombotic therapy and the potential
dietary restrictions) that they are willing to accept. complications.
The novel OACs (direct thrombin inhibitors and factor Xa
inhibitors) that do not appear to require regular monitoring, Special situations with additional bleeding
unlike VKA therapy, with few drug, food, and alcohol interactions, risk considerations
suggest that OAC will be available to a wider range of people in the Periablation
future. It is conceivable that some patients will be more accepting Catheter ablation carries a small but relevant risk of severe bleed-
of new OACs but we do not currently have data on the impact of ing, 0.5% in older series,122,123 associated with vascular access—
such therapy on patients’ values and preferences. often with relatively large diameter sheaths and peri-interventional
In summary, patients’ preferences for antithrombotic treatment anticoagulation, increasing when ablation is performed in the left
are largely influenced by the type and format of information atrium or left ventricle. Interestingly, bleeding events do not
736 G.Y.H. Lip et al.

appear to be related to pre- and peri-procedural antithrombotic The issue of bleeding with anticoagulation and catheter ablation
therapy (aspirin, VKAs, or others). The available data suggest is increased for patients treated with intra-coronary stents and
that bleeding events during or shortly after catheter ablation pro- with antiplatelet agents, such as clopidogrel and aspirin, which
cedures are largely due to mechanical factors such as vascular cannot be antagonized. For the introduction and manoeuvring of
access, transseptal puncture, and the ablation lesions themselves. sheaths and catheters, the risk of peripheral bleeding or compli-
Catheter ablation also carries a risk for thrombotic events due cations when using aspirin and clopidogrel is low. Additional use
to the scars created in the endocardial walls. Furthermore, an of effective UFH adds to the risk of bleeding. There are no relevant
increasing number of patients undergoing catheter ablation are at data in the literature on the specific question of the management of
long-term risk for embolic stroke and therefore require continu- cardiac tamponade when the patient is on aspirin and clopidogrel.
ous OAC, e.g. most patients with AF or atrial flutter. Right-sided If life-threatening bleeding occurs, platelet transfusions may help.
ablation procedures carry a relatively low risk of peri-procedural The risk of perforation in catheter ablation is extremely low
thrombotic events,122,124 and venous access required for such pro- except in the ablation of AF, which carries higher incidences of
cedures appears to be securable without major bleeding risk when tamponade, where tamponade is the cause of approximately half

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


anticoagulation is continued using VKAs, at least in a relatively small of the procedure-related deaths.123 It can be assumed that bleeding
series. Continuation of VKA therapy even demonstrated a trend is more severe and more difficult to be managed when the patient
towards less minor bleeding events.123,125 is on aspirin and clopidogrel.
Catheter ablation for AF combines the difficulties related to Most guidelines recommend continued anticoagulant therapy
bleeding (from transseptal puncture and requirement for anticoagu- for 2–3 months following an AF ablation in all patients regardless
lation in many patients) and stroke risk (left atrial lesions, long of stroke risk factors.2 The optimal duration of this therapy has not
periods with foreign material in the left atrium, and often long endo- been clearly established. Owing to the risk of relapse, the EHRA
cardial lesions in already diseased atria). The stroke and transient and ESC guidelines recommend that anticoagulation should be
ischaemic attack (TIA) rate is 1% in recent surveys, while bleeding continued long term as per the original indication in subjects
events occur at 1% for cardiac tamponade and 1 –2% for access with stroke risk factors.2,128 This is in line with current recommen-
site bleeds.125 – 127 To avoid bleeding complications and to allow dations for AF and with the observation that AF tends to recur in
rapid adaptation of antithrombotic regimens, a consensus document many patients, including late recurrences in patients after AF
from EHRA in 2008 recommended stopping warfarin 4– 5 days ablation.131
before the ablation procedure and bridging with heparin.128
In a large population of 3052 patients with continuation of VKA Peri-devices (implantable cardioverter-defibrillator,
therapy at a therapeutic INR at the time of AF ablation without the pacemakers)
use of heparin [unfractionated heparin (UFH) or low molecular It may be necessary to interrupt oral anticoagulant therapy for
weight heparin (LMWH)] for bridging, 1 patient had a haemorrha- elective implantation or replacement of a pacemaker or an implan-
gic stroke but recovered completely and bleeding complications table cardioverter defibrillator (ICD), although smaller procedures
occurred in 34 (1.11%) patients.129 Major haemorrhagic compli- can often be performed without interrupting anticoagulation. In
cations (tamponade, haematomas requiring intervention, and patients with mechanical prosthetic heart valves, it may be appro-
necessity for transfusion) occurred in 10 (0.33%) patients.129 In a priate to substitute UFH or LMWH to prevent thrombosis.132 In
case-control analysis, no significant difference in terms of vascular patients with AF who do not have mechanical valves, however,
events or bleeding was detected between patients with a thera- based on extrapolation from the annual rate of thrombo-
peutic INR and the control group with normal INR.130 Thus, unin- embolism in patients with non-valvular AF, it was the consensus
terrupted OAC is a potential alternative to strategies that use of the Task Force for the 2010 ESC guidelines that anticoagulation
bridging with heparin or LMWH. In summary, recent data may be interrupted temporarily for procedures that carry a risk of
suggest that continuation of OAC during catheter ablation pro- bleeding, such as ICD or pacemaker implantation, without substi-
cedures for AF may be safe with respect to bleeding events and tuting heparin.2 In high-risk patients (particularly those with prior
may help to prevent peri-procedural strokes. stroke, TIA, or systemic embolism), UFH or LMWH may be admi-
A higher complication rate has been reported in the elderly (.65 nistered. In the 2008 American College of Chest Physicians
years old) than in young patients undergoing electrophysiological (ACCP) guidelines,133 bridging is ‘suggested’ for patients with a
study (2.2 vs. 0.5%) or radiofrequency ablation (6.1 vs. 2.0%).127 CHADS2 score of 3 or 4 (considered to be at moderate risk of
The incidence of vascular complications depends on the type of vas- thrombo-embolism after interruption of antithrombotic therapy)
cular access (arterial, venous, or both), site of vascular access (i.e. and ‘recommended’ for those with a CHADS2 score of 5 or 6
femoral vs. subclavian or jugular), number of introduced catheters, (considered at high risk, i.e. .10% risk per year). It is possible
length of the procedure, patient profile (i.e. obesity and baseline that such operations can in part be performed without interrup-
coagulation parameters), type of anticoagulation used, management tion of anticoagulation, as for vascular procedures.
of catheterization site during the procedure (i.e. flush) and after- The use of LMWH instead of UFH in patients with AF is based on
wards (immediate vs. delayed catheter removal, duration and extrapolation from venous thrombo-embolic disease states and from
strength of compression, and the use of protamine), and operator limited observational studies or trials.134 LMWH has several pharma-
experience. The introduction of two sheaths through one puncture cological advantages over UFH including a longer half-life, more pre-
site probably increases the risk of haematoma, as does the use of dictable bioavailability, and a predictable antithrombotic response
wide-calibre sheaths for several simultaneous catheters. based on body weight, which permits fixed-dose treatment
Bleeding risk assessment and management in atrial fibrillation patients 737

without laboratory monitoring, except under special circumstances additional use of a GPIIb/IIIa inhibitor (GPI; e.g. in bailout situations
such as obesity, renal insufficiency, or pregnancy.135 Treatment in elective patients or in very high-risk ACS patients); left main or
with LMWH is associated with a lower risk of heparin-induced three-vessel disease; older age (e.g. .75 years); female gender;
thrombocytopenia than treatment with UFH. The favourable prop- smoking; chronic kidney disease; and high INR value (.2.6).
erties of LMWH may simplify the treatment of AF in acute situations Some measures have been taken to reduce this increased bleed-
and may shorten the need for hospitalization. However, excess ing risk: radial instead of femoral access was associated with fewer
dosing of LMWH should be avoided in this vulnerable population access site bleeding events in ‘all-comers’.146 The use of femoral
(elderly and with frequent subclinical kidney impairment). closure devices, although believed to reduce bleeding risk when
Haematoma in the region of the generator pocket mainly occurs compared with manual compression, was not associated with
in patients who are taking antiplatelet or anticoagulant medication. reduced bleeding events.147
Haematoma formation after implantation remains rare among Owing to the lack of prospective randomized investigations in
those who are anticoagulated with a rate that may be similar to this field, a group of experts recently published recommen-
that in patients with a normal INR.136,137 Indeed, there are prob- dations,145 which in the majority are level C, i.e. largely based on

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


ably more patients with haematoma on dual-antiplatelet therapy expert opinion (Table 7).
(incidence up to 20%) than on warfarin therapy (5–10%). Our recommendations can be summarized as follows: (i) avoid
Pocket revision for evacuation may be needed in 20 –50% of the use of DES for patients who require triple antithrombotic
patients with this complication.136,138 In patients with non-valvular therapy; (ii) when OAC is given in combination with clopidogrel
AF, post-operative high-dose heparinization or post-operative and/or low-dose aspirin, the intensity must be carefully regulated,
LMWH bridging substantially increases the haematoma rate (10 – with a target INR of 2.0 –2.5;148,149 and (iii) in the case of combined
20 vs. 2 –8%) without reducing the rate of arterial embolism antithrombotic strategies, gastric protection is recommended at
within the first month after implantation.138 – 140 least for the duration of combination therapy.150
In a systematic review of the literature including eight studies on In elective PCI for patients with chronic stable coronary disease,
the peri-operative management of anticoagulation in patients DES, which require a more prolonged antithrombotic combination
having implantation of a pacemaker or ICD, a strategy involving therapy compared with bare metal stents (BMS),151 should be
bridging anticoagulation with therapeutic-dose heparin was associ- avoided or strictly limited to those clinical and/or anatomical situ-
ated with an incidence of pocket haematoma of 12 –20%, while a ations, such as long lesions, small vessels, diabetes, etc., where a
strategy involving peri-operative continuation of a coumarin was significant benefit is expected as compared with BMS. Triple
associated with an incidence of pocket bleeding of 2–7%.141 The therapy (OAC, aspirin, and clopidogrel) should be given for the
incidence of thrombo-embolic events was 0 –1%, irrespective of duration and at the dosages shown in Table 7. After implantation
the peri-operative anticoagulation strategy used.141 In a recent pro- of a BMS, clopidogrel needs to be given in combination with
spective randomized study including patients with high risk of OAC plus aspirin only for 1 month after implantation, but at
thrombo-embolic events in whom 80% had AF, implant of least 3 months after the use of a ‘-limus’ (sirolimus, everolimus,
devices while maintaining OAC was as safe as bridging with and tacrolimus) eluting stent and at least 6 months after a
heparin infusion and allowed a significant reduction of in-hospital paclitaxel-eluting stent. For OAC patients at moderate–high risk
stay.142 When drainage systems are used, device implantation of thrombo-embolism, an uninterrupted anticoagulation strategy
appears to be safe and can be performed without significantly can be followed, and radial access can be used as the first choice
increased risk of clinically relevant haematoma in patients on even during therapeutic anticoagulation (INR 2.0 –3.0).
continued dual-antiplatelet therapy.143 In patients presenting with NSTE-ACS (including unstable angina
Thus, it has been recommended that for such implantations and non-ST elevation myocardial infarction [NSTEMI])
treatment be interrupted pre-operatively and replaced by dual-antiplatelet therapy with aspirin plus clopidogrel (or other
heparin only if needed.2 If that is not feasible and implantation P2Y12 receptor blockers) is recommended for 12 months. Those
must be performed under anticoagulant (whether maintaining with AF and a moderate–high risk of stroke should also receive
OAC or bridging with heparin) and/or antiplatelet therapy (see or continue anticoagulation therapy. The majority of these patients
the section Patients with ACS and/or requiring PCI/stents), the will undergo cardiac catheterization and/or PCI with/without stent-
procedure should be carried out by an experienced operator ing. Again DES should be avoided or be strictly limited to those clini-
who will pay close attention to haemostasis in the area of the gen- cal and/or anatomical situations, such as long lesions, small vessels,
erator pocket.144 or diabetes. Given the risk of bleeding with combination antithrom-
botic therapies, it may be prudent to temporarily stop OAC, and
Acute coronary syndromes and coronary angiography/ administer short-acting antithrombins or GPIs only if the INR is
intervention ≤2. However, in anticoagulated patients at very high risk of
Several factors associated with coronary angiography or PCI bear thrombo-embolism, an uninterrupted strategy of OAC can be fol-
an increased bleeding risk in patients with a need for OAC (for lowed and radial access can be used as the first choice even
a detailed review see145). These include: ‘triple therapy’ using an during therapeutic anticoagulation (INR 2.0–3.0).
oral anticoagulant and dual platelet inhibition, most often aspirin For longer-term management after an acute event, triple therapy
and clopidogrel; factors prolonging the duration of combined (OAC, aspirin, and clopidogrel) should be used in the mid-term
antithrombotic therapy [e.g. use of drug-eluting stents (DES)]; (3– 6 months), or longer in selected patients at low bleeding risk
OAC, when compared with non-anticoagulated patients; the (Table 7).
738 G.Y.H. Lip et al.

Table 7 Recommended antithrombotic strategies following coronary artery stenting in patients with atrial fibrillation at
moderate-to-high thrombo-embolic risk (in whom oral anticoagulation therapy is required)

Haemorrhagic risk Clinical setting Stent implanted Anticoagulation regime


...............................................................................................................................................................................
Low/intermediate risk Elective Bare metal 1 month: triple therapy of VKA (INR 2.0–2.5)+aspirin ≤100 mg/
day+Clopidogrel 75 mg/day
Up to 12th month: combination of VKA (INR 2.0–2.5)+Clopidogrel 75 mg/
dayb (or aspirin ≤100 mg/day)
Lifelong: VKA (INR 2.0–20) alone
Elective Drug eluting 3 (-olimusa group) to 6 (paclitaxel) months: triple therapy of VKA (INR 2.0–
2.5)+aspirin ≤100 mg/day+Clopidogrel 75 mg/day
Up to 12th month: combination of VKA (INR 2.0–2.5)+Clopidogrel 75 mg/
dayb (or aspirin ≤100 mg/day)

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


Lifelong: warfarin (INR 2.0–3.0) alone
ACS Bare metal/drug eluting 6 months: triple therapy of VKA (INR 2.0– 2.5)+aspirin ≤100 mg/
day+Clopidogrel 75 mg/day
Up to 12 th month: combination of VKA (INR 2.0–2.5)+Clopidogrel
75 mg/dayb (or aspirin ≤100 mg/day)
Lifelong: warfarin (INR 2.0–3.0) alone
High Elective Bare metalc 2– 4 weeks: triple therapy of VKA (INR 2.0– 2.5)+aspirin
≤100 mg/day+Clopidogrel 75 mg/day
Lifelong: VKA (INR 2.0–3.0) alone
ACS Bare metalc 4 weeks: triple therapy of VKA (INR 2.0– 2.5)+aspirin≤100 mg/
day+Clopidogrel 75 mg/day
Up to 12th month: combination of VKA (INR 2.0–2.5)+Clopidogrel 75 mg/
dayb (or aspirin ≤100 mg/day)
Lifelong: VKA (INR 2.0–3.0) alone

Gastric protection with a proton pump inhibitor (PPI) should be considered where necessary.
ACS, acute coronary syndrome; AF, atrial fibrillation; INR, international normalized ratio; VKA, vitamin K antagonist.
a
Srolimua everolimus, and tacrolimus.
b
Combination of VKA [INR 2.0 –3.0]+aspirin≤100 mg/day (with PPI, if indicated) may be considered as an alternative.
c
Drug-eluting stents should be avoided as far as possible, but, if used, consideration of more prolonged (3 –6 months) triple antithrombotic therapy is necessary.

Table 8 Resuming oral antiplatelet agents after invasive procedure

Initial therapy Treatment during Expected treatment immediately Practicalities during re-initiation of therapy
invasive procedure after invasive procedure
...............................................................................................................................................................................
Aspirin Aspirin Aspirin Aspirin same dosage
Aspirin None Aspirin Aspirin same dosage
Clopidogrel None Clopidogrel Clopidogrel 75 mg+300 mg
Clopidogrel Aspirin Aspirin Aspirin same dosage
Apirin + clopidogrel Aspirin Aspirin Aspirin same dosage+clopidogrel
75 mg+300 mg (major thrombotic risk)
Aspirin + clopidogrel None Aspirin Aspirin same dosage+clopidogrel
75 mg+300 mg (major thrombotic risk)
Aspirin+prasugrel Aspirin Aspirin Aspirin same dosage+prasugrel same dosage
Aspirin+ticagrelor Aspirin Aspirin Aspirin same dosage+ticagrelor same dosage

In patients with acute STEMI referred for primary PCI, usually GPIs in patients on OAC should be considered only in rare
unfractionated heparin or bivalirudin is used as an anticoagulant bailout situations and should not be part of a routine procedure.
agent in combination with dual-antiplatelet therapy,152 but their Also, potent P2Y12 inhibitors (prasugrel and ticagrelor) should
use, however, should be ideally limited to patients with a known not be used in combination with VKA until more data are available,
INR of ≤2. The use of more effective ADP receptor blockers in unless there is an urgent clinical need. Again, radial access for
this setting (prasugrel, ticagrelor) may further increase peri- primary PCI is probably the best option to avoid procedural bleed-
interventional bleeding rates. Accordingly, the additional use of ing in patients on OAC, depending on operator expertise and
Bleeding risk assessment and management in atrial fibrillation patients 739

preference. The use of DES should be avoided. Long-term treat- Practical approaches in patients taking dual-antiplatelet
ment after primary PCI follows similar recommendations to therapy
those in patients with NSTE-ACS (Table 7). In surgical procedures with high to very high bleeding risk:
(i) stop clopidogrel 5 days before surgery and stay on ASA,
Surgical procedures and bridging therapy unless very high bleeding risk surgery;
Dual-antiplatelet therapy with aspirin and clopidogrel is a well- (ii) if prasugrel is used, therapy should be stopped 7 days before
established strategy to prevent thrombotic complications in surgery based on its prolonged and less unpredictable action
patients with high platelet reactivity following plaque rupture in compared with clopidogrel;
ACS or PCI.151 Current practice guidelines for antiplatelet (iii) the advantage of ticagrelor in patients referred for surgery
therapy advocate a 4 week dual-antiplatelet therapy in elective would be its fast offset of action after stopping intake.160 In
procedures after BMS—PCI and a (6–) 12 months dual-antiplatelet the PLATelet inhibition and clinical Outcomes (PLATO)
therapy after PCI in patients with ACS and/or the use of DES trial, ticagrelor was stopped 3–5 days before CABG and peri-
PCI.151,152

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


operative bleeding rate was broadly similar to that seen with
Withdrawal of oral-antiplatelet drugs may cause a ‘rebound effect’ clopidogrel.47
associated with the clustering of thrombotic events.153 Importantly, (iv) the substitution of combined antiplatelet therapy with LMWH
patients requiring early surgery after coronary artery stenting face an or UFH alone is ineffective;161
up to 30% incidence of in-hospital major adverse cardiac (v) restart clopidogrel (prasugrel and ticagrelor) as soon as poss-
events.154,155 Although oral-antiplatelet therapy is associated with ible with loading dose (if possible ,24 h after operation);
both short- and long-term clinical efficacy, platelet inhibition, (vi) in very high-risk patients, in whom cessation of antiplatelet
especially when combined with anticoagulant therapy, carries a sub- therapy before surgery seems to be dangerous (e.g. shortly
stantial risk of in-hospital and long-term bleeding and may be a par- after stent implantation), it has been suggested that to switch
ticular limitation in patients presenting for cardiac and non-cardiac from clopidogrel 5 days before surgery to a short half-life antipla-
surgery.156 The risk of major bleeding has been demonstrated to telet agent, e.g. the GPIIb/IIIa-inhibitor tirofiban, and stop infusion
be particularly pronounced in ACS patients treated with prasugrel, of these agents 4 h before surgery.162 This strategy has, however,
as compared with clopidogrel.157 Ticagrelor is an oral, reversible not been investigated in prospective randomized trials.
and more potent P2Y antagonist than clopidogrel and has shown
A summary of how to resume oral-antiplatelet drugs after an inva-
better efficacy with a comparable bleeding risk including coronary
sive procedure is summarized in Table 8.
artery bypass graft surgery (CABG)-related bleeding; however,
In surgical procedures with low to moderate bleeding risk:
similar to prasugrel, there was a statistically significant increase in
consider operating on dual-antiplatelet therapy (ASA +
spontaneous severe bleeding rate.47
clopidogrel) if the specific type of surgery would allow.

Patients on dual-antiplatelet therapy


Practical approaches in patients taking oral
Management of patients undergoing dual-antiplatelet therapy who are
anticoagulation
referred for surgical procedures depends on the urgency of the situ-
Tables 9 and 10 give an overview of bridging therapy in relation to
ation and the thrombotic and bleeding risk of the individual
the choice of antithrombotic regimens and related dosage,
patient.156,158 Many surgical procedures are not immediately necess-
whereby the actual risk of developing a thrombotic complication
ary and should be delayed until combination antithrombotic treatment
and the actual bleeding risk determine the respective therapeutic
is no longer necessary.158 If surgery, however, cannot be delayed, most
approach.133,163 There may be evidence that continuation of
surgical procedures can be performed under dual-antiplatelet therapy
OAC with an INR of 2 is better for the prevention of
or at least under acetyl salicylic acid (ASA) alone with an acceptable
thrombo-embolism and bleeding events for PCI—but limited
rate of bleeding; a multidisciplinary approach is required (cardiologist,
data are available concerning surgery while on therapeutic OAC.
anesthesiologist, haematologist, and surgeon) to determine the
patient’s risk and choose the best method of treatment.
The assessment of these patients requires a balance between the Managing bleeding complications
risk of thrombo-embolic events such as stent thrombosis or stroke, Management of bleeding consists of measures to preserve ade-
and bleeding risk.159 – 161 Those at ‘high risk’ for thrombotic events quate circulation, local control (e.g. endoscopic treatment or sur-
include those with any stent ,6 weeks; DES,6–12 months; gical haemostasis), and proper transfusion procedures. If serious
NSTEMI , 6 weeks; and STEMI , 12 months. Those at ‘moderate bleeding occurs in a VKA user it may be necessary to reverse
risk’ include those with a BMS . 6 weeks, a DES . 12 months, and the anticoagulant effect of the agent. When interrupting the admin-
those presenting with an ACS from 6 weeks to 1 year. istration of VKAs important differences in the half-lives of the
various agents (9 h for acenocoumarol, 36–42 h for warfarin,
Patients on triple therapy and 90 h for phenprocoumon, respectively) need to be taken
In patients taking triple therapy, it is even more important to choose into account.164 There is debate over the best management strat-
the right time point of surgery. For high-risk patients, OAC should egy when a patient on VKA bleeds, given the competing concern
be stopped 3–5 days before surgery and replaced by LMWH or for thrombo-embolic risk. Adequacy of haemostasis and duration
UFH when the INR falls below the therapeutic range. of VKA interruption would vary with different patient scenarios.165
740 G.Y.H. Lip et al.

Table 9 A recommended approach to bridging therapy, in relation to risk of thrombo-embolism (adapted from ACCP8
guidelines, with permission133). Thrombo-embolic risk category of patient

Risk Indication for vitamin K antagonist therapy


stratum ..........................................................................................................................................................
Mechanical heart valve Atrial fibrillation VTE
...............................................................................................................................................................................
High Any mitral value prosthesis; older (caged-ball CHADS2, score: 5 or 6; recent Recent VTE (within 3 months); severe
or tilting-disc) aortic valve prosthesis; (within 3 months) stroke or thrombophilia: e.g. deficiency of protein C,
recent stroke or transient ischemic attack transient ischemic attack; protein S or antithrombin, antiphospholipid
(within 6 months) rheumatic valvular heart disease antibodies or multiple abnormalities
Moderate Bileaflet aortic valve prosthesis and one of the CHADS2, score: 3 or 4 VTE within the past 3–12 months: non-severe
following atrial fibrillation, prior stroke or thrombophilic conditions (e.g. heterozygous.
transient ischemic attack, hypertension, Factor V Leiden mutation or heterozygous.

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


diabetes, congestive heart failure or age Factor II mutation); recurrent VTE
.75 years
Active cancer (treated within 6 months or
palliative)
Low Bileaflet aortic valve prosthesis without atrial CHADS2; score. 0 –2 (and no prior Single VTE occurred .12 months ago and no
fibrillation and no other risk factors for stroke or transient ischaemic other risk factors
stroke attack)

Table 10 A recommended approach to bridging therapy, in relation to risk of thrombo-embolism (adapted from
ACCP8 guidelines, with permission133). Bridging anticoagulant therapy

Patient group Bridging recommendation Grade of


recommendation
...............................................................................................................................................................................

Patients with an MHV or NVAF or VTE at Bridging anticoagulation with therapeutic-dose subcutaneous LMWH or Grade 1C
high risk for thrombo-embolism intra-venous UFH over no bridging during temporary interruption of
OAC
Patients with a MHV, NVAF, or VTE at Bridging anticoagulation with therapeutic-dose subcutaneous LMWH, Grade 2C
moderate risk for ‘thrombo-embolism’ therapeutic-dose intra-venous UFH, or low-dose subcutaneous LMWH
over no bridging curing. Temporary interruption of OAC (grade 2C)
Patients with MHV, NFAV, or VTE at low Low-dose subcutaneous LMWH or no bridging over bridging with Grade 2C
risk for ‘thrombo-embolism’ therapeutic-dose subcutaneous LMWH or intra-venous UFH
Patients who are undergoing minor dental Continuing VKAs at the lime of the procedure and co-administering an oral Grade 1E
procedures and are receiving VKAs prohaemostatic agent
Patients who are undergoing cataract Continuing VKAs at the time at the procedure Grade 1C
removal and are receiving VKAs

LMWH, low-molecular-weight heparin; MHV, mechanical heart value; NVAF, non-valvular artial fibrillation; OAC, oral anticoagulation; UFH, unfractionated heparin; VKA, vitamin
K antagonist; VTE, venous thrombo-embolism.

The most straightforward intervention to counteract the effect normalize the INR.3 Intra-muscular injections of vitamin K should
of VKAs is the administration of vitamin K.164 – 167 There is some be avoided in patients who are anticoagulated, and subcutaneous
debate on the need for vitamin K in the management of a administration of vitamin K results in a less predictable
patient with a very high INR but with no signs of bleeding. bioavailability.164,169
A recent randomized controlled trial did not find any difference in When the INR is ,7, a dose range of 2.5– 5 mg vitamin K has
bleeding or other complications in non-bleeding patients with INR been advocated, whereas a dose of 5– 10 mg may be required
values of 4.5–10 who were treated with vitamin K or for correcting higher INRs. Higher doses of vitamin K are equally
placebo.168,169 In patients with clinically significant bleeding, adminis- effective but may lead to VKA resistance for more than a week,
tration of vitamin K is crucial to reverse the anticoagulant effect of which may hamper long-term management.169
VKAs. Vitamin K can be given orally and intravenously (iv), but the In the case of very serious or even life-threatening bleeding,
parenteral route has the advantage of a more rapid onset of the immediate correction of the INR is essential and can be achieved
treatment.167 After the administration of iv vitamin K, the INR will by the administration of vitamin K-dependent coagulation factors.
start to drop within 2 h and will be completely normalized within Theoretically, these factors are present in fresh frozen plasma;
12–16 h.168 After oral administration it will take up to 24 h to however, the amount of plasma that is required for correcting
Bleeding risk assessment and management in atrial fibrillation patients 741

the INR is very large, carries the risk of fluid overload, and will A perspective on newer anticoagulants
probably take hours to administer.170 Therefore, prothrombin At the time of writing of this summary, the direct thrombin inhibitor
complex concentrates (PCCs), most of which contain all vitamin dabigatran is approved for clinical use in the USA and in Canada, and
K-dependent coagulation factors, are more useful, and individua- is close to approval in Europe. The pharmacokinetics of the drug and
lized dosing regimens based on INR at presentation and body its apparent safety compared with warfarin render dabigatran a
weight are more effective.171 – 174 The risk of disseminated intra- potentially attractive therapeutic option in patients in need of antic-
vascular coagulation (DIC) due to traces of activated coagulation oagulation and at risk for bleeding; at the lower dose tested [110 mg
factors in PCCs comes from the older literature and modern twice a day (bid)] the rate of intra-cerebral bleeding events was
PCCs seem not to be associated with precipating precipitating higher in the VKA group in the RE-LY trial than in the dabigatran
DIC.175 However, PCCs do confer a small thrombotic risk, and group. At the higher tested dose (150 mg bid), dabigatran prevented
this needs to be weighed when considering their use.176 ischaemic strokes more effectively than VKAs. It is conceivable that
some of the other new anticoagulants may confer similar benefits,

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


but large trials in AF patients are not available as full publications
at the time of writing of this document. In addition to these long-
Left atrial appendage closure
term benefits, the shorter half-life of direct thrombin or factor Xa
A substantial number of patients eligible for OAC will suffer bleed- antagonists compared with VKAs suggests that the management of
ing complications that may be potentially life threatening. Patients bleeding complications and the antithrombotic regimen during oper-
at high risk of embolic stroke, but with contraindications for ations and invasive procedures could become simpler with those
OAC are in need of an alternative strategy that is not associated substances compared to VKAs. This assumption is supported by
with long-term bleeding risk. This is particularly true for those the orthopaedic experience, e.g. with dabigatran 110 mg bid which
after an intra-cranial haemorrhage. is often initiated 12–24 h after orthopedic surgery.
The high frequency of thrombus formation in the left ventricular
appendage (LAA) of patients with AF and its role as a source of
embolism have led to the hypothesis that resection or obliteration
of the LAA may reduce the risk of stroke. Surgical closure of the
LAA has been practiced, and indeed, current guidelines suggest Consensus statements
obliteration of the LAA during mitral valve surgery.177 Currently,
excision of the LAA at the time of mitral valve surgery is rec- General atrial fibrillation
ommended for reduction of future stroke risk. Exclusion of the populations1 – 3
LAA during coronary artery bypass graft surgery has also been (i) In most patients, thrombo-embolic rates without anticoagula-
proposed, but with suboptimal results.177,178 tion are markedly (five- to eight-fold) higher than bleeding
A reasonable alternative may be the exclusion of the LAA cavity rates. Therefore, most patients with AF—including the
from the circulation, using either surgical or percutaneous majority of patients at high bleeding risk—are in need of
catheter-based procedures. The percutaneous left atrial appendage anticoagulant therapy.
transcatheter occlusion device was the first to be successfully (ii) For AF patients requiring permanent effective anticoagula-
deployed.179 The second device specifically designed for percuta- tion, it is recommended that the 2010 ESC Guidelines for
neous transcatheter LAA exclusion is the WATCHMAN left the management of patients with AF be applied.
atrial appendage system. The WATCHMAN Left Atrial Appendage (iii) The bleeding risk with aspirin should be considered as similar
System for Embolic PROTECTion in Patients With Atrial Fibrilla- to that with VKA, especially in the elderly.
tion (PROTECT AF) study was designed to demonstrate the (iv) Most patients with a high CHA2DS2-VASC score would
safety, efficacy, and non-inferiority of the WATCHMAN device benefit from OAC even if their bleeding risk is high. Only
compared to chronic warfarin therapy in those patients with non- in rare patients with a relatively low stroke risk and an extre-
valvular AF who are eligible for long-term OAC.180 The rate of mely increased risk of bleeding may the withholding of OAC
ischaemic stroke was higher in the intervention group than in be considered.
the control group, whereas haemorrhagic strokes were less fre- (v) An assessment of the (long-term) risk of bleeding in the
quent in the intervention group than in the control group. More- general AF population is recommended.
over, an important risk of serious procedural complications was (vi) In specific AF patient subsets (i.e. post-ablation, post-LAA
observed (e.g., peri-cardial tamponade), perhaps related to a learn- closure, post-percutaneous coronary intervention/acute cor-
ing curve of device implantation. Several new devices, the onary syndrome, etc.), the assessment of bleeding risk is part
AMPLATZER and Coherex WaveCrest, are a novel alternative, of overall management, balancing this risk against the risk of
and initial results are encouraging.181 Major bleeding was higher thrombo-embolic complications.
with warfarin (4.1%) compared with the WATCHMAN device (vii) The HAS-BLED score should be considered as a calculation
(3.5%). to assess bleeding risk, whereby a score of ≥3 indicates
Careful selection of patients and a meticulous technique are ‘high risk’ and some caution and regular review is needed, fol-
required to improve the risk –benefit ratio. Further data are lowing the initiation of antithrombotic therapy, whether with
required in order to recommend the use of LAA closure in our OAC or antiplatelet drugs.
high-risk patients.
742 G.Y.H. Lip et al.

Peri-ablation2,129 (ii) Bleeding risk assessment should be regularly performed, during


(i) Start OAC (e.g. VKA, such as warfarin INR 2.0 –3.0) for at regular review of the patient. Correctable bleeding risk factors
least 4 weeks prior to the ablation procedure. should be managed.
(ii) In many cases, OAC can be continued throughout the abla-
tion procedure. References
1. Kirchhof P, Auricchio A, Bax J, Crijns H, Camm J, Diener HC et al. Outcome
(iii) Where a bridging strategy is planned, stop VKA 2–5 days
parameters for trials in atrial fibrillation: executive summary. Recommendations
before the ablation procedure and bridging therapy with from a consensus conference organized by the German Atrial Fibrillation Com-
heparin (either LMWH or UFH) until the day before the abla- petence NETwork (AFNET) and the European Heart Rhythm Association
(EHRA). Eur Heart J 2007;28:2803 –17.
tion procedure.
2. Camm AJ, Kirchhof P, Lip GY, Schotten U, Savelieva I, Ernst S et al. Guidelines for
(iv) Peri-procedure anticoagulation: after sheath insertion and the management of atrial fibrillation: the Task Force for the Management of
transseptal puncture, administration of a bolus of intravenous Atrial Fibrillation of the European Society of Cardiology (ESC). Europace 2010;
(IV) heparin (bolus dose empirically 5000–10 000 U or 50– 12:1360 –420.
3. Singer DE, Chang Y, Fang MC, Borowsky LH, Pomernacki NK, Udaltsova N et al.

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


100 U/kg) followed by continuous infusion of 1000–1500 U/ The net clinical benefit of warfarin anticoagulation in atrial fibrillation. Ann Intern
h in order to achieve an ACT at least in excess of 300 s Med 2009;151:297 –305.
that is checked every 30–45 min. On the completion of the 4. Fang MC, Go AS, Chang Y, Borowsky LH, Pomernacki NK, Udaltsova N et al.
Warfarin discontinuation after starting warfarin for atrial fibrillation. Circ Cardio-
procedure, IV heparin is discontinued and sheaths removed vasc Qual Outcomes 2010;3:624–31.
when the ACT is subtherapeutic (,160 s) or if high, reversed 5. Ahrens I, Lip GY, Peter K. New oral anticoagulant drugs in cardiovascular
by protamine. IV heparin to be resumed for 12–24 h at a disease. Thromb Haemost 2010;104:49– 60.
6. Roskell NS, Lip GY, Noack H, Clemens A, Plumb JM. Treatments for stroke pre-
maintenance dose of 1000 U/h without a bolus that will main- vention in atrial fibrillation: a network meta-analysis and indirect comparisons
tain activated partical thromboplastin time at 60–80 s or at versus dabigatran etexilate. Thromb Haemost 2010;104:1106 –15.
least twice the baseline level. Oral anticoagulation to be 7. van Walraven C, Hart RG, Connolly S, Austin PC, Mant J, Hobbs FD et al., Effect
of age on stroke prevention therapy in patients with atrial fibrillation: the Atrial
resumed on the day of the procedure. Fibrillation Investigators. Stroke 2009;40:1410 –6.
(v) Replace IV heparin with subcutaneous LMWH after 12 –24 h 8. Gage BF, Waterman AD, Shannon W, Boechler M, Rich MW, Radford MJ. Vali-
and reinitiate OAC. Stop LMWH when the target INR 2–3 is dation of clinical classification schemes for predicting stroke: results from the
National Registry of Atrial Fibrillation. JAMA 2001;285:2864 –70.
reached. 9. Lip GY, Nieuwlaat R, Pisters R, Lane DA, Crijns HJ. Refining clinical risk stratifica-
(vi) Continue therapeutic warfarin for a minimum of 12 weeks tion for predicting stroke and thromboembolism in atrial fibrillation using a
after the ablation procedure. Patients who have a CHA2DS2- novel risk factor-based approach: the Euro Heart Survey on Atrial Fibrillation.
Chest 2010;137:263 –72.
VASC score of ≥2 should continue OAC long term. 10. Lip GY. Anticoagulation therapy and the risk of stroke in patients with atrial
fibrillation al ‘moderate risk’ [CHADS2 score¼1]: simplifying stroke risk assess-
Peri-devices (implantable ment and thromboprophylaxis in real-life clinical practice. Thromb Haemost 2010;
103:683 –5.
cardioverter-defibrillator, 11. Mannucci PM, Levi M. Prevention and treatment of major blood loss. N Engl J
pacemakers)2,145 Med 2007;356:2301 – 11.
12. Lin H, Wolf PA, Kelly-Hayes M, Beiser AS, Kase CS, Benjamin EJ et al. Stroke
(i) Implant of devices maintaining OAC may be as safe as bridging severity in atrial fibrillation: The Framingham study. Stroke 1996;27:1760 –4.
with heparin infusion and should allow a significant reduction 13. Flaherty MLKB, Woo D, Kleindorfer D, Alwell K, Sekar P, Moomaw CJ et al. The
increasing incidence of anticoagulant-associated intracerebral hemorrhage. Neu-
of in-hospital stay. rology 2007;68:116–21.
(ii) In some circumstances, anticoagulant treatment should be 14. Nieuwlaat R, Capucci A, Lip GY, Olsson SB, Prins MH, Nieman FH et al. on
interrupted pre-operatively and be replaced by heparin. behalf of the Euro Heart Survey Investigators. Antithrombotic treatment in
real-life atrial fibrillation patients: a report from the Euro Heart Survey on
(iii) If implantation must be performed while on anticoagulant
Atrial Fibrillation. Eur Heart J 2006;27:3018 –26.
(whether maintaining OAC or bridging with heparin infusion) 15. Mazzaglia G, Filippi A, Alacqua M, Cowell W, Shakespeare A, Mantovani LG et al.
and/or antiplatelet therapy, the procedure should be carried A national survey of the management of atrial fibrillation with antithrombotic
drugs in Italian primary care. Thromb Haemost 2010;103:968–75.
out by an experienced operator who will pay close attention
16. Garcia DA, Lopes RD, Hylek EM. New-onset atrial fibrillation and warfarin
to haemostasis in the area of the generator pocket. initiation: high risk periods and implications for new antithrombotic drugs.
Thromb Haemost 2010;104:1099 – 105.
17. Ezekowitz MD, Wallentin L, Connolly SJ, Parekh A, Chernick MR, Pogue J et al.,
Presentation with ACS and/or requiring the RE-LY Steering Committee and Investigators. Dabigatran and Warfarin in
PCI/stents2,146 Vitamin K antagonist-naive and experienced cohorts with atrial fibrillation. Circu-
lation 2010;122:2246 – 53.
(i) For antithrombotic therapy management in anticoagulated AF 18. Atrial Fibrillation Investigators. Risk factors for stroke and efficacy of antithrom-
patients presenting with an ACS and/or undergoing PCI/stent- botic therapy in atrial fibrillation. Analysis of pooled data from five randomized
ing, the recommendations in the 2010 ESC Guidelines for the controlled trials. Arch Intern Med 1994;154:1449 – 57.
19. SPAF Investigators. Warfarin versus aspirin for prevention of thromboembolism
management of patients with AF or the ESC thrombosis in atrial fibrillation: Stroke Prevention in Atrial Fibrillation II Study. Lancet 1994;
working group consensus document should be applied. 343:687 –91.
20. DiMarco JP, Flaker G, Waldo AL, Corley SD, Greene HL, Safford RE et al.
Factors affecting bleeding risk during anticoagulant therapy in patients with
The management of bleeding atrial fibrillation: observations from the AFFIRM Study. Am Heart J 2005;149:
complications 650 –6.
21. Executive Steering Committee for the SPORTIF III Investigators. Stroke preven-
(i) Appropriate strategies should be implemented both in the long tion with the oral direct thrombin inhibitor ximelagatran compared with war-
term and peri-intervention, in order to prevent bleeding. farin in patients with non-valvular atrial fibrillation. Lancet 2003;362:1691 –8.
Bleeding risk assessment and management in atrial fibrillation patients 743

22. Executive Steering Committee for the SPORTIF V Investigators. Ximelagatran vs 45. Stone GW, White HD, Ohman EM, Bertrand ME, Lincoff AM, McLaurin BT et al.
warfarin for stroke prevention in patients with nonvalvular atrial fibrillation. Acute Catheterization and Urgent Intervention Triage Strategy (ACUITY) trial
JAMA 2005;293:690 –8. investigators,Bivalirudin in patiens with acute coronary syndromes undergoing
23. Active Writing Group on behalf of the Active Investigators. Clopidogrel plus percutaneous coronary intervention: a subgroup analysis from the Acute Cathe-
aspirin versus oral anticoagulation for atrial fibrillation in the Atrial Fibrillation terization and Urgent Intervention Triage strategy (ACUITY) trial. Lancet 2007;
Clopidogrel Trial with Irbesartan for prevention of vascular events. Lancet 369:907 –19.
2006;367:1903 –12. 46. Yusuf S, Mehta SR, Chrolavicius S, Afzal R, Pogue J, Granger CB et al., Oasis-6
24. Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A et al.; Trial Group. Effects of fondaparinux on mortality and reinfarction in patients
RE-LY Steering Committee and Investigators. Dabigatran vs. Warfarin in patients with acute ST-segment elevation myocardial infarction: the OASIS-6 randomized
with atrial fibrillation. N Engl J Med 2009;361:1139 – 51. trial. JAMA 2006;295:1519 –30.
25. O’Riordan M. Off orbit? ROCKET AF: Rivaroxaban noninferior to warfarin, but super- 47. Wallentin L, Becker R, Budaj A, Cannon C, Emanuelsson H, Held C et al., for the
iority analyses at odds. theheart.org. [Clinical Conditions.Arrhythmia/EP.Arrhythmia/ PLATO Investigators. Ticagrelor versus clopidogrel in patients with acute coron-
EP]; 15November 2010. http://www.theheart.org/article/1148785.do (29 ary syndromes. N Engl J Med 2009;361:1045 –57.
December 2010, date last accessed). 48. Wells PS, Forgie MA, Simms M, Greene A, Touchie D, Lewis G et al. The Out-
26. Landefeld CS, Goldman OL. Major bleeding in outpatients treated with warfarin: patient Bleeding Risk Index: validation of a tool for predicting bleeding rates in
incidence and prediction by factors known at the start of outpatient therapy. Am patients treated for deep vein thrombosis and pulmonary embolism. Arch
J Med 1989;87:144 – 52.

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


Intern Med 2003;163:917 –20.
27. Steffensen F, Kristensen K, Ejlersen E, Dahlerup J, Sorensen H. Major haemorrha-
49. Kuijer PMM, Hutten BA, Prins MH et al. Prediction of the risk of bleeding during
gic complications during oral anticoagulant therapy in a Danish population-based
anticoagulant treatment for venous thromboembolism. Arch Intern Med 1999;
cohort. J Intern Med 1997;242:497–503.
159:457 –60.
28. Beyth RJ, Quinn LM, Landefeld CS. Prospective evaluation of an index for pre-
50. Schulman S, Kearon C. Definition of major bleeding in clinical investigations of
dicting the risk of major bleeding in outpatients treated with warfarin. Am J
antihemostatic medicinal products in non-surgical patients. J Thromb Haemost
Med 1998;105:91– 9.
2005;3:692 – 4.
29. Pengo V, Legnani C, Noventa F, Palareti G, ISCOAT Study Group. Italian Study
51. Aspinall SL, De Sanzo BE, Trilli LE, Good CB. Bleeding risk index in an anticoa-
on Complications of Oral Anticoagulant Therapy. Oral anticoagulant therapy in
gulation clinic. Assessment by indication and implications for care. J Gen Intern
patients with nonrheumatic atrial fibrillation and risk of bleed. A multicenter
Med 2005;20:1008 –13.
inception cohort study. Thromb Haemost 2001;85:418–22.
52. Gage BF, Yan Y, Milligan PE, Waterman AD, Culverhouse R, Rich MW et al.
30. Hylek EM, Evans-Molina C, Shea C, Henault LE, Regan S. Major hemorrhage and
tolerability of warfarin in the first year of therapy among elderly patients with Clinical classification schemes for predicting haemorrhage: Results from the
atrial fibrillation. Circulation 2007;115:2689 –96. National Registry of Atrial Fibrillation (NRAF). Am Heart J 2006;151:713 –9.
31. van der Meer FJ, Rosendaal FR, Vandenbroucke JP, Briet E. Bleeding compli- 53. Shireman TI, Mahnken JD, Howard PA, Kresowik TF, Hou Q, Ellerbeck EF.
cations in oral anticoagulant therapy: an analysis of risk factors. Arch Intern Development of a contemporary bleeding risk model for elderly warfarin reci-
Med 1993;153:1557 –62. pients. Chest 2006;130:1390 –6.
32. Fihn SD, McDonell M, Martin D. Risk factors for complications of chronic antic- 54. Ruı́z-Giménez N, Suárez C, González R, Nieto JA, Todolı́ JA, Samperiz AL et al.,
oagulation: a multicenter study. Ann Intern Med 1993;118:511 – 20. RIETE Investigators. Predictive variables for major bleeding events inpatients
33. Go AS, Hylek EM, Chang Y, Phillips KA, Henault LE, Capra AM et al. Anticoagu- presenting with documented acute venous thromboembolism. Findings from
lation therapy for stroke prevention in atrial fibrillation: How well do random- the RIETE Registry. Thromb Haemost 2008;100:26–31.
ized trials translate into clinical practice? JAMA 2003;290:2685 –91. 55. Cutlip DE, Windecker S, Mehran R, Boam A, Cohen DJ, van Es G-A et al., and on
34. Poli D, Antonucci E, Grifoni E, Abbate R, Gensini GF, Prisco D. Bleeding risk behalf of the Academic Research Consortium. Clinical End Points in Coronary
during oral anticoagulation in atrial fibrillation patients older than 80 years. Stent Trials: A case for standardized definitions. Circulation 2007;115:2344 –51.
J Am Coll Cardiol 2009;54:999 –1002. 56. Palareti G, Cosmi B. Bleeding with anticoagulation therapy—Who is at risk, and
35. Rose A, Ozonoff A, Henault LE, Hylek EM. Warfarin for atrial fibrillation in how best to identify such patients. Thromb Haemost 2009;102:268 –78.
community-based practice. J Thromb Haemost 2008;6:1647 –54. 57. Hughes M, Lip GY, Guideline Development Group for the NICE national clinical
36. Hutten BA, Lensing AW, Kraaijenhagen RA, Prins MH. Safety of treatment with guideline for management of atrial fibrillation in primary and secondary care.
oral anticoagulants in the elderly. A systematic review. Drugs Aging 1999;14: Risk factors for anticoagulation-related bleeding complications in patients with
303– 12. atrial fibrillation: a systematic review. QJM 2007;100:599 –607.
37. Fang MC, Chang Y, Hylek EM, Rosand J, Greenberg SM, Go AS et al. Advanced 58. Schulman S, Angerås U, Bergqvist D, Eriksson B, Lassen MR, Fisher W, Subcom-
age, anticoagulation intensity, and risk for intracranial hemorrhage among mittee on Control of Anticoagulation of the Scientific and Standardization Com-
patients taking warfarin for atrial fibrillation. Ann Intern Med 2004;141:745 –52. mittee of the International Society on Thrombosis and Haemostasis. Definition
38. Schulman S, Beyth RJ, Kearon C, Levine MN. Hemorrhagic complications of of major bleeding in clinical investigations of antihemostatic medicinal products
anticoagulant and thrombolytic treatment: American College of Chest Physicians in surgical patients. J Thromb Haemost 2010;8:202 –4.
Evidence-based Clinical Practice Guidelines (8th Edition). Chest 2008;133: 59. Eikelboom JW, Mehta SR, Anand SS, Xie C, Fox KA, Yusuf S. Adverse impact of
257S– 298S. bleeding on prognosis in patients with acute coronary syndromes. Circulation
39. Steger C, Pratter A, Martinek-Bregel M, Avanzini M, Valentin A, Slany J et al. 2006;114:774 – 82.
Stroke patients with atrial fibrillation have a worse prognosis than patients 60. Ndrepepa G, Berger PB, Mehilli J, Seyfarth M, Neumann FJ, Schöming A et al.
without: data from the Austrian Stroke registry. Eur Heart J 2004;25:1734 –40. Periprocedural bleeding and 1-year outcome after percutaneous coronary inter-
40. Steinhubl SR, Kastrati A, Berger PB. Variation in the definitions of bleeding in ventions: appropriateness of including bleeding as a component of a quadruple
clinical trials of patients with acute coronary syndromes and undergoing percu- end point. J Am Coll Cardiol 2008;51:690 –7.
taneous coronary interventions and its impact on the apparent safety of antith- 61. Berkowitz SD, Granger CB, Pieper KS, Lee KL, Gore JM, Simoons M et al. Inci-
rombotic drugs. Am Heart J 2007;154:3 –11.
dence and predictors of bleeding after contemporary thrombolytic therapy for
41. Chesebro JH, Knatterud G, Roberts R, Borer J, Cohen LS, Dalen J et al. Throm-
myocardial infarction. The Global Utilization of Streptokinase and Tissue Plasmi-
bolysis in Myocardial Infarction (TIMI) Trial, Phase I: A comparison between
nogen activator for Occluded coronary arteries (GUSTO) I Investigators. Circula-
intravenous tissue plasminogen activator and intravenous streptokinase. Clinical
tion 1997;95:2508 –16.
findings through hospital discharge. Circulation 1987;76:142 –54.
62. Moscucci M, Fox KA, Cannon CP, Klein W, López-Sendón J, Montalescot G et al.
42. The GUSTO Investigators. An international randomized trial comparing four
Predictors of major bleeding in acute coronary syndromes: the Global Registry
thrombolytic strategies for acute myocardial infarction. N Engl J Med 1993;
of Acute Coronary Events (GRACE). Eur Heart J 2003;24:1815 –23.
329:673 –82.
63. Segev A, Strauss BH, Tan M, Constance C, Langer A, Goodman SG. Canadian
43. Assessment of the Safety and Efficacy of a New Thrombolytic Regimen
Acute Coronary Syndromes Registries Investigators. Predictors and 1-year
(ASSENT)-3 Investigators. Efficacy and safety of tenecteplase in combination
outcome of major bleeding in patients with non-ST-elevation acute coronary
with enoxaparin, abciximab, or unfractionated heparin: the ASSENT-3 random-
syndromes: insights from the Canadian Acute Coronary Syndrome Registries.
ised trial in acute myocardial infarction. Lancet 2001;358:605 –13.
44. Yusuf S, Zhao F, Mehta SR, Chrolavicius S, Tognoni G, Fox KK, Clopidogrel in Am Heart J 2005;150:690 –4.
Unstable Angina to Prevent Recurrent Events Trial Investigators. Effects of clo- 64. Andersen KK, Olsen TS, Dehlendorff C, Kammersgaard LP. Hemorrhagic and
pidogrel in addition to aspirin in patients with acute coronary syndromes ischemic strokes compared: stroke severity, mortality, and risk factors. Stroke
without ST-segment elevation. N Engl J Med 2001;345:494 –502. 2009;40:2068 –72.
744 G.Y.H. Lip et al.

65. Rosand J, Echman MH, Knudsen KA, Singer DE, Greenberg SM. The effect of 89. Hallas J, Dall M, Andries A, Andersen BS, Aalykke C, Hansen JM et al. Use of
warfarin and intensity of anticoagulation on outcome of intracerebral hemor- single and combined antithrombotic therapy and risk of serious upper gastroin-
rhage. Arch Intern Med 2004;164:880 –4. testinal bleeding: population based case-control study. BMJ 2006;333:726.
66. Michel P, Odier C, Rutgers M, Reichhart M, Maeder P, Meuli R et al. The Acute 90. Sørensen R, Hansen ML, Abildstrom SZ, Hvelplund A, Andersson C,
STroke Registry and Analysis of Lausanne (ASTRAL): design and baseline analysis Jørgensen C et al. Risk of bleeding in patients with acute myocardial infarction
of an ischemic stroke registry including acute multimodal imaging. Stroke 2010; treated with different combinations of aspirin, clopidogrel, and vitamin K antag-
41:2491 – 8. onists in Denmark: a retrospective analysis of nationwide registry data. Lancet
67. Rao SV, Jollis JG, Harrington RA, Granger CB, Newby LK, Armstrong PW et al. 2009;374:1967 –74.
Relationship of blood transfusion and clinical outcomes in patients with acute 91. Mellemkjaer L, Blot WJ, Sorensen HT, Thomassen L, McLaughlin JK, Nielsen GL
coronary syndromes. JAMA 2004;292:1555 –62. et al. Upper gastrointestinal bleeding among users of NSAIDs: a population-
68. Arioldi F, Colombo A, Morici N, Latib A, Cosgrave J, Buellesfeld L et al. Circula- based cohort study in Denmark. Br J Clin Pharmacol 2002;53:173 –81.
tion 2007;116:745 – 54. 92. Battistella M, Mamdami MM, Juurlink DN, Rabeneck L, Laupacis A. Risk of upper
69. Kinnaird T, Stabile E, Mintz GS, Lee CW, Canos DA, Gevorkian N et al. Inci- gastrointestinal hemorrhage in warfarin users treated with nonselective NSAIDs
dence, predictors, and prognostic implications of bleeding and blood transfusion or COX-2 inhibitors. Arch Intern Med 2005;165:189 –92.
following percutaneous coronary interventions. Am J Cardiol 2003;92:930 – 5. 93. Man-Son-Hing M, Nichol G, Lau A, Laupacis A. Choosing antithrombotic
70. Collet JP, Montalescot G. Optimizing long-term dual aspirin/clopidogrel therapy therapy for elderly patients with atrial fibrillation who are at risk for falls. Arch
in acute coronary syndromes: when does the risk outweigh the benefit? Int J Intern Med 1999;159:677 – 85.

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


Cardiol 2009;133:8– 17. 94. Levi M, Hovingh K. Bleeding complications in patients on anticoagulants who
71. Goldstein JN, Greenberg SM. Should anticoagulation be resumed after intracer- would have been disqualified for clinical trials. Thromb Haemost 2008;100:
ebral hemorrhage? Cleve Clin J Med. 2010;77:791 –9. 1047 –51.
72. Saour JN, Sieck JO, Mamo LA, Gallus AS. Trial of different intensities of antic- 95. Pisters R, Lane DA, Nieuwlaat R, de Vos CB, Crijns HJGM, Lip GYH. A novel
oagulation in patients with prosthetic heart valves. N Engl J Med 1990;322: user-friendly score (HAS-BLED) to assess one year risk of major bleeding in
428 –32. atrial fibrillation patients: The Euro Heart Survey. Chest 2010;138:1093 –100.
73. Palareti G, Leali N, Coccheri S, Poggi M, Manotti C, D’Angelo A et al. Bleeding 96. Fang M, Go A, Chang Y, Borowsky LH, Pmernacki NK, Udaltsova N et al.
complications of oral anticoagulant treatment: an inception-cohort, prospective Abstract 16443: Development of a new risk stratification scheme to predict
collaborative study (ISCOAT). Italian Study on Complications of Oral Anticoa- warfarin-associated hemorrhage: the AnTicoagulation and Risk Factors in
gulant Therapy. Lancet 1996;348:423 –8. Atrial Fibrillation (ATRIA) Study. Circulation 2010;122:A16443.
74. Vink R, Kraaijenhagen RA, Hutten BA, van den Brink RB, de Mol BA, Buller HR 97. Lip GY, Frison L, Halperin JL, Lane D. Comparative validation of a novel risk
et al. The optimal intensity of vitamin K antagonists in patients with mechanical score for predicting bleeding risk in anticoagulated patients with atrial fibrillation.
heart valves: a meta-analysis. J Am Coll Cardiol 2003;42:2042 –8. The HAS-BLED (Hypertension, Abnormal Renal/Liver Function, Stroke, Bleed-
75. Hylek EM, Singer DE. Risk factors for intracranial hemorrhage in outpatients ing History or Predisposition, Labile INR, Elderly, Drugs/Alcohol Concomitantly)
taking warfarin. Ann Intern Med 1994;120:897 –902. Score. J Am Coll Cardiol 2011;57:173 – 80.
76. Cannegieter SC, Rosendaal FR, Wintzen AR, van der Meer FJ, Vandenbroucke JP, 98. Cairns JA, Connolly S, McMurtry S, Stephenson M, Talajic M, CCS Atrial Fibrilla-
Briet E. Optimal oral anticoagulant therapy in patients with mechanical heart tion Guidelines Committee. Canadian cardiovascular society atrial fibrillation
valves. N Engl J Med 1995;333:11–7. guidelines 2010: prevention of stroke and systemic thromboembolism in atrial
77. White HD, Gruber M, Feyzi J, Kaatz S, Tse HF, Husted S et al. Comparison of fibrillation and flutter. Can J Cardiol. 2011;27:74– 90.
outcomes among patients randomized to warfarin therapy according to anticoa- 99. Lane DA, Ponsford J, Shelley A, Sirpal A, Lip GYH. Patient knowledge and per-
gulant control: results from SPORTIF III and V. Arch Intern Med 2007;167: ceptions of atrial fibrillation and anticoagulant therapy: effects of an educational
239 –45. intervention programme. The West Birmingham Atrial Fibrillation Project. Int J
78. Connolly SJ, Pogue J, Eikelboom J, Flaker G, Commerford P, Franzosi MG et al. Cardiol 2006;110:354 –8.
Benefit of oral anticoagulant over antiplatelet therapy in atrial fibrillation 100. Lip GYH, Agnelli G, Thach AA, Knight E, Rost D, Tangelder MJ. Oral anticoagula-
depends on the quality of international normalized ratio control achieved by tion in atrial fibrillation: a Pan-European Patient Survey. Eur J Intern Med 2007;18:
centers and countries as measured by time in therapeutic range. Circulation 202 –8.
2008;118:2029 – 37. 101. Dantas GC, Thompson BV, Manson JA, Tracy CS, Upshur RE. Patients’ perspec-
79. van Walraven C, Jennings A, Oake N, Fergusson D, Forster AJ. Effect of study tives on taking warfarin: qualitative study in family practice. BMC Fam Pract 2004;
setting on anticoagulation control: a systematic review and metaregression. 5:15.
Chest 2006;129:1155 –66. 102. Department of Health. Patient and Public Involvement in the New NHS. London:
80. Matchar DB, Jacobson A, Dolor R, Edson R, Uyeda L, Phibbs CS et al., THINRS Department of Health; 1999.
Executive Committee and Site Investigators. Effect of home testing of inter- 103. Gage BF, Cardinalli AB, Albers GW, Owens DK. Cost-effectiveness of warfarin
national normalized ratio on clinical events. N Engl J Med 2010;363:1608 – 20. and aspirin for prophylaxis of stroke in patients with nonvalvular atrial fibrilla-
81. Levi M, de Peuter OR, Kamphuisen PW. Management strategies for optimal tion. J Am Med Assoc 1995;274:1839 –45.
control of anticoagulation in patients with atrial fibrillation. Semin Thromb 104. Gage BF, Cardinalli AB, Owens DK. The effect of stroke and stroke prophylaxis
Hemost 2009;35:560 –7. with aspirin or warfarin on quality of life. Arch Intern Med 1996;156:1829 – 36.
82. Higashi MK, Veenstra DL, Kondo LM, Wittkowsky AK, Srinouanprachanh SL, 105. Man Son Hing M, Laupacis A, O’Connor A, Wells G, Lemelin J, Wood W et al.
Farin FM et al. Association between CYP2C9 genetic variants and Warfarin for atrial fibrillation: The patient’s perspective. Arch Intern Med 1996;
anticoagulation-related outcomes during warfarin therapy. JAMA 2002;287: 156:1841 –8.
1690 –8. 106. Gage BF, Cardinalli AB, Owens DK. Cost-effectiveness of preference-based
83. Reitsma PH, van der Heijden JF, Groot AP, Rosendaal FR, Buller HR. A C1173T antithrombotic therapy for patients with non-valvular atrial fibrillation. Stroke
dimorphism in the VKORC1 gene determines coumarin sensitivity and bleeding 1998;29:1083 –91.
risk. PLoS Med 2005;2:e312. 107. Sudlow M, Thomson R, Kenny RA, Rodgers H. A community survey of patients
84. Namazi S, Azarpira N, Hendijani F, Khorshid MB, Vessal G, Mehdipour AR. The with atrial fibrillation: associated disabilities and treatment preferences. Br J Gen
impact of genetic polymorphisms and patient characteristics on warfarin dose Pract 1998;48:1775 –8.
requirements: a cross-sectional study in Iran. Clin Ther 2010;32:1050 –60. 108. Man-Son-Hing M, Laupacis A, O’Connor AM, Biggs J, Drake E, Yetisir E et al., for
85. Lip GYH. Chronic renal disease and stroke in atrial fibrillation: balancing the pre- the Stroke Prevention in Atrial Fibrillation Investigators. A patient decision aid
vention of thromboembolism and bleeding risk. Europace 2011;13:145 –8. regarding antithrombotic therapy for stroke prevention in atrial fibrillation: a
86. Levi M, Hovingh GK, Cannegieter SC, Vermeulen M, Buller HR, Rosendaal FR. randomised controlled trial. J Am Med Assoc 1999;282:737–43.
Bleeding in patients receiving vitamin K antagonists who would have been 109. Howitt A, Armstrong D. Implementing evidence based medicine in general prac-
excluded from trials on which the indication for anticoagulation was based. tice: audit and qualitative study of antithrombotic treatment for atrial fibrillation.
Blood 2008;111:4471 – 6. Br Med J 1999;318:1324 –7.
87. Hart RG, Benavente O, Pearce LA. Increased risk of intracranial hemorrhage 110. Protheroe J, Fahey T, Montgomery AA, Peters TJ. The impact of patients’ pre-
when aspirin is combined with warfarin: a meta-analysis and hypothesis. Cerebro- ferences on the treatment of atrial fibrillation: observational study of patient
vasc Dis 1999;9:215 –7. based decision analysis. Br Med J 2000;320:1380 –4.
88. Rothberg MB, Celestin C, Fiore LD, Lawler E, Cook JR. Warfarin plus aspirin 111. Thomson R, Parkin D, Eccles M, Sudlow M, Robinson A. Decision analysis and
after myocardial infarction or the acute coronary syndrome: meta-analysis guidelines for anticoagulant therapy to prevent stroke in patients with atrial
with estimates of risk and benefit. Ann Intern Med 2005;143:241 –50. fibrillation. Lancet 2000;355:956 –62.
Bleeding risk assessment and management in atrial fibrillation patients 745

112. McAlister FA, Man Son Hing M, Straus SE, Ghali WA, Anderson D, Majumdar SR of Cardiology; ESC Committee for Practice Guidelines. Guidelines on the man-
et al., for the Decision Aid in Atrial Fibrillation (DAAFI) Investigators. Impact of a agement of valvular heart disease. The task force on the management of valvular
patient decision aid on care among patients with nonvalvular atrial fibrillation: a heart disease of the European Society of Cardiology. Eur Heart J 2007;28:
cluster randomized trial. CMAJ 2005;173:496 –501. 230 –68.
113. Thomson RG, Eccles MP, Steen IN, Greenaway J, Stobbart L, Murtagh MJ et al. A 133. Douketis JD, Berger PB, Dunn AS, Jaffer AK, Spyropoulos AC, Becker RC et al.,
patient decision aid to support shared decision-making on antithrombotic treat- American College of Chest Physicians. The perioperative management of antith-
ment of patients with atrial fibrillation: randomised controlled trial. Qual Saf rombotic therapy: American College of Chest Physicians Evidence-based Clinical
Health Care 2007;16:216–23. Practice Guidelines (8th Edition). Chest 2008;133(6 Suppl.):299S–339S.
114. Devereaux PJ, Anderson DR, Gardner MJ, Putnam W, Flowerdew GJ, 134. Stellbrink C, Nixdorff U, Hofmann T, Lehmacher W, Daniel WG, Hanrath P
Brownell BF et al. Differences between perspectives of physicians and patients et al. Safety and efficacy of enoxaparin compared with unfractionated heparin
on anticoagulation in patients with atrial fibrillation; observational study. Br and oral anticoagulants for prevention of thromboembolic complications in car-
Med J 2001;323:1 –7. dioversion of nonvalvular atrial fibrillation: the Anticoagulation in Cardioversion
115. Fuller R, Dudley N, Blacktop J. Avoidance hierarchies and preferences for antic- using Enoxaparin (ACE) trial. Circulation 2004;109:997 – 1003.
oagulation—semi-qualitative analysis of older patients’ views about stroke pre- 135. Hirsh J, Bauer K, Donati MB, Gould M, Samama MM, Weitz JI. Parenteral antic-
vention and the use of warfarin. Age Ageing 2004;33:608 –11. oagulants: American College Of Chest Physicians Evidence-Based Clinical Prac-
116. Man Son Hing M, O’Connor AM, Drake E, Biggs J, Hum V, Laupacis A. The effect tice Guidelines (8th Edition). Chest 2008;133:141S –159S.
of qualitative vs. quantitative presentation of probability estimates on patient 136. Thal S, Moukabary T, Boyella R, Shanmugasundaram M, Pierce MK, Thai H et al.

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


decision-making: a randomised trial. Health Expect 2002;5:246 –55. The relationship between warfarin, aspirin, and clopidogrel continuation in the
117. Holbrook A, Labiris R, Goldsmith CH, Ota K, Harb S, Sebaldt RJ. Influence of peri-procedural period and the incidence of hematoma formation after device
decision aids on patient preferences for anticoagulant therapy: a randomised implantation. Pacing Clin Electrophysiol 2010;33:385 –8.
trial. Can Med Assoc J 2007;176:1583 –7. 137. Giudici MC, Paul DL, Bontu P, Barold SS. Pacemaker and implantable cardiover-
118. Alonso-Coello P, Montori VM, Solà I, Schünemann HJ, Devereaux PJ, Charles C ter defibrillator implantation without reversal of warfarin therapy. Pacing Clin
et al. Values and preferences in oral anticoagulation in patients with atrial fibrilla- Electrophysiol 2004;27:358 – 60.
tion, physicians’ and patients’ perspectives: protocol for a two-phase study. BMC 138. Wiegand UK, LeJeune D, Boguschewski F, Bonnemeier H, Eberhardt F,
Health Serv Res 2008;8:221 –33. Schunkert H et al. Pocket hematoma after pacemaker or implantable cardiover-
119. Fuller R, Dudley N, Blacktop J. Risk communication and older people—under- ter defibrillator surgery: influence of patient morbidity, operation strategy, and
standing of probability and risk information by medical inpatients aged 75 perioperative antiplatelet/anticoagulation therapy. Chest 2004;126:1177 –86.
years and older. Age Ageing 2001;30:473 – 6. 139. Michaud GF, Pelosi F Jr, Noble MD, Knight BP, Morady F, Strickberger SA. A ran-
120. Thrall G, Lane DA, Carroll D, Lip GYH. Quality of life in patients with atrial domized trial comparing heparin initiation 6 h or 24 h after pacemaker or defi-
fibrillation: a systematic review. Am J Med 2006;119:448.e1 –19. brillator implantation. J Am Coll Cardiol 2000;35:1915 –8.
121. Aliot E, Briethardt G, Brugada J, Camm J, Lip GYH, Vardas PE, Wagner M for the 140. Robinson M, Healey JS, Eikelboom J, Schulman S, Morillo CA, Nair GM et al.
Atrial Fibrillation Awareness and Risk Education (AF AWARE) group [compris- Postoperative low-molecular-weight heparin bridging is associated with an
ing the Atrial Fibrillation Association (AFA), the European Heart Rhythm Associ- increase in wound hematoma following surgery for pacemakers and implantable
ation (EHRA), Stroke Alliance for Europe (SAFE), and the World Heart defibrillators. Pacing Clin Electrophysiol 2009;32:378 – 82.
Federation (WHF)]. An international survey of physician and patient understand- 141. Jamula E, Douketis JD, Schulman S. Perioperative anticoagulation in patients
ing, perception, and attitudes to atrial fibrillation and its contribution to cardio- having implantation of a cardiac pacemaker or defibrillator: a systematic
vascular disease morbidity and mortality. Europace 2010;12:626–33. review and practical management guide. J Thromb Haemost 2008;6:1615 – 21.
122. Calkins H, Yong P, Miller JM, Olshansky B, Carlson M, Saul JP et al. Catheter abla- 142. Tolosana JM, Berne P, Mont L, Heras M, Berruezo A, Monteagudo J et al. Prep-
tion of accessory pathways, atrioventricular nodal reentrant tachycardia, and the aration for pacemaker or implantable cardiac defibrillator implants in patients
atrioventricular junction: final results of a prospective, multicenter clinical trial. with high risk of thrombo-embolic events: oral anticoagulation or bridging
The Atakr Multicenter Investigators Group. Circulation 1999;99:262 –70. with intravenous heparin? A prospective randomized trial. Eur Heart J 2009;
123. Finlay M, Sawhney V, Schilling R, Thomas G, Duncan E, Hunter R et al. Uninter- 30:1880 – 4.
rupted warfarin for periprocedural anticoagulation in catheter ablation of typical 143. Dreger H, Grohmann A, Bondke H, Gast B, Baumann G, Melzer C. Is antiar-
atrial flutter: a safe and cost-effective strategy. J Cardiovasc Electrophysiol 2010;21: rhythmia device implantation safe under dual antiplatelet therapy? Pacing Clin
150– 4. Electrophysiol 2010;33:394 – 9.
124. Alvarez-Lopez M, Rodriguez-Font E, Garcia-Alberola A, Spanish Catheter Abla- 144. Vardas PE, Auricchio A, Blanc JJ, Daubert JC, Drexler H, Ector H et al., European
tion Registry. Fifth official report of the Spanish Society of Cardiology Working Society of Cardiology; European Heart Rhythm Association. Guidelines for
Group on Electrophysiology and Arrhythmias (2005). Rev Esp Cardiol 2006;59: cardiac pacing and cardiac resynchronization therapy. The Task Force for
1165–74. Cardiac Pacing and Cardiac Resynchronization Therapy of the European
125. Di Biase L, Burkhardt JD, Mohanty P, Sanchez J, Horton R, Gallinghouse GJ et al. Society of Cardiology. Developed in collaboration with the European Heart
Periprocedural stroke and management of major bleeding complications in Rhythm Association. Europace 2007;9:959 –98.
patients undergoing catheter ablation of atrial fibrillation: the impact of peripro- 145. Lip G, Huber K, Andreotti F, Arnesen H, Airaksinen K, Cuisset T et al. European
cedural therapeutic internatioinal normalized ratio. Circulation 2010;121:2550 – 6. Society of Cardiology Working Group on Thrombosis. Management of antith-
126. Gaita F, Caponi D, Pianelli M, Scaglione M, Toso E, Cesarani F et al. Radiofre- rombotic therapy in atrial fibrillation patients presenting with acute coronary
quency catheter ablation of atrial fibrillation: a cause of siltent thromboembo- syndrome and/or undergoing percutaneous coronary intervention/stenting.
lism? Magnetic resonance imaging assessment of cerebral thromboembolism in Thromb Haemost 2010;103:13–28.
patients undergoing ablation of atrial fibrillation. Circulation 2010;122:1667 –73. 146. Agostoni P, Biondi-Zoccai G, de Benedictis M, Rigattieri S, Turri M, Anselmi M
127. Cappato R, Calkins H, Chen SA, Davies W, Iesaka Y, Kalman J et al. Updated et al. Radial versus femoral approach for percutaneous coronary diagnostic and
worldwide survey on the methods, efficacy, and safety of catheter ablation for interventional procedures; Systematic Overview and Meta-analysis of Random-
human atrial fibrillation. Circ Arrhythm Electrophysiol 2010;3:32 –8. ized Trials. J Am Coll Cardiol 2004;44:349 – 56.
128. Blanc JJ, Almendral J, Brignole M, Fatemi M, Gjesdal K, Gonzalez-Torrecilla E 147. Jolly S, Amlani S, Hamon M, Yusuf S, Mehta S. Radial versus femoral access for
et al. Consensus document on antithrombotic therapy in the setting of electro- coronary angiography or intervention and the impact on major bleeding and
physiological procedures. Europace 2008;10:513–27. ischemic events: a systematic review and meta-analysis of randomized trials.
129. Hussein AA, Martin DO, Saliba W, Patel D, Karim S, Batal O et al. Radiofre- Am Heart J 2009;157:132 –40.
quency ablation of atrial fibrillation under therapeutic international normalized 148. Rossini R, Musumeci G, Lettieri C, Molfese M, Mihalcsik L, Mantovani P et al.
ratio: a safe and efficacious periprocedural anticoagulation strategy. Heart Long-term outcomes in patients undergoing coronary stenting on dual oral anti-
Rhythm 2009;6:1425 –9. platelet treatment requiring oral anticoagulant therapy. Am J Cardiol 2008;102:
130. Schmidt M, Segerson NM, Marschang H, Akoum N, Rittger H, Clifford SM et al. 1618 –23.
Atrial fibrillation ablation in patients with therapeutic international normalized 149. Sarafoff N, Ndrepepa G, Mehilli J, Dörrler K, Schulz S, Iijima R et al. Aspirin and
ratios. Pacing Clin Electrophysiol 2009;32:995 –9. clopidogrel with or without phenprocoumon after drug eluting coronary stent
131. Ouyang F, Tilz R, Chun J, Schmidt B, Wissner E, Zerm T et al. Long-term results placement in patients on chronic oral anticoagulation. J Intern Med 2008;264:
of catheter ablation in paroxysmal atrial fibrillation: lessons from a 5-year 472 –80.
follow-up. Circulation 2010;122:2368 –77. 150. Bhatt D, Scheiman J, Abraham N, Antman E, Chan F, Furberg C et al. ACCF/
132. Vahanian A, Baumgartner H, Bax J, Butchart E, Dion R, Filippatos G et al., Task ACG/AHA 2008 expert consensus document on reducing the gastrointestinal
Force on the Management of Valvular Hearth Disease of the European Society risks of antiplatelet therapy and NSAID use: a report of the American College
746 G.Y.H. Lip et al.

of Cardiology Foundation Task Force on Clinical Expert Consensus Documents. 165. Ananthasubramaniam K, Beattie JN, Rosman HS et al. How safely and for how
J Am Coll Cardiol 2008;52:1502 –17. long can warfarin therapy be withheld in prosthetic heart valve patients hospi-
151. Silber S, Albertsson P, Aviles F, Camici PG, Colombo A, Hamm CW et al. Guide- talized with a major hemorrhage? Chest 2001;119:478 –84.
lines for percutaneous coronary interventions. The Task Force for Percutaneous 166. Hackman DG, Kopp A, Redelmeier DA. Prognostic implications of warfarin ces-
Coronary Interventions of the European Society of Cardiology. Eur Heart J 2005; sation after major trauma. A population-based cohort analysis. Circulation 2005;
26:804 –47. 111:2250 –6.
152. Cuisset T, Valgimigli M, Mudra H, Muller O, Wijns W, Huber K. Rationale and 167. Dentali F, Ageno W, Crowther M. Treatment of coumarin-associated coagulo-
use of antiplatelet and antithrombotic drugs during cardiovascular interventions: pathy: a systematic review and proposed treatment algorithms. J Thromb
May 2010 update. EuroIntervention 2010;6:39–45. Haemost 2006;4:1853 – 63.
153. Ho P, Peterson E, Wang L, Magid D, Fihn S, Larsen G et al. Incidence of death 168. Crowther MA, Ageno W, Garcia D, Wang L, Witt DM, Clark NP et al. Oral
and acute myocardial infarction associated with stopping clopidogrel after acute vitamin K versus placebo to correct excessive anticoagulation in patients receiv-
coronary syndrome. JAMA 2008;299:532 –9. ing warfarin. Ann Intern Med 2009;150:293 – 300.
154. Nuttall G, Brown M, Stombaugh J, Michon P, Hathaway M, Lindeen K et al. Time 169. Crowther MA, Douketis JD, Schnurr T, Steidl L, Mera V, Ultori C et al. Oral
and cardiac risk of surgery after bare-metal stent percutaneous coronary inter- vitamin K lowers the international normalized ratio more rapidly than subcu-
vention. Anesthesiology 2008;109:588 –95. taneous vitamin K in the treatment of warfarin-associated coagulopathy. A ran-
155. Rabbitts J, Nuttall G, Brown M, Hanson A, Oliver W, Holmes D et al. Cardiac domized, controlled trial. Ann Intern Med 2002;20:251 –4.
risk of noncardiac surgery after percutaneous coronary intervention with 170. Lubetsky A, Yonath H, Olchovsky D, Loebstein R, Halkin H, Ezra D. Comparison

Downloaded from https://academic.oup.com/europace/article/13/5/723/469556 by guest on 16 August 2021


drug-eluting stents. Anesthesiology 2008;109:596–604. of oral vs intravenous phytonadione (vitamin K1) in patients with excessive
156. Metzler H, Kozek-Langenecker S, Huber K. Antiplatelet therapy and coronary anticoagulation: a prospective randomized controlled study. Arch Intern Med
stents in perioperative medicine—the two sides of the coin. Best Pract Res Clin 2003;163:2469 –73.
Anaesthesiol 2008;22:81 –94. 171. van Geest-Daalderop JH, Hutten BA, Pequeriaux NC, de
157. Wiviott SD, Braunwald E, McCabe CH, Horvath I, Keltai M, Herrman JP et al. Vries-Goldschmeding HJ, Rakers E, Levi M. Invasive procedures in the outpatient
setting: managing the short-acting acenocoumarol and the long-acting phenpro-
TRITON-TIMI 38 Investigators. Intensive oral antiplatelet therapy for reduction
coumon. Thromb Haemost 2007;98:747 –55.
of ischaemic events including stent thrombosis in patients with acute coronary
172. Aguilar MI, Hart RG, Kase CS, Freeman WD, Hoeben BJ, Garcia RC et al. Treat-
syndromes treated with percutaneous coronary intervention and stenting in
ment of warfarin-associated intracerebral hemorrhage: literature review and
the TRITON-TIMI 38 trial: a subanalysis of a randomised trial. Lancet 2008;19:
expert opinion. Mayo Clin Proc 2007;82:82–92.
1353 –63.
173. van AL, Eijkhout HW, Kamphuis JS, Dam M, Schattenkerk ME, Schouten TJ et al.
158. Fleisher L, Beckman J, Brown K, Calkins H, Chaikof E, Fleischmann K et al. ACC/
Individualized dosing regimen for prothrombin complex concentrate more effec-
AHA 2007 guidelines on perioperative cardiovascular evaluation and care for
tive than standard treatment in the reversal of oral anticoagulant therapy: an
noncardiac surgery: a report of the American College of Cardiology/American
open, prospective randomized controlled trial. Thromb Res 2006;118:313–20.
Heart Association Task Force on Practice Guidelines (Writing Committee to
174. Pabinger I, Brenner B, Kalina U, Knaub S, Nagy A, Ostermann H. Prothrombin
Revise the 2002 Guidelines on Perioperative Cardiovascular Evaluation for Non-
complex concentrate (Beriplex P/N) for emergency anticoagulation reversal: a
cardiac Surgery). Circulation 2007;116:e418 –499.
prospective multinational clinical trial. J Thromb Haemost 2008;6:622 –31.
159. Samama MM, Horellou MH, Achkar A, Conard J; collaboration de F Mauriat.
175. Levi M. Disseminated intravascular coagulation. Crit Care Med 2007;35:2191 –5.
Perioperative use of antithrombotic agents: recommendations of the American
176. Ehrlich HJ, Henzl MJ, Gomperts ED. Safety of factor VIII inhibitor bypass activity
College of Chest Physicians (ACCP) and the French Superior Health Authority
(FEIBA): 10 year compilation of thrombotic adverse events. Haemophilia 2002;8:
(HAS). J Mal Vasc 2010;35:220–34. 83 –90.
160. Gurbel PA, Bliden KP, Butler K, Tantry US, Gesheff T, Wei C et al. Randomized 177. Bonow RO, Carabello BA, Kanu C et al. ACC/AHA 2006 guidelines for the man-
double-blind assessment of the ONSET and OFFSET of the antiplatelet effects of agement of patients with valvular heart disease: a report of the American
ticagrelor versus clopidogrel in patients with stable coronary artery disease: the College of Cardiology/American Heart Association Task Force on Practice
ONSET/OFFSET study. Circulation 2009;120:2577 –85. Guidelines (Writing Committee to Revise the 1998 Guidelines for the Manage-
161. Oscarsson A, Gupta A, Fredrikson M, Järhult J, Nyström M, Pettersson E et al. ment of Patients With Valvular Heart Disease): developed in collaboration with
To continue or discontinue aspirin in the perioperative period: a randomized, the Society of Cardiovascular Anesthesiologists: endorsed by the Society for
controlled clinical trial. Br J Anaesth 2010;104:305 – 12. Cardiovascular Angiography and Interventions and the Society of Thoracic Sur-
162. Grines C, Bonow R, Casey DJ, Gardner T, Lockhart P, Moliterno D et al. Amer- geons. J Am Coll Cardiol 2006;48:e1 –148.
ican Heart Association; American College of Cardiology; Society for Cardiovas- 178. Kanderian AS, Gillinov M, Pettersson GB, Blackstone E, Klein AL. Success of sur-
cular. Prevention of premature discontinuation of dual antiplatelet therapy in gical left atrial appendage closure. Assessment by transesophageal echocardio-
patients with coronary artery stents: a science advisory from the American graphy. J Am Coll Cardiol 2008;52:924 –9.
Heart Association, American College of Cardiology, Society for Cardiovascular 179. Ostermayer SH, Reisman M, Kramer PH, Matthews RV, Gray WA, Block PC
Angiography and Interventions, American College of Surgeons, and American et al. Percutaneous Left Atrial Appendage Transcatheter Occlusion (PLAATO
Dental Association, with representation from the American College of Phys- system) to prevent stroke in high-risk patients with non-rheumatic atrial
icians. J Am Dent Assoc 2007;138:652 –5. fibrillation: results from the international multi-center feasibility trials. J Am
163. Savonitto S, D’Urbano M, Caracciolo M, Barlocco F, Mariani G, Nichelatti M Coll Cardiol 2005;46:9 –14.
et al. Urgent surgery in patients with a recently implanted coronary drug-eluting 180. Holmes DR, Reddy VY, Turi ZG, Doshi SK, Sievert H, Buchbinder M et al.;
stent: a phase II study of ‘bridging’ antiplatelet therapy with tirofiban during tem- PROTECT AF Investigators. Percutaneous closure of the left atrial appendage
porary withdrawal of clopidogrel. Br J Anaesth 2010;104:285–91. versus warfarin therapy for prevention of stroke in patients with atrial fibrilla-
164. Ansell J, Hirsh J, Hylek E, Jacobson A, Crowther M, Palareti G. Pharmacology and tion: a randomised noninferiority trial. Lancet 2009;374:534 –42.
management of the vitamin K antagonists: American College of Chest Physicians 181. Park JW, Bethencourt A, Sievert H, Santoro G, Meier B, Walsh K et al. Left atrial
Evidence-based Clinical Practice Guidelines (8th Edition). Chest 2008;133: appendage closure with amplatzer cardiac plug in atrial fibrillation—Initial Euro-
160S –198S. pean experience. Catheter Cardiovasc Interv 2011;77:700 –6.

You might also like