Skip to main content
Erschienen in: Advances in Therapy 1/2023

Open Access 16.10.2022 | Review

Risk and Management of Bleeding Complications with Direct Oral Anticoagulants in Patients with Atrial Fibrillation and Venous Thromboembolism: a Narrative Review

verfasst von: Stefano Ballestri, Elisa Romagnoli, Dimitriy Arioli, Valeria Coluccio, Alessandra Marrazzo, Afroditi Athanasiou, Maria Di Girolamo, Cinzia Cappi, Marco Marietta, Mariano Capitelli

Erschienen in: Advances in Therapy | Ausgabe 1/2023

Abstract

Atrial fibrillation (AF) and venous thromboembolism (VTE) are highly prevalent conditions with a significant healthcare burden, and represent the main indications for anticoagulation. Direct oral anticoagulants (DOACs) are the first choice treatment of AF/VTE, and have become the most prescribed class of anticoagulants globally, overtaking vitamin K antagonists (VKAs). Compared to VKAs, DOACs have a similar or better efficacy/safety profile, with reduced risk of intracerebral hemorrhage (ICH), while the risk of major bleeding and other bleeding harms may vary depending on the type of DOAC. We have critically reviewed available evidence from randomized controlled trials and observational studies regarding the risk of bleeding complications of DOACs compared to VKAs in patients with AF and VTE. Special patient populations (e.g., elderly, extreme body weights, chronic kidney disease) have specifically been addressed. Management of bleeding complications and possible resumption of anticoagulation, in particular after ICH and gastrointestinal bleeding, are also discussed. Finally, some suggestions are provided to choose the optimal DOAC to minimize adverse events according to individual patient characteristics and bleeding risk.
Key Summary Points
Atrial fibrillation and venous thromboembolism are highly prevalent conditions posing a significant healthcare burden and representing the main indications for anticoagulation.
Compared to vitamin K antagonists, direct oral anticoagulants (DOACs) have a similar or better efficacy/safety profile, with significantly reduced of risk of intracerebral hemorrhage, while the risk of major bleeding and gastrointestinal bleeding may vary considering DOAC type and special populations.
Supportive measures can be used in most patients, while anticoagulant reversal agents are indicated for life-threatening bleeding complications. In most cases, resuming anticoagulation after a bleeding event will provide a net clinical benefit.
Careful assessment of patient characteristics and bleeding risk is key to choosing the most appropriate DOAC to try to minimize the risk of bleeding complications.
Apixaban, followed by edoxaban, shows the best efficacy/safety profile in the majority of patients.

Introduction

Atrial fibrillation (AF), the most common cardiac arrhythmia, shows an escalating age-dependent prevalence [1], and is significantly associated with an increased risk of thromboembolic events, hospitalization, and mortality [2, 3]. Venous thromboembolism (VTE), including deep vein thrombosis (DVT) and pulmonary embolism (PE), is the third most frequent acute cardiovascular syndrome after myocardial infarction and stroke, ranking among the main causes of cardiovascular mortality [4]. Therefore, both AF and VTE carry a high healthcare financial and social burden.
Direct oral anticoagulants (DOACs), the first-line anticoagulant agents for stroke prevention in non-valvular AF (NVAF) and treatment of VTE, compared to vitamin K antagonists (VKAs), show similar or superior efficacy and better safety, significantly reducing the risk of major bleeding (MB), mainly intracerebral hemorrhage (ICH) [35]. DOACs exert their anticoagulant activity through direct inhibition of thrombin (dabigatran) or activated factor X (FXa) (rivaroxaban, apixaban, edoxaban) and do not require routine laboratory monitoring to assess their antithrombotic activity [3]. The recommended doses of DOACs according to different therapeutic indications, including the relevant dose-reduction criteria, are reported in Table 1 [3].
Table 1
Recommended doses of DOACs in NVAF and VTE patients according to phase-3 randomized controlled trials (RCTs)
 
NVAF
VTE
Standard dose
Dose reduction
Standard dose
Dose reduction
Apixaban
5 mg bid
2.5 mg bid if: age ≥ 80 yrs, weight ≤ 60 kg, creatinine ≥ 1.5 mg/dl (at least two); eGFR 15–29 ml/min (single criterion)
10 mg bid for 7 days followed by 5 mg bid
No in acute VTEa
Extended treatment: possible 2.5 mg bid after 3–6 months
Edoxaban
60 mg od
30 mg od if eGFR 15–49 ml/min, weight ≤ 60 kg, concomitant potent P-Gp inhibitor
60 mg od preceded by LMWH for 5 days
30 mg od if dose reduction criteria as for AF satisfied
Rivaroxaban
20 mg od
15 mg od if eGFR 15–49 ml/min
15 mg bid for 21 days followed by 20 mg od
No in acute VTEb
Extended treatment: possible 10 mg od after 3–6 months
Dabigatran
150 mg bid/110 mg bid
110 mg bid if age ≥ 80 yrs, concomitant verapamil, increased bleeding riskc
150 mg bid preceded by LMWH for 5 days
Nod
Adapted from [3]
bid twice a day, od once daily, eGFR estimated glomerular filtration rate, LMWH low molecular weight heparin, P-Gp P-glycoprotein, RCT randomized controlled trial, SmPc summary of product characteristics (from European Medicines Agency), yrs years
aPer SmPc: it should be used with caution in patients eGFR 15–29 ml/min
bPer SmPc: in patients with eGFR 15–49 ml/min reduced dose 15 mg od should be considered only if risk of bleeding outweighs risk for recurrent DVT/PE (based on pharmacokinetics/ pharmacodynamic analyses; not studied in this setting)
cPer SmPc: no prespecified dose reduction criteria in phase-3 RCT
dPer SmPc: possible dose-reduction criteria as for AF (based on pharmacokinetics/pharmacodynamic analyses; not studied in this setting)
VKAs, such as warfarin, are still widely used, playing an important therapeutic role in patients in whom DOACs are contraindicated: e.g., in those with estimated glomerular filtration rate (eGFR) < 15 ml/min, mechanical heart valves, valvular AF (moderate/severe mitral stenosis), VTE with triple-positive antiphospholipid syndrome, or in the presence of major drug interactions [24]. Both DOACs and VKAs are contraindicated in pregnancy and lactation.
All anticoagulants are associated with a risk of bleeding complications, which can occur at different anatomical sites and with variable severity. This comprehensive review aims at critically discussing available evidence regarding the risk of bleeding complications and efficacy of DOACs compared to VKAs in patients with NVAF and VTE, including a focus on special patient populations. Furthermore, practical principles to help the clinician in assessing bleeding risk, managing bleeding complications, and choosing the appropriate anticoagulant based on patient profile are provided. To this end, we searched the PubMed database for publications up to June 2022, using the following MESH terms and their combination: direct oral anticoagulants, apixaban, dabigatran, edoxaban, rivaroxaban, venous thromboembolism, atrial fibrillation, bleeding, major, gastrointestinal, intracerebral, elderly, obesity, chronic kidney disease, liver disease, cancer, and COVID-19. Clinical trials, observational studies, systematic reviews, meta-analyses, editorials, expert consensus, and clinical guidelines were considered if relevant to the issue. This article is based on previously conducted studies and does not contain any new investigations involving human participants or animals performed by any of the authors.

Risk of Bleeding Complications Under Different Oral Anticoagulants

Overview in NVAF Patients

The risks of various bleeding complications reported in pivotal RCTs and real-world studies comparing DOACs to VKAs (warfarin) for the prevention of stroke or systemic embolism (SE) in patients with NVAF are presented in Table 2 [612].
Table 2
Risk of bleeding with DOACs versus Warfarin in RCTs and real-world studies on NVAF stroke prevention
RCTs
 
Dabigatran
Rivaroxaban
Apixaban
Edoxaban
RE-LY [6]
ROCKET-AF [7]
ARISTOTLE [8]
ENGAGE- AF TIMI 48 [9]
110 mg
150 mg
  
30 mg
60 mg
Stroke or SE
=
=
=
=
MB or CRNMB
=
MB
=
=
ICH
MGIB
=
=
Real-world data
 
Dabigatran [10, 11]
Rivaroxaban [10, 11]
Apixaban [10, 11]
Edoxaban [12]a
Stroke or SE
=
=
=
MB
= ↑
ICH
GIB
b
CRNMB clinically relevant non-major bleeding, GIB gastrointestinal bleeding, ICH intracerebral hemorrhage, MB major bleeding, MGIB major gastrointestinal bleeding, NVAF non-valvular atrial fibrillation, SE systemic embolism
Symbols: = , similar; ↓, minor; ↑ higher [hazard ratio (HR) of events with DOACs versus warfarin]
aOnly one study available
bMGIB

Data from RCTs

In the four pivotal RCTs of DOACs versus warfarin in patients with NVAF (RE-LY, ROCKET-AF, ARISTOTLE, ENGAGE AF-TIMI 48) the risk of MB [expressed as relative risk (RR) or hazard ratio (HR)] was significantly lower with dabigatran 110 mg (RR 0.80), apixaban (HR 0.69), and edoxaban 30/60 mg (HR 0.47/0.80) compared to warfarin, while it was similar with dabigatran 150 mg and rivaroxaban. The risk of ICH was significantly lower with all DOACs compared to warfarin (RR dabigatran 110/150 mg: 0.31/0.40; HR rivaroxaban: 0.67; apixaban: 0.42; edoxaban 30 mg: 0.30; edoxaban 60 mg: 0.47). The risk of major gastrointestinal bleeding (MGIB) was higher with dabigatran 150 mg (RR 1.50), rivaroxaban (3.2% versus 2.2%), and edoxaban 60 mg (HR 1.23), while it was lower with edoxaban 30 mg (HR 0.67) and comparable to warfarin with dabigatran 110 mg and apixaban [69].
A network meta-analysis, including 23 RCTs on AF patients confirmed that the risk of ICH was significantly lower with all DOACs compared to warfarin [odds ratio (OR) range 0.31–0.46), while the risk of MB and GIB varied with each DOAC, reflecting the results of the pivotal RCTs [13]. Apixaban 5 mg, dabigatran 110 mg, and edoxaban 30/60 mg were associated with a lower risk of MB compared to warfarin, while rivaroxaban and dabigatran 150 mg had similar risk. Dabigatran 150 mg, rivaroxaban 20 mg, and edoxaban 60 mg were associated with higher risks of GIB, while apixaban 5 mg and dabigatran 110 mg were associated with similar risk. Edoxaban 30 mg was associated with lower risk of GIB but higher risk of ischemic stroke than warfarin. Apixaban 5 mg ranked the best for most outcomes [13].

Real-World Data

A recent pooled analysis of results from multiple population-based cohort studies from Europe and Canada (421,523 NVAF patients) showed a slightly higher risk of MB for rivaroxaban (HR 1.11), whereas a lower risk of MB was observed for apixaban (HR 0.76) and dabigatran (HR 0.85) compared to VKAs [11]. Superior effectiveness (ischemic stroke/SE HR: 0.82) and safety (ICH/GIB HR: 0.58) outcomes of Apixaban versus Rivaroxaban have been reported in a US nationwide commercial health care claims database [14]. Compared to other DOACs or VKAs, there is scant real-world evidence on the efficacy and safety of edoxaban, which is the last licensed DOAC. In the only study available on AF patients from a large German health insurance database (globally enrolling 21,038 patients), edoxaban demonstrated a favorable safety profile with lower risk of MB compared to rivaroxaban (HR 0.74) and VKA (HR 0.47), while no differences in the risk of MB were found between edoxaban and apixaban or dabigatran [12].
A milestone meta-analysis of 28 real-world studies in AF patients treated with DOACs (dabigatran, rivaroxaban, and apixaban) versus VKAs, found that all three DOACs were associated with a significant reduction in ICH risk (HR 0.42, 0.64, and 0.45, respectively) and similar rates of ischemic stroke/SE; apixaban and dabigatran with reduced MB risk (HR 0.83 and 0.72, respectively) and mortality (HR 0.65 and 0.63, respectively); apixaban with reduced risk of GIB (HR 0.63); and dabigatran (HR 1.20) and rivaroxaban (HR 1.24) with increased risk of GIB [10]. Other recent systematic review and meta-analyses found that among three DOACs (apixaban, dabigatran, rivaroxaban), apixaban had the most favorable safety profile, with lower risk of MB. No significant difference was observed in the risk of stroke/SE between DOACs [15, 16].

Overview in VTE Patients

Acute VTE Treatment

The risks of various bleeding complications reported in the pivotal RCTs and real-world studies comparing DOACs to VKAs (warfarin) for acute VTE treatment are presented in Table 3 [1731].
Table 3
Risk of bleeding with DOACs versus Warfarin in RCTs and real-world studies on acute treatment of VTE
RCTs
 
Dabigatran
Rivaroxaban
Apixaban
Edoxaban
RE-COVER (150 mg) [17]
EINSTEIN DVT-PE [1820]
AMPLIFY [21]
Hokusai-VTE [22]
Recurrent VTE
=
=
=
=
MB or CRNMB
=
MB
=
=
ICH
↓*
↓*
↓*
↓*
MGIB
↑*
=*
↓*
=*
Real-world data
 
Dabigatran [2325]
Rivaroxaban [23, 24, 2628]
Apixaban [24, 2831]
Edoxaban
Recurrent VTE
=
=
=↓
CRNMB
=
MB
=
=
ICH
=↓
GIB
=
=
CRNMB clinically relevant non-major bleeding, GIB gastrointestinal bleeding, ICH intracerebral hemorrhage, MB major bleeding, MGIB major gastrointestinal bleeding, NVAF non-valvular atrial fibrillation, SE systemic embolism, VTE venous thromboembolism
Symbols: = similar; ↓ minor; ↑ higher [hazard ratio (HR) of events with DOACs versus warfarin]; – no data
*Only the number of cases/frequency was reported but not HR and/or statistical significance
Data from RCTs
In RCTs of DOACs on patients with VTE (RE-COVER, EINSTEIN-DVT, EINSTEIN-PE, AMPLIFY, Hokusai-VTE), the risk of MB was significantly lower with rivaroxaban (HR 0.54) and apixaban (HR 0.31), while it was similar with dabigatran 150 mg and edoxaban compared to warfarin; cases of ICH were lower with all four DOACs; cases of MGIB were higher with dabigatran 150 mg, lower with apixaban, and similar with rivaroxaban and edoxaban compared to warfarin [1722].
Real-World Data
International registries XALIA and GARFIELD-VTE. have shown similar safety and efficacy profiles of DOACs (mostly rivaroxaban) versus standard anticoagulation for VTE treatment over a 6–12-month follow-up [26, 27, 32]. All-cause mortality was significantly lower with DOACs than with VKAs [26, 27]. Three recent studies using real-world data from large US registries showed that apixaban was associated with a significantly lower risk of MB and CRNMB and a similar risk of recurrent VTE compared to warfarin overall and among elderly patients (≥ 65 years old) over a 6-month follow-up [2931]. The RE-COVERY DVT/PE international observational study found that dabigatran had similar efficacy to VKA, but lower rates of MB/CRNMB over a 12-month follow-up, although the difference was not statistically significant [25]. A nationwide French cohort study on treatment-naïve patients hospitalized for VTE showed that, compared to VKAs, safety and effectiveness of DOACs were superior for apixaban and similar for rivaroxaban over a 6-month follow-up [28].
In summary, evidence from observational studies for DOAC treatment in acute VTE largely reflect findings of pivotal RCTs. In addition, two recent meta-analyses directly comparing different DOACs in patients with acute VTE have shown that apixaban had a significantly lower risk of major and minor bleeding events than rivaroxaban, with equivalent efficacy in prevention of recurrent VTE events [33, 34].

VTE Extended Treatment

The risks of various bleeding complications reported in the pivotal RCTs comparing DOACs with VKA (warfarin) for VTE extended treatment are presented in Table 4 [18, 3537].
Table 4
Risk of bleeding in RCTs of extended VTE treatment with DOACs
 
Dabigatran
Rivaroxaban
Apixaban
RE-MEDY (versus VKA) [35]
RE-SONATE (versus placebo) [35]
EINSTEIN-EXT (versus placebo) [18]
EINSTEIN-CHOICE (versus ASA 100 mg) [36]
AMPLIFY-EXT (versus placebo) [37]
150 mg
150 mg
20 mg
20 mg
10 mg
5 mg
2.5 mg
Recurrent VTE
=
MB or CRNM
=
=
=
=
MB
=
=
=
=
=
GIB
ICH
ASA aspirin, CRNMB clinically relevant non-major bleeding, GIB gastrointestinal bleeding, ICH intracerebral hemorrhage, MB major bleeding, MGIB major gastrointestinal bleeding, NVAF non-valvular atrial fibrillation, SE systemic embolism, VKA vitamin k antagonist, VTE venous thromboembolism
Symbols: = similar; ↓ minor; ↑ higher [hazard ratio (HR) of events with DOACs versus warfarin]; – no data
Dabigatran was as effective as warfarin in the extended treatment of VTE, exhibiting a lower risk of MB/CRNMB compared to warfarin but a higher risk compared to placebo [35]. Rivaroxaban 20 mg was superior to placebo for reducing recurrent VTE events, while MB/CRNMB events were significantly higher with rivaroxaban [18]. The risk of recurrent VTE was significantly lower with rivaroxaban at either therapeutic (20 mg) or thromboprophylactic dose (10 mg) compared to aspirin, without a significant increase in bleeding rates [36]. Apixaban at either therapeutic (5 mg) or thromboprophylactic dose (2.5 mg) reduced the risk of recurrent VTE without increasing the MB rate compared to placebo [37]. Edoxaban has not yet been evaluated in an RCT of extended therapy for secondary prevention of VTE.
A network meta-analysis of 16 RCTs on VKAs, DOACs, or aspirin for secondary prevention of VTE beyond 3 months, globally including over 22,000 patients, showed that all oral anticoagulant regimens, but not aspirin, were associated with a lower risk of recurrent VTE compared to placebo, while only VKAs were associated with a higher risk of MB compared to both placebo/observation and aspirin [38]. Finally, a recent meta-analysis pooling 17,220 patients (from 14 RCTs and 13 prospective cohort studies) receiving extended anticoagulant therapy for a first unprovoked VTE beyond the initial 3 months (for a minimum of six additional months), reported that the long-term risks and outcomes of anticoagulant-related MB were considerable with both VKAs and DOACs [39]. These results seemingly conflict with evidence from RCTs indicating a low risk of bleeding with reduced dose of FXa inhibitors for secondary prevention of VTE [36, 37]. However, it should be noted that a standard dose of DOAC was used in all but one of the studies included in the meta-analysis. Moreover, data were insufficient to estimate incidence of MB beyond 1 year of extended anticoagulation with DOACs.
No real-life data are available yet on the extended treatment of VTE comparing DOACs with warfarin, aspirin, or placebo in patients who have completed 6–12 months of oral anticoagulation.

ICH

ICH has a heterogeneous etiology, including spontaneous and traumatic ICH. Spontaneous ICH related to anticoagulant therapy occurs in 0.5–1.0% of treated patients annually. It is the most feared complication of anticoagulation, resulting in highest disability and mortality rates (50% at 30-day) [40]. Risk factors for ICH include increasing age, hypertension, concomitant antiplatelet drug use, reduced platelet count, cerebral amyloid angiopathy, history of stroke/transient ischemic attack, history of bleeding, and ethnicity (Asian, Latin American, or Black) [41]. Interestingly, two recent meta-analyses focused on the risk of ICH in patients taking DOACs versus VKAs [42, 43]. The first, including 82,404 NVAF patients from 19 RCTs confirmed, in agreement with literature data, an almost 50% reduction in risk of ICH with DOACs compared to VKAs showing that, among the four DOACs, dabigatran 110 mg was associated with the lowest risk of ICH [42]. The second meta-analysis used data from 55 RCTs on 184,839 patients with various clinical conditions (AF, VTE treatment, and prophylaxis). Compared to VKAs, all DOACs reduced the risk of ICH: dabigatran by 60%, apixaban by 57%, edoxaban by 56%, and rivaroxaban by 41%. If only RCTs on secondary stroke prevention in AF were considered, compared to warfarin, DOACs reduced the risk of ICH by 46%, with a risk of ICH similar to aspirin [43].

GIB

GIB poses a relevant health care burden given that it is the most common type of bleeding among patients on oral anticoagulants and is associated with substantial morbidity and mortality (5–15%) [44, 45].
Data from RCTs and real-world studies showing the risks of MGIB and GIB with DOACs use in NVAF and VTE patients have been anticipated in the above sections (Overview in NVAF patients and Overview in VTE patients). Available evidence from several large observational studies confirms that apixaban is associated with the best safety profile for GIB compared to VKAs and other DOACs, especially in elderly patients; conversely, rivaroxaban probably has the worst profile in patients with AF [11, 4651] and VTE [23, 24, 50]. A systematic review and meta-analysis of data from 43 RCTs and 41 real-world studies showed that there was no significant difference in the risk of MGIB bleeding among patients receiving DOAC (1.19%) compared to conventional treatment (0.92%) for various indications; only the use of rivaroxaban was associated with a 39% increase in the risk of MGIB [52].
The GIB risk of dabigatran and edoxaban in AF patients is dose dependent, and that of dabigatran and rivaroxaban is more pronounced in patients aged ≥ 75 years [23, 47, 53]. The risk of GIB associated with any anticoagulant is increased by the concomitant use of antiplatelet agents [44, 54].
In patients under treatment with different DOACs, coadministration of proton pump inhibitors (PPIs) may decrease the risk of upper GIB, [55], but this effect may be relevant only in high-risk patients (i.e., ≥ 75 years, HAS-BLED score ≥ 3, or on concomitant antiplatelet therapy) [56]. A more pronounced protective effect of PPIs on upper GIB risk has been observed for dabigatran [55], probably because PPIs exert a curative effect on esophageal mucosal lesions potentially induced by the drug’s tartaric-acid core.

Risk of Bleeding Complications in Special Populations

Elderly and Frail Patients

Elderly patients with AF carry an elevated risk of both stroke and bleeding. A recent review summarizing the available evidence from five RCTs (ROCKET AF, ENGAGE AF-TIMI 48, RE-LY, ARISTOTLE, J-ROCKET AF) reported that, while all four DOACs demonstrated a similar/better efficacy compared to warfarin, only apixaban and edoxaban significantly reduced MB events in elderly patients with AF. Age was more strictly associated with bleeding than with ischemic events, resulting in a greater net benefit of apixaban and edoxaban over warfarin in elderly compared to younger patients [57]. A meta-analysis of these RCTs conducted in > 75-year old patients showed that, compared to warfarin, apixaban was the only DOAC significantly associated with the reduction all three outcomes of SE, MB, and ICH (by 29%, 36%, and 66%, respectively) [58]. Recent real-world data confirmed a better efficacy and safety of apixaban compared to rivaroxaban among Medicare beneficiaries with AF who were ≥ 65 years old [59].
Frailty, a common geriatric syndrome characterized by weakness and reduced physiologic reserve, is an emerging health concern [60]. A recent meta-analysis of ten studies involving 97,413 patients has shown that AF patients with frailty had a 2.77-fold higher risk of all-cause mortality and a 1.83-fold higher risk of MB [61]. A post-hoc analysis of the ENGAGE AF-TIMI 48 trial (20,867 participants) showed that edoxaban was as effective as warfarin across the frailty spectrum, and was associated with lower rates of bleeding except in those with severe frailty [62]. In a recent Medicare registry-based study, apixaban was associated with lower rates of adverse events across all frailty levels, while rivaroxaban and dabigatran were associated with lower event rates only among non-frail patients [63].
Frailty carries an increased risk of falls. It is therefore important to highlight that two post-hoc subanalyses of phase 3 RCTs ENGAGE-AF and ARISTOTLE showed that efficacy and safety profiles of edoxaban and apixaban were preserved, regardless of risk of falls in elderly patients [64, 65].
In summary, DOACs have been found to be safer and more effective than warfarin for the treatment of NVAF in older and frail patients. Apixaban and edoxaban seem to provide the best combination of efficacy and safety in these patient populations [3, 57, 66].
Overall, data from RCTs on VTE treatment have shown that, for both young and elderly patients, DOACs have similar efficacy and improved safety compared to VKAs [67, 68]. Limited data are available on frail patients with VTE treated with DOACs. Published subanalyses of the DOAC phase 3 RCTs have shown that rivaroxaban and edoxaban offer at least as much benefit to frail patients as they do to those who are not frail [20, 22]. Data from the observational registry RIETE suggest that the use of DOACs may be more effective and safer than standard therapy in VTE fragile patients (defined as age ≥ 75 years and/or eGFR ≤ 50 mL/min and/or body weight ≤ 50 kg) [69]

Extreme Weight Patients

Registration studies of DOACs for NVAF or VTE did not have any body mass index (BMI)/weight restrictions, but data on the efficacy and safety of DOACs in patients with extreme weights are limited.

Severe Obesity

Recent analyses of the four pivotal RCTs on DOACs versus warfarin for NVAF stratified by BMI has demonstrated preserved efficacy with DOACs versus warfarin in obese patients, with similar risk of MB [70]. However, data are limited in patients with severe class III obesity (BMI ≥ 40 kg/m2). Post-hoc analyses of ARISTOTLE and ENGAGE AF-TIMI 48 trials demonstrated that edoxaban and apixaban were as effective and safe as warfarin in patients with BMI ≥ 40 kg/m2 [7173]. Edoxaban and rivaroxaban demonstrated consistent drug levels in severely obese patients [71, 73, 74]. Two recent large observational studies have shown that rivaroxaban was associated with a reduced risk of stroke/SE and similar or reduced risk of MB compared to warfarin in NVAF patients with mild-to-severe obesity [75, 76]. Another large observational study (ARISTOPHANES) found no significant difference in the risk of stroke/SE between DOAC and warfarin in NVAF patients with morbid obesity. Apixaban had a lower risk of MB compared to warfarin, dabigatran, and rivaroxaban. Conversely, compared to warfarin, dabigatran and rivaroxaban were both associated with a similar risk of MB [77].
Available data from RCTs and real-life studies on DOACs for the treatment of VTE suggest that in patients with BMI ≥ 40 kg/m2 or weight ≥ 120 kg, apixaban and rivaroxaban appear to be effective and safe compared to VKAs [74, 7880].
Current guidelines end expert opinions advocate using DOACs with caution in AF or VTE patients with weight ≥ 120 kg/BMI ≥ 40 kg/m2 while, given limited data available in those with weight ≥ 140 kg/BMI ≥ 50 kg/m2, using VKAs or performing trough plasma-level measurements of DOACs may be reasonable in such patients [3, 70, 81].

Low Body Weight

Low body weight may raise plasma concentrations of any DOAC and VKA, thus increasing the risk of bleeding compared to normal-weight patients [3]. Notably, underweight patients may frequently have associated conditions that increase the risk of stroke as well as of bleeding. These conditions include advanced age, frailty, chronic kidney disease (CKD), and cancer. Data from RCTs have shown that both apixaban and edoxaban have consistent efficacy and safety compared to warfarin in underweight patients with AF when compared to the overall study population [7173, 82]. Conversely, data regarding dabigatran and rivaroxaban were less convincing [3]. No specific comparative analyses on the safety and efficacy of DOACs versus warfarin in the treatment of VTE in low body weight patients are currently available [83]. However, no difference in safety and efficacy of apixaban and edoxaban versus warfarin was reported in phase 3 RCTs on a prespecified small subgroup analysis of patients weighing ≤ 60 kg [84]. Edoxaban is the only DOAC requiring a reduced dose based on body weight for the treatment of both VTE and AF [3].
Current guidelines end expert opinions generally suggest using DOACs Apixaban and Edoxaban (following any proper dose-reduction criteria) in patients weighing 60–40 kg with AF or VTE, while using VKAs or performing plasma level measurements of DOACs may be preferred in patients weighing less than 40 kg with AF or VTE [3, 70, 81, 83, 85]. However, no evidence-based recommendations exist regarding (further) dose reduction in cases where trough levels are above the expected range.

Diabetics

Evidence from RCTs suggests that patients with diabetes benefit from DOAC versus VKA therapy to an extent similar to patients without diabetes [86, 87]. A recent meta-analysis of observational studies reported that DOACs had a superior efficacy and safety profile over VKAs in patients with NVAF and diabetes [88]. Interestingly, a recent observational nationwide study has shown that DOACs were associated with a lower risk of diabetes vascular complications and mortality than warfarin in patients with AF and diabetes [89].

CKD Patients

A bidirectional relationship links AF with CKD, which share multiple risk factors. CKD is associated with an increased risk of both ischemic stroke and bleeding, and this complicates the decision of which optimal stroke prevention strategies should be adopted among patients with AF and CKD [90]. CKD is also associated with increased risk of a first episode/recurrent VTE and is a well-recognized risk factor for bleeding [91, 92].
Data from systematic reviews and meta-analyses have shown that, in patients with moderate CKD (eGFR 30–59 mL/min) and AF or VTE, DOACs compared to warfarin have a superior or equivalent safety and efficacy profile [93, 94]. Post-hoc analyses of phase 3 RCTs and observational data revealed that DOACs, in particular dabigatran and rivaroxaban, were associated with reduced loss of renal function compared to warfarin [90, 95].
Patients with severe CKD (eGFR 15–29 mL/min) were generally excluded from pivotal phase 3 RCTs, both in AF and VTE patients. Nevertheless, pharmacokinetic data indicate that a change in DOAC plasma levels of the three factor Xa inhibitors (apixaban, edoxaban, and rivaroxaban) were similar in patients with severe and moderate renal impairment; this finding led to their being approved in patients with severe CKD, a special patient population in which DOACs should, nevertheless, be used with caution [90]. A recent large US observational cohort study showed consistently lower risk of CVD and bleeding events with DOACs compared to warfarin across CKD stages (including end-stage disease) in patients with AF, while the risk was equal in those with VTE [96]. Available data suggest that apixaban may have a similar or better efficacy and safety profile than warfarin in AF or VTE patients with severe to end-stage CKD [94, 97, 98].
In summary, the limited available data suggest that DOACs may have a favorable safety and efficacy profile in moderate-to-severe CKD.

Chronic Liver Disease Patients

Anticoagulant therapy is challenging in cirrhotic patients, who exhibit an unstable hemostatic balance fluctuating between thrombosis and bleeding [99].
Patients with significant active liver disease and cirrhosis have been excluded from all pivotal RCTs on DOACs both in AF and VTE [99, 100]. Nevertheless, accumulating real-world data from relatively small-sized cohort studies to large retrospective longitudinal population studies suggest that DOACs are at least as effective as, and may be safer than VKAs in patients with advanced liver disease [99, 101]. A recent meta-analysis has shown that, similar to what has been observed among non-cirrhotic patients, in cirrhotic patients treated with DOACs owing to various indications, DOACs have a similar efficacy and lower risk of MB/ICH compared to VKAs [102]. The risk of GIB was variably reported as being either reduced or equivalent compared to warfarin [102, 103].
All four DOACs are recommended in patients with cirrhosis Child–Pugh A, while they are contraindicated in those patients with liver-related coagulopathy and clinically relevant bleeding risk, including those with cirrhosis Child–Pugh C class. Recommendations regarding patients with cirrhosis Child–Pugh B vary: rivaroxaban is contraindicated, while dabigatran, apixaban, and edoxaban may be used with caution [3].

Corona Virus Disease-19

An increased incidence of VTE, attributed to micro- and macrovascular thrombosis and systemic coagulopathy, has been observed among hospitalized patients with coronavirus disease-19 (COVID-19). Critically ill COVID-19 patients may also be at increased bleeding risk, which can result from platelet dysfunction, thrombocytopenia, organ dysfunction, or consumption coagulopathy, making anticoagulant therapy challenging in this scenario [104, 105].
Given that potentially significant drug interactions of both VKA and DOACs with concomitant immunosuppressive/antiviral medications may occur, switching to treatment dose low molecular weight heparin (LMWH) or unfractionated heparin (UFH) may be considered in hospitalized patients with moderate-to-severe COVID-19 [105, 106].

Oncological Patients

Patients with cancer, compared to cancer-free individuals, have a strongly increased thrombotic and bleeding risk. Bleeding risk is affected by anemia, thrombocytopenia, renal failure (which are common in these patients), and drug interactions with chemotherapeutics [107]
Anticoagulation and risk of bleeding complications in patients with cancer VTE have been extensively reviewed elsewhere [107, 108]. Results of RCTs showed non-inferior efficacy of factor Xa inhibitors (edoxaban, rivaroxaban, or apixaban) compared to LMWH for cancer associated VTE treatment, although rivaroxaban and edoxaban displayed higher bleeding rates, especially in gastrointestinal cancers [108]
Although cancer is emerging as an important risk factor for NVAF [109], the efficacy and safety of DOACs compared to VKAs in NVAF cancer patients have received scarce attention in RCTs [110]. Post-hoc analyses of pivotal DOAC RCTs, recent real-world and metanalysis data have shown that DOAC had a similar or better efficacy/safety profile than VKAs among NVAF patients with cancer compared to other NVAF patients [110, 111].

Stratification of Bleeding Risk

Predictors of Bleeding

History of previous bleeding is the most important risk factor associated with MB events in patients on anticoagulant therapy. Other relevant risk factors are advanced age, liver disease, renal failure, cancer, thrombocytopenia, coadministration of antiplatelet drugs, and excessive anticoagulation secondary to inappropriately high doses of DOAC or poor INR control [112, 113].
Preexisting, though clinically occult, mucosal lesions of gastrointestinal or urinary tracts should always be considered in case of bleeding events. In fact, both VKA- and DOAC-related bleeding are associated with an increased rate of new cancer diagnoses [114116]. Therefore, patients should be evaluated for age-appropriate colorectal cancer screening prior to initiation of oral anticoagulation [114, 115].

Scores for the Assessment of Bleeding Risk

Several clinical scoring systems are available for the stratification of bleeding risk in patients on anticoagulant therapy (Table 5, 6) [117128].
Table 5
Major bleeding prediction models in AF patients
Score
Author, year
Bleeding risk factors included
Risk scoring and MB annual incidence
OBRI
Beyth [117], 1998
Age ≥ 65 yrs, history of GIB, previous stroke, co-morbidities (recent MI, hematocrit < 30%, diabetes, creatinine > 1.5 mg/dL): 1 pt each
Low risk: 0 = 3% at 48 months
Intermediate risk: 1–2 = 12% at 48 months
High-risk: 3–4 = 53% at 48 months
HEMORR2HAGES
Gage [118], 2006
Prior bleed (2 pts). Liver/renal disease, ethanol abuse, malignancy, age > 75 yrs, low platelet count or function, uncontrolled hypertension, anemia, genetic factors (CYP2C9), risk of fall, stroke (1 pt each)
Low risk: 0 = 1.9%, 1 = 2.5%
Intermediate risk: 2 = 5.3%, 3 = 8.4%
High risk: 4 = 10.4%, 5 = 12.3%
ATRIA
Fang [119], 2011
Anemia, renal disease (eGFR < 30 ml/min): 3 pts each; age > 75 yrs: 2 pts; any prior bleeding, hypertension: 1 pt each
Low risk: 0–3 = 0.76%
Intermediate risk: 4 = 2.62%
High risk: 5–10 = 5.76%
HAS-BLED
Pisters [120], 2010
Hypertension (uncontrolled), abnormal renal or liver function, stroke, bleeding history/predisposition, labile INR (TTR < 60%), elderly (> 65 yrs), drugs (antiplatelets/NSAIDs) or excess alcohol (1 pt each)
Low risk: 0 = 1.13%
Moderate risk: 1 = 1.02%, 2 = 1.88%
High-risk: 3 = 3.74%, 4 = 8.70%, 5–9 = insufficient data
ORBIT
O’Brien [121], 2015
Reduced Hb (< 13 mg/dL M; < 12 mg/dL F), Ht (< 40% M, < 36% F), history of anemia and bleeding history: 2 pts each; eGFR < 60 mg/dL/1.73 m2, age ≥ 75 yrs, antiplatelet agent: 1 pt each
Low risk: 0–2 = 2.4%
Medium risk: 3 = 4.7%
High risk: ≥ 4 = 8.1%
ABC
Hijazi [122], 2016
Age, biomarkers (GDF-15, cTnT-hs, and Hb), and clinical history (previous bleeding)
Nomogram: Low risk = 0.77%
High risk = 30%
NBP
Barnett-Griness [123], 2022
Thrombocytopenia (< 99 × 103/μL): 9 pts; hypertension: 8 pts; M sex, antiplatelet therapy: 7 pts each; anemia (Hb < 13 g/dL M, < 12 g/dL F): 6 pts; prior MB: 5 pts; known fall risk: 4 pts; serum CH (mg/dL): 200–239 = 1 pt, 160–199 = 2 pts, < 160 = 5 pts; eGFR (mL/min): 60–89 = 4 pts, 30–59 = 6 pts, ≤ 29 = 8 pts
Continuous variable:
from 0.3% (0 points)
to 10.3% (59 points)
CH cholesterol, cTnT-hs high-sensitivity cardiac troponin T, eGFR estimated glomerular filtration rate, F female, GDF-15 growth differentiation factor-15, GIB gastrointestinal bleeding, Hb hemoglobin, Ht hematocrit, INR international normalized ratio, M male, MB major bleeding, MI myocardial infarction, NSAIDs nonsteroidal antiinflammatory drugs, pt point, TTR time in therapeutic range, yrs years
Table 6
Major bleeding prediction models in VTE patients
Score
Author, year
Bleeding risk factors included
High risk scoring and MB incidence
Nieuwenhuis [124], 1991
WHO performance status grade 2: 1 pt; WHO grade 3 or 4: 2 pts; history of a bleeding diathesis: 2 pts; recent (< 2 months) trauma or surgery: 1 pt; BSA < 2 m2: 2 pts
≥ 5 = 44% at 8 daysa
OBRI
Beyth [117], 1998
Age ≥ 65 yrs, history of GIB, previous stroke, co-morbidities (recent MI, hematocrit < 30%, diabetes, creatinine > 1.5 mg/dL): 1 pt each
≥ 3 = 53% at 48 months
Kuijer [125], 1999
Age ≥ 60 yrs, F sex, BSA ≤ 2, malignancy, long-acting coumarin (1 pt each, otherwise 0 points). Score = (1.6 × age) + (1.3 × sex) + (2.2 × malignancy) + (2.4 × BSA) + (1.3 × coumarin type)
≥ 6.25 = 7% at 3 months
HEMORR2HAGES
Gage [118], 2006
Prior bleed (2 pts). Liver/renal disease, ethanol abuse, malignancy, age > 75 yrs, low platelet count or function, uncontrolled hypertension, anemia, genetic factors (CYP2C9), risk of fall, stroke (1 pt each)
≥ 4 = 10.4% annually
RIETE
Ruíz-Gimenez [126], 2008
Recent MB: 2 pts; creatinine levels > 1.2 mg/dL, anemia: 1.5 pts each; cancer, clinically overt PE, age > 75 yrs: 1 pt each
≥ 5 = 6.2% at 3 months
ATRIA
Fang [119], 2011
Anemia, renal disease (eGFR < 30 ml/min): 3 pts each; age ≥ 75 years: 2 pts; any prior bleeding, hypertension: 1 pt each
≥ 5 = 5.8% annually
ACCP
Kearon [127], 2012
Age > 65 years, previous bleeding, active/metastatic cancer, renal failure, liver failure, thrombocytopenia, previous stroke, diabetes, anemia, antiplatelet therapy, poor AC control therapy, comorbidity and reduced functional capacity, recent surgery, frequent falls, NSAIDs, alcohol abuse: 1 pt each
≥ 2 = 12.8% 0–3 months; ≥ 6.5% annually
VTE-BLEED (XALIA register)
Klok [128], 2016
Active cancer: 2 pts; male with uncontrolled hypertension: 1 pt; anemia (Hb < 13 g/dL M or < 12 g/dL F): 1.5 pts; history of bleeding: 1.5 pts; age ≥ 60 years: 1.5 pt; eGRF 30–60 ml/min: 1.5 pts
≥ 2 = 12.6% at 6 months
AC anticoagulant, BSA body surface area, CYP2C9 cytochrome P450 2C9, eGFR estimated glomerular filtration rate, F female, GIB gastrointestinal bleeding, Hb hemoglobin, M male, MB major bleeding, MI myocardial infarction, NSAIDs nonsteroidal antiinflammatory drugs, pt point, WHO world health organization
aInitial VTE treatment with heparin or dalteparin
The HAS-BLED (“Hypertension, Abnormal renal/liver function, Stroke, Bleeding history or predisposition, Labile INR, Elderly, Drugs/alcohol”) is probably the most widely adopted score in the AF population, being extensively validated in large cohorts and in many important patient subgroups [2, 120, 129] (Table 5). A HAS-BLED score ≥ 3 is associated with a high bleeding risk [2]. However, as recently shown by Barnett Griness et al., HAS-BLED score shows the worst performance among the various available scores, with an area under the curve (AUC) of 0.5765 compared to the value of 0.6263 of the ORBIT score or to that of 0.6579 of the nine-items score developed by the same authors [123].
Different bleeding risk scores have also been developed for patients with VTE, including Registro Informatizado de Enfermedad TromboEmbólica (RIETE), American College of Chest Physician (ACCP), and VTE-BLEED score [126128] (Table 6). ACCP and VTE-BLEED scores appear to be the best validated available tools [129]. However, a recent systematic review and meta-analysis has shown that these scores have quite low specificity and modest sensitivity in identifying patients at high risk of MB events [130].
Therefore, clinical judgment after careful evaluation of individual patient modifiable and non-modifiable bleeding risk factors still remains the unreplaceable approach to minimize the individual risk of bleeding, thus improving the benefit/risk ratio of anticoagulant treatments [113, 129, 131].

Treatment of Bleeding Complications

Figure 1 summarizes the therapeutic management of bleeding complications on DOACs according to current guidelines [2, 3, 132]. The type of bleeding and patient/anticoagulant drug characteristics are key aspects to consider for successful bleeding management. Given the short half-life of DOACs, most non-MB complications can be safely managed only with discontinuation of the anticoagulant and supportive measures. Systolic blood pressure should be lowered to 140 mm Hg in all patients with ICH [133]. Specific coagulation tests that measure the anticoagulant activity of DOACs [diluted thrombin time (dTT) and anti-Xa chromogenic assay] play a key role in the management of MB that is not immediately life-threatening, in as much as they enable selecting only those patients who, having therapeutic plasma concentration of DOACs are truly in need of antifibrinolytic therapy and/or anticoagulant reversal agents [2, 3, 132]. Tranexamic acid should be considered in cases of MB, especially in trauma patients [2, 3, 132]. Conversely, high doses of this should be avoided in patients with GIB in whom it will not reduce mortality and may even increase the risk of VTE events, particularly in patients with liver disease or with suspected variceal bleeding [134]. Specific measures should be adopted in cirrhotic patients, as extensively reviewed elsewhere [99]. In the case of bleeding at a critical site (e.g., intracranial, ocular, thoracic, abdominal, pericardial, retroperitoneal)/life-threatening MB or MB not responding to the general control measures, specific (idarucizumab for dabigatran; andexanet alfa for FXa inhibitors) or unspecific [4-factor prothrombin complex concentrates (4-F PCC)] anticoagulant reversal agents can be indicated as a life-saving measure [2, 3, 132]. In patients receiving VKA treatment, the administration of 10 mg intravenous (IV) vitamin K and CCP-4F according to body weight and INR value is recommended [132]. Endoscopic, radiological, or surgical mechanical hemostasis procedures should be performed whenever necessary [2, 3, 132].

Resumption of Anticoagulant Therapy

Assessment of resumption of anticoagulant therapy after bleeding complications should consider location/severity of bleeding, whether there is an identifiable cause, the profile of the individual patient’s thrombotic and hemorrhagic risk, the presence of modifiable bleeding risk factors (e.g., supratherapeutic INR, acute or worsening renal failure, concomitant antiplatelet agent), and the appropriateness of anticoagulant prescription based on patient characteristics/clinical indication [132]. In most cases, resuming anticoagulation after a bleeding event provides a net clinical benefit [132]. In case of bleeding due to secondary causes (e.g., post-trauma) or reversible (e.g., genitourinary due to cancer), anticoagulation can generally be resumed once the cause of the bleeding has been eliminated [3].
In the event of MB, particularly if life-threatening, a careful risk–benefit assessment for resuming anticoagulation should be conducted in collaboration with other specialists (e.g., neurologist, surgeon, endoscopist, interventional radiologists) and shared with the patient. None of the bleeding risk scores have been studied in the specific situation of active or very recent bleeding. In case of high thrombotic risk conditions (e.g., mechanical heart valve prosthesis, valvular AF, NVAF with CHA2DS2-VASc score ≥ 4, transient ischemic attack/ischemic stroke within 3 months, VTE within 3 months, or recurrent or cancer-associated VTE), restarting the anticoagulant will likely result in a benefit for the patient even if the risk of rebleeding is high, and should occur early once hemostasis and clinical stability have been achieved [132]. In patients at both high thrombotic and bleeding risk with relative or absolute contraindication to resume anticoagulation (e.g., severe and life-threatening bleeding without a clear secondary or reversible/treatable cause), nonpharmacological therapies may be considered, such as devices for closing the left atrial appendage to reduce the thrombotic risk in AF or retrievable inferior vena cava (IVC) filters in case of DVT and/or PE.

Resumption of Anticoagulation After ICH

Limited data are available on resuming anticoagulation after ICH [135]. Factors associated with a high risk of ICH recurrence include the mechanism of bleeding (spontaneous versus traumatic), lobar location, and presence/number of microbleeds on magnetic resonance imaging (suggestive of amyloid angiopathy) [133]. A history of spontaneous ICH is a contraindication to anticoagulation according to labeling of VKAs and DOACs, unless the cause of the bleeding (such as uncontrolled hypertension, aneurysm or arteriovenous malformation, or dual/triple arteriothrombotic therapy) has been removed [3]. Anticoagulation resumption should be considered in patients with non-lobar ICH, depending on bleeding characteristics, modification of risk factors, and indication for anticoagulation [132, 133]. Conversely, resuming anticoagulation in those with lobar ICH secondary to amyloid angiopathy or spontaneous subdural hematomas should be implemented very cautiously under neurologist and/or neurosurgeon supervision, given a particularly high risk of rebleeding [132].
An important argument to switch from VKA to a safer DOAC is the lower risk of spontaneous as well as posttraumatic de novo and recurrent ICH [136138].
Optimal timing of anticoagulation resumption after ICH has been evaluated only in observational studies [135, 139]. Current guidelines recommend discontinuing oral anticoagulation for at least 4 weeks in patients without high thrombotic risk (e.g., mechanical heart valves) [3, 132, 133]. In stable patients, a prophylactic dose of heparin or LMWH can be started 2–4 days after the onset of bleeding [140]. Oral anticoagulation can be resumed after 14 days in patients with a stable ICH and a high risk of cerebral ischemia (e.g., those with mechanical valve prosthesis or NVAF with CHA2DS2VASc score > 4) [141, 142].

Resumption of Anticoagulation After GIB

Up to 25–50% of patients still do not restart oral anticoagulation after GIB, although available evidence suggests that the benefits of resuming anticoagulation (decreased thromboembolic events and mortality) outweigh the risk of bleeding in patients with NVAF as well as in those with VTE [45, 143, 144].
Timing of anticoagulation resumption of after GIB has not been systematically studied [132] and remains unclear. However, based on available information, it appears that approximately 7–14 days may provide the best balance between recurrent GIB, thromboembolism and risk of mortality [145, 146]. In fact, some studies have reported that starting earlier than 5–7 days may increase the risk of bleeding [132, 146]. European guidelines suggest restarting anticoagulation after GIB as early as clinically feasible [3].
The prescription of PPIs should be evaluated and is always indicated in patients with GIB associated with peptic ulcer disease, and so is eradication of Helicobacter pylori, whenever this infection is present [146]. Effective gastroprotection exerted by PPIs in preventing GIB complications, especially in patients with a prior history of peptic ulcer/GIB or requiring concomitant use of antiplatelet therapy, has been exhaustively demonstrated in patients receiving antiplatelet or VKA therapy, while data in DOAC treated patients are more limited [3, 55, 56].
A recent study on 948 patients (531 on VKAs and 417 on DOACs) hospitalized for GIB followed-up for up to 2 years has shown that the risk of recurrent bleeding associated with restarting anticoagulant is more influenced by patient characteristics (previous bleeding, index MB, lower eGFR) than by the time of anticoagulation resumption [147].
Patient features against resuming anticoagulant therapy after GIB include unidentifiable bleeding site, multiple GI tract angiodysplasias, no reversible/treatable causes, bleeding during treatment discontinuation, chronic alcohol abuse, need for dual antiplatelet therapy after percutaneous coronary intervention, and advanced age [3].

Choosing the Appropriate Anticoagulant

The choice of the proper anticoagulant should carefully consider the patient thrombotic and bleeding risk profile. In some patients (e.g., severe CKD, mechanical heart valves, valvular AF or major drug interactions, extreme body weights), only VKAs are indicated. In DOAC-eligible patients, based on the above reported literature evidence and expert opinions, it can be suggested to match the optimal DOAC to the patient profile, especially in those with AF, with the aim of reducing the risk of complications [66, 85, 148150] (Fig. 2).
In the elderly (≥ 75 years old) or frail NVAF patients, apixaban followed by edoxaban may be the first choice due to the excellent efficacy–safety profile demonstrated in these subpopulations, while dabigatran and rivaroxaban may be considered secondarily in those at low-bleeding risk [57, 63, 66]. All FXa inhibitors are viable options in elderly patients with VTE, while dabigatran 150 mg should not be the first choice, given the increased risk of MB reported in this subgroup in pooled data of RECOVER I and II [151].
Apixaban may be the first choice in AF as well as in VTE patients at high risk of GIB, while in those with AF, dabigatran 110 mg may also be a reasonable option [24, 48, 50, 148, 150]. In patients with dyspepsia/gastroesophageal reflux, dabigatran should be avoided; or treatment with PPI should be associated [148, 150].
In patients at risk of genitourinary bleeding there is no evidence supporting that any specific DOAC type is safer than warfarin [24]. Reduced risk of heavy menstrual bleeding, however, has been reported with dabigatran in a post-hoc analysis of the RE-COVER and RE-MEDY studies [152].
In AF patients with moderate-to-severe CKD (eGFR 15–49 ml/min) the first therapeutic choice may be apixaban 5 mg twice daily (2.5 mg twice daily if eGFR 15–29 ml/min or two dose reduction criteria are satisfied), edoxaban 30 mg once daily or rivaroxaban 15 mg daily; dabigatran 110 mg twice a day as a second choice in patients with moderate CKD (eGFR 30–49 ml/min), while it is contraindicated in patients with severe CKD (eGFR 15–30 ml/min) owing to its predominant renal elimination [3, 85, 148, 150]. Likewise, using DOACs less dependent on renal excretion may be preferred in VTE patients with moderate-severe CKD but DOAC dose reduction is not recommended in these patients, except for edoxaban [3, 22].
No evidence is available regarding the best efficacy and safety profile of individual DOAC in patients with liver disease [99]. Dabigatran may be preferred based on the prevailing renal excretion; while apixaban may appear preferable based on optimal GIB safety profile, it undergoes hepatic metabolism to a larger extent than other DOACs. Rivaroxaban is contraindicated in Child–Pugh class B patients and all DOACs should be avoided in Child–Pugh class C patients [3].
In severely obese patients (either BMI 40–49 kg/m2 or weight 120–140 kg), DOACs may cautiously be used, preferring apixaban, edoxaban or rivaroxaban in those with AF and apixaban or rivaroxaban in those with VTE [7174, 7880].
In low body weight patients (60–40 kg) with AF or VTE, apixaban and edoxaban may be preferred [3, 70, 81, 84, 85].
DOACs with low incidence of MB (such as apixaban, dabigatran 110 mg, and edoxaban) should be considered in AF patients with high bleeding risk (HASBLED ≥ 3) [148], while in those with high thrombotic risk and a low bleeding risk, dabigatran at a dose of 150 mg would be preferable, given that it is the only DOAC more effective than warfarin in reducing ischemic stroke [6].
Drug interactions must always be evaluated before prescribing DOACs: certain antiarrhythmics require a dose reduction of DOACs (e.g., dabigatran and edoxaban in verapamil users); moreover, many antineoplastic, antiepileptic, and antifungal drugs contraindicate the use of DOACs [2, 3, 150].
In case of therapeutic compliance concerns, once daily dosing DOAC (rivaroxaban or edoxaban) should be preferred because this could improve treatment adherence [3, 148, 149].
Finally, a structured follow-up involving general practitioners/consultants (cardiologist, internist, or hematologist) with surveillance of renal–hepatic function, symptoms/signs of bleeding, and medication compliance should be considered for all patients to optimize management and reduce the risk of complications, such as recommended by current guidelines [24]. A closer follow-up should be adopted for patients at high risk of bleeding, addressing all modifiable risk factors.

Conclusions

DOACs compared to VKAs have at least a comparable or better efficacy–safety profile for multiple indications, with significantly reduced of risk of ICH, while the risk of MB and GIB vary according to each individual DOAC.
Bleeding severity and intensity requested for its management are generally lower during therapy with DOACs than with VKAs. In most patients, supportive measures can be used as the therapeutic effect decreases due to the shorter half-life of DOACs than VKAs, while anticoagulant reversal agents are indicated for life-threatening bleeding complications. In most cases, resuming anticoagulation after a bleeding event will provide a net clinical benefit.
Correct patient selection, including careful assessment of bleeding risk profile and choice of appropriate DOAC type/dosing based on patient characteristics/comorbidities and presence of drug interactions is key to minimizing adverse events. Aimed at optimizing management efficacy while reducing the risk of complications, a closer, more structured follow-up should be considered for all patients at high risk of bleeding.

Acknowledgements

We are indebted to Doctor Giorgio Cioni for his outstanding clinical mentorship.

Funding

No funding or sponsorship was received for publication of this article.

Authorship

All named authors meet the International Committee of Medical Journal Editors (ICMJE) criteria for authorship for this manuscript, take responsibility for the integrity of the work as a whole, and have given final approval for the version to be published.

Author Contributions

SB conceived the study design. SB, ER, and DA wrote the first draft of the manuscript, and all the Authors contributed in the preparation of the final version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Disclosures

Stefano Ballestri, Elisa Romagnoli, Dimitriy Arioli, Valeria Coluccio, Alessandra Marrazzo, Athanasiou Afroditi, Di Girolamo Maria, Cappi Cinzia, Marco Marietta and Mariano Capitelli have nothing to disclose.
This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.

Compliance with Ethics Guidelines

This article is based on previously conducted studies and does not contain any studies with human participants or animals performed by any of the authors.

Data Availability

This article is based on previously conducted studies, the results of which are in the public domain.
Open AccessThis article is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License, which permits any non-commercial use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by-nc/​4.​0/​.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Jetzt e.Med zum Sonderpreis bestellen!

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Jetzt bestellen und 100 € sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Jetzt bestellen und 100 € sparen!

Literatur
1.
Zurück zum Zitat Di Carlo A, Bellino L, Consoli D, et al. Prevalence of atrial fibrillation in the Italian elderly population and projections from 2020 to 2060 for Italy and the European Union: the FAI Project. Europace. 2019;21:1468–75.CrossRef Di Carlo A, Bellino L, Consoli D, et al. Prevalence of atrial fibrillation in the Italian elderly population and projections from 2020 to 2060 for Italy and the European Union: the FAI Project. Europace. 2019;21:1468–75.CrossRef
2.
Zurück zum Zitat Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): the task force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur Heart J. 2021;42:373–498.CrossRef Hindricks G, Potpara T, Dagres N, et al. 2020 ESC guidelines for the diagnosis and management of atrial fibrillation developed in collaboration with the European Association for Cardio-Thoracic Surgery (EACTS): the task force for the diagnosis and management of atrial fibrillation of the European Society of Cardiology (ESC) developed with the special contribution of the European Heart Rhythm Association (EHRA) of the ESC. Eur Heart J. 2021;42:373–498.CrossRef
3.
Zurück zum Zitat Steffel J, Collins R, Antz M, et al. 2021 European Heart Rhythm Association practical guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation. Europace. 2021;23:1612–76.CrossRef Steffel J, Collins R, Antz M, et al. 2021 European Heart Rhythm Association practical guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation. Europace. 2021;23:1612–76.CrossRef
4.
Zurück zum Zitat Konstantinides SV, Meyer G, Becattini C, et al. 2019 ESC Guidelines for the diagnosis and management of acute pulmonary embolism developed in collaboration with the European Respiratory Society (ERS). Eur Heart J. 2020;41:543–603.CrossRef Konstantinides SV, Meyer G, Becattini C, et al. 2019 ESC Guidelines for the diagnosis and management of acute pulmonary embolism developed in collaboration with the European Respiratory Society (ERS). Eur Heart J. 2020;41:543–603.CrossRef
5.
Zurück zum Zitat Ortel TL, Neumann I, Ageno W, et al. American Society of Hematology 2020 guidelines for management of venous thromboembolism: treatment of deep vein thrombosis and pulmonary embolism. Blood Adv. 2020;4:4693–738.CrossRef Ortel TL, Neumann I, Ageno W, et al. American Society of Hematology 2020 guidelines for management of venous thromboembolism: treatment of deep vein thrombosis and pulmonary embolism. Blood Adv. 2020;4:4693–738.CrossRef
6.
Zurück zum Zitat Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361:1139–51.CrossRef Connolly SJ, Ezekowitz MD, Yusuf S, et al. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med. 2009;361:1139–51.CrossRef
7.
Zurück zum Zitat Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883–91.CrossRef Patel MR, Mahaffey KW, Garg J, et al. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883–91.CrossRef
8.
Zurück zum Zitat Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365:981–92.CrossRef Granger CB, Alexander JH, McMurray JJ, et al. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2011;365:981–92.CrossRef
9.
Zurück zum Zitat Giugliano RP, Ruff CT, Braunwald E, et al. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093–104.CrossRef Giugliano RP, Ruff CT, Braunwald E, et al. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093–104.CrossRef
10.
Zurück zum Zitat Ntaios G, Papavasileiou V, Makaritsis K, Vemmos K, Michel P, Lip GYH. Real-world setting comparison of nonvitamin-K antagonist oral anticoagulants versus vitamin-K antagonists for stroke prevention in atrial fibrillation: a systematic review and meta-analysis. Stroke. 2017;48:2494–503.CrossRef Ntaios G, Papavasileiou V, Makaritsis K, Vemmos K, Michel P, Lip GYH. Real-world setting comparison of nonvitamin-K antagonist oral anticoagulants versus vitamin-K antagonists for stroke prevention in atrial fibrillation: a systematic review and meta-analysis. Stroke. 2017;48:2494–503.CrossRef
11.
Zurück zum Zitat van den Ham HA, Souverein PC, Klungel OH, et al. Major bleeding in users of direct oral anticoagulants in atrial fibrillation: a pooled analysis of results from multiple population-based cohort studies. Pharmacoepidemiol Drug Saf. 2021;30:1339–52.CrossRef van den Ham HA, Souverein PC, Klungel OH, et al. Major bleeding in users of direct oral anticoagulants in atrial fibrillation: a pooled analysis of results from multiple population-based cohort studies. Pharmacoepidemiol Drug Saf. 2021;30:1339–52.CrossRef
13.
Zurück zum Zitat López-López JA, Sterne JAC, Thom HHZ, et al. Oral anticoagulants for prevention of stroke in atrial fibrillation: systematic review, network meta-analysis, and cost effectiveness analysis. BMJ. 2017;359: j5058.CrossRef López-López JA, Sterne JAC, Thom HHZ, et al. Oral anticoagulants for prevention of stroke in atrial fibrillation: systematic review, network meta-analysis, and cost effectiveness analysis. BMJ. 2017;359: j5058.CrossRef
14.
Zurück zum Zitat Fralick M, Colacci M, Schneeweiss S, Huybrechts KF, Lin KJ, Gagne JJ. Effectiveness and safety of apixaban compared with rivaroxaban for patients with atrial fibrillation in routine practice: a cohort study. Ann Intern Med. 2020;172:463–73. https://doi.org/10.7326/M19-2522.CrossRef Fralick M, Colacci M, Schneeweiss S, Huybrechts KF, Lin KJ, Gagne JJ. Effectiveness and safety of apixaban compared with rivaroxaban for patients with atrial fibrillation in routine practice: a cohort study. Ann Intern Med. 2020;172:463–73. https://​doi.​org/​10.​7326/​M19-2522.CrossRef
15.
Zurück zum Zitat Li G, Lip GYH, Holbrook A, et al. Direct comparative effectiveness and safety between non-vitamin K antagonist oral anticoagulants for stroke prevention in nonvalvular atrial fibrillation: a systematic review and meta-analysis of observational studies. Eur J Epidemiol. 2019;34:173–90.CrossRef Li G, Lip GYH, Holbrook A, et al. Direct comparative effectiveness and safety between non-vitamin K antagonist oral anticoagulants for stroke prevention in nonvalvular atrial fibrillation: a systematic review and meta-analysis of observational studies. Eur J Epidemiol. 2019;34:173–90.CrossRef
17.
Zurück zum Zitat Schulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med. 2009;361:2342–52.CrossRef Schulman S, Kearon C, Kakkar AK, et al. Dabigatran versus warfarin in the treatment of acute venous thromboembolism. N Engl J Med. 2009;361:2342–52.CrossRef
18.
Zurück zum Zitat Bauersachs R, Berkowitz SD, Brenner B, et al. Oral rivaroxaban for symptomatic venous thromboembolism. N Engl J Med. 2010;363:2499–510.CrossRef Bauersachs R, Berkowitz SD, Brenner B, et al. Oral rivaroxaban for symptomatic venous thromboembolism. N Engl J Med. 2010;363:2499–510.CrossRef
19.
Zurück zum Zitat Büller HR, Prins MH, Lensin AW, et al. Oral rivaroxaban for the treatment of symptomatic pulmonary embolism. N Engl J Med. 2012;366:1287–97.CrossRef Büller HR, Prins MH, Lensin AW, et al. Oral rivaroxaban for the treatment of symptomatic pulmonary embolism. N Engl J Med. 2012;366:1287–97.CrossRef
20.
Zurück zum Zitat Prins MH, Lensing AW, Bauersachs R, et al. Oral rivaroxaban versus standard therapy for the treatment of symptomatic venous thromboembolism: a pooled analysis of the EINSTEIN-DVT and PE randomized studies. Thromb J. 2013;11:21.CrossRef Prins MH, Lensing AW, Bauersachs R, et al. Oral rivaroxaban versus standard therapy for the treatment of symptomatic venous thromboembolism: a pooled analysis of the EINSTEIN-DVT and PE randomized studies. Thromb J. 2013;11:21.CrossRef
21.
Zurück zum Zitat Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med. 2013;369:799–808.CrossRef Agnelli G, Buller HR, Cohen A, et al. Oral apixaban for the treatment of acute venous thromboembolism. N Engl J Med. 2013;369:799–808.CrossRef
22.
Zurück zum Zitat Buller HR, Décousus H, Grosso MA, et al. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism. N Engl J Med. 2013;369:1406–15.CrossRef Buller HR, Décousus H, Grosso MA, et al. Edoxaban versus warfarin for the treatment of symptomatic venous thromboembolism. N Engl J Med. 2013;369:1406–15.CrossRef
23.
Zurück zum Zitat Abraham NS, Singh S, Alexander GC, et al. Comparative risk of gastrointestinal bleeding with dabigatran, rivaroxaban, and warfarin: population based cohort study. BMJ. 2015;350: h1857.CrossRef Abraham NS, Singh S, Alexander GC, et al. Comparative risk of gastrointestinal bleeding with dabigatran, rivaroxaban, and warfarin: population based cohort study. BMJ. 2015;350: h1857.CrossRef
24.
Zurück zum Zitat Vinogradova Y, Coupland C, Hill T, Hippisley-Cox J. Risks and benefits of direct oral anticoagulants versus warfarin in a real world setting: cohort study in primary care. BMJ. 2018;362: k2505.CrossRef Vinogradova Y, Coupland C, Hill T, Hippisley-Cox J. Risks and benefits of direct oral anticoagulants versus warfarin in a real world setting: cohort study in primary care. BMJ. 2018;362: k2505.CrossRef
25.
Zurück zum Zitat Goldhaber SZ, Ageno W, Casella IB, et al. Safety and effectiveness of dabigatran in routine clinical practice: the RE-COVERY DVT/PE study. J Thromb Thrombolysis. 2022;53:399–409.CrossRef Goldhaber SZ, Ageno W, Casella IB, et al. Safety and effectiveness of dabigatran in routine clinical practice: the RE-COVERY DVT/PE study. J Thromb Thrombolysis. 2022;53:399–409.CrossRef
26.
Zurück zum Zitat Bounameaux H, Haas S, Farjat AE, et al. Comparative effectiveness of oral anticoagulants in venous thromboembolism: GARFIELD-VTE. Thromb Res. 2020;191:103–12.CrossRef Bounameaux H, Haas S, Farjat AE, et al. Comparative effectiveness of oral anticoagulants in venous thromboembolism: GARFIELD-VTE. Thromb Res. 2020;191:103–12.CrossRef
27.
Zurück zum Zitat Haas S, Mantovani LG, Kreutz R, et al. Anticoagulant treatment for venous thromboembolism: a pooled analysis and additional results of the XALIA and XALIA-LEA noninterventional studies. Res Pract Thromb Haemost. 2021;5:426–38.CrossRef Haas S, Mantovani LG, Kreutz R, et al. Anticoagulant treatment for venous thromboembolism: a pooled analysis and additional results of the XALIA and XALIA-LEA noninterventional studies. Res Pract Thromb Haemost. 2021;5:426–38.CrossRef
28.
Zurück zum Zitat Bertoletti L, Gusto G, Khachatryan A, et al. Effectiveness and safety of oral anticoagulants in the treatment of acute venous thromboembolism: a Nationwide Comparative Cohort Study in France. Thromb Haemost. 2022. https://doi.org/10.1055/a-1731-3922 (Epub ahead of print).CrossRef Bertoletti L, Gusto G, Khachatryan A, et al. Effectiveness and safety of oral anticoagulants in the treatment of acute venous thromboembolism: a Nationwide Comparative Cohort Study in France. Thromb Haemost. 2022. https://​doi.​org/​10.​1055/​a-1731-3922 (Epub ahead of print).CrossRef
29.
Zurück zum Zitat Weycker D, Li X, Wygant GD, et al. Effectiveness and safety of apixaban versus warfarin as outpatient treatment of venous thromboembolism in US clinical practice. Thromb Haemost. 2018;118:1951–61.CrossRef Weycker D, Li X, Wygant GD, et al. Effectiveness and safety of apixaban versus warfarin as outpatient treatment of venous thromboembolism in US clinical practice. Thromb Haemost. 2018;118:1951–61.CrossRef
30.
Zurück zum Zitat Dawwas GK, Smith SM, Dietrich E, Lo-Ciganic WH, Park H. Comparative effectiveness and safety of apixaban versus warfarin in patients with venous thromboembolism. Am J Health Syst Pharm. 2020;77:188–95.CrossRef Dawwas GK, Smith SM, Dietrich E, Lo-Ciganic WH, Park H. Comparative effectiveness and safety of apixaban versus warfarin in patients with venous thromboembolism. Am J Health Syst Pharm. 2020;77:188–95.CrossRef
31.
Zurück zum Zitat Guo JD, Hlavacek P, Rosenblatt L, et al. Safety and effectiveness of apixaban compared with warfarin among clinically-relevant subgroups of venous thromboembolism patients in the United States Medicare populatioldon. Thromb Res. 2021;198:163–70.CrossRef Guo JD, Hlavacek P, Rosenblatt L, et al. Safety and effectiveness of apixaban compared with warfarin among clinically-relevant subgroups of venous thromboembolism patients in the United States Medicare populatioldon. Thromb Res. 2021;198:163–70.CrossRef
32.
Zurück zum Zitat Giustozzi M, Franco L, Vedovati MC, Becattini C, Agnelli G. Safety of direct oral anticoagulants versus traditional anticoagulants in venous thromboembolism. J Thromb Thrombolysis. 2019;48:439–53.CrossRef Giustozzi M, Franco L, Vedovati MC, Becattini C, Agnelli G. Safety of direct oral anticoagulants versus traditional anticoagulants in venous thromboembolism. J Thromb Thrombolysis. 2019;48:439–53.CrossRef
33.
Zurück zum Zitat Aryal MR, Gosain R, Donato A, et al. Systematic review and meta-analysis of the efficacy and safety of apixaban compared to rivaroxaban in acute VTE in the real world. Blood Adv. 2019;3:2381–7.CrossRef Aryal MR, Gosain R, Donato A, et al. Systematic review and meta-analysis of the efficacy and safety of apixaban compared to rivaroxaban in acute VTE in the real world. Blood Adv. 2019;3:2381–7.CrossRef
34.
Zurück zum Zitat Liu ZY, Zhang HX, Ma LY, et al. Non-vitamin K antagonist oral anticoagulants in venous thromboembolism patients: a meta-analysis of real-world studies. BMC Cardiovasc Disord. 2022;22:105.CrossRef Liu ZY, Zhang HX, Ma LY, et al. Non-vitamin K antagonist oral anticoagulants in venous thromboembolism patients: a meta-analysis of real-world studies. BMC Cardiovasc Disord. 2022;22:105.CrossRef
35.
Zurück zum Zitat Schulman S, Kearon C, Kakkar AK, et al. Extended use of dabigatran, warfarin, or placebo in venous thromboembolism. N Engl J Med. 2013;368:709–18.CrossRef Schulman S, Kearon C, Kakkar AK, et al. Extended use of dabigatran, warfarin, or placebo in venous thromboembolism. N Engl J Med. 2013;368:709–18.CrossRef
36.
Zurück zum Zitat Weitz JI, Lensing AWA, Prins MH, et al. Rivaroxaban or aspirin for extended treatment of venous thromboembolism. N Engl J Med. 2017;376:1211–22.CrossRef Weitz JI, Lensing AWA, Prins MH, et al. Rivaroxaban or aspirin for extended treatment of venous thromboembolism. N Engl J Med. 2017;376:1211–22.CrossRef
37.
Zurück zum Zitat Agnelli G, Buller HR, Cohen A, et al. Apixaban for extended treatment of venous thromboembolism. N Engl J Med. 2013;368:699–708.CrossRef Agnelli G, Buller HR, Cohen A, et al. Apixaban for extended treatment of venous thromboembolism. N Engl J Med. 2013;368:699–708.CrossRef
38.
Zurück zum Zitat Wang KL, van Es N, Cameron C, Castellucci LA, Büller HR, Carrier M. Extended treatment of venous thromboembolism: a systematic review and network meta-analysis. Heart. 2019;105:545–52.CrossRef Wang KL, van Es N, Cameron C, Castellucci LA, Büller HR, Carrier M. Extended treatment of venous thromboembolism: a systematic review and network meta-analysis. Heart. 2019;105:545–52.CrossRef
39.
Zurück zum Zitat Khan F, Tritschler T, Kimpton M, et al. Long-term risk for major bleeding during extended oral anticoagulant therapy for first unprovoked venous thromboembolism: a systematic review and meta-analysis. Ann Intern Med. 2021;174:1420–9.CrossRef Khan F, Tritschler T, Kimpton M, et al. Long-term risk for major bleeding during extended oral anticoagulant therapy for first unprovoked venous thromboembolism: a systematic review and meta-analysis. Ann Intern Med. 2021;174:1420–9.CrossRef
40.
Zurück zum Zitat Halvorsen S, Storey RF, Rocca B, et al. Management of antithrombotic therapy after bleeding in patients with coronary artery disease and/or atrial fibrillation: expert consensus paper of the European Society of Cardiology Working Group on thrombosis. Eur Heart J. 2017;38:1455–62. Halvorsen S, Storey RF, Rocca B, et al. Management of antithrombotic therapy after bleeding in patients with coronary artery disease and/or atrial fibrillation: expert consensus paper of the European Society of Cardiology Working Group on thrombosis. Eur Heart J. 2017;38:1455–62.
41.
Zurück zum Zitat Meschia JF. Predicting who will experience cerebral hemorrhage when anticoagulated. Mayo Clin Proc. 2020;95:2057–9.CrossRef Meschia JF. Predicting who will experience cerebral hemorrhage when anticoagulated. Mayo Clin Proc. 2020;95:2057–9.CrossRef
42.
Zurück zum Zitat Lv M, Wu T, Jiang S, Chen W, Zhang J. Risk of intracranial hemorrhage caused by direct oral anticoagulants for stroke prevention in patients with atrial fibrillation (from a network meta-analysis of randomized controlled trials). Am J Cardiol. 2022;1(162):92–9.CrossRef Lv M, Wu T, Jiang S, Chen W, Zhang J. Risk of intracranial hemorrhage caused by direct oral anticoagulants for stroke prevention in patients with atrial fibrillation (from a network meta-analysis of randomized controlled trials). Am J Cardiol. 2022;1(162):92–9.CrossRef
43.
Zurück zum Zitat Wu T, Lv C, Wu L, et al. Risk of intracranial hemorrhage with direct oral anticoagulants: a systematic review and meta-analysis of randomized controlled trials. J Neurol. 2022;269:664–75.CrossRef Wu T, Lv C, Wu L, et al. Risk of intracranial hemorrhage with direct oral anticoagulants: a systematic review and meta-analysis of randomized controlled trials. J Neurol. 2022;269:664–75.CrossRef
44.
Zurück zum Zitat Cheung KS, Leung WK. Gastrointestinal bleeding in patients on novel oral anticoagulants: risk, prevention and management. World J Gastroenterol. 2017;23:1954–63.CrossRef Cheung KS, Leung WK. Gastrointestinal bleeding in patients on novel oral anticoagulants: risk, prevention and management. World J Gastroenterol. 2017;23:1954–63.CrossRef
46.
Zurück zum Zitat Noseworthy PA, Yao X, Abraham NS, et al. Direct comparison of dabigatran, rivaroxaban, and apixaban for effectiveness and safety in nonvalvular atrial fibrillation. Chest. 2016;150:1302–12.CrossRef Noseworthy PA, Yao X, Abraham NS, et al. Direct comparison of dabigatran, rivaroxaban, and apixaban for effectiveness and safety in nonvalvular atrial fibrillation. Chest. 2016;150:1302–12.CrossRef
47.
Zurück zum Zitat Abraham NS, Noseworthy PA, Yao X, et al. Gastrointestinal safety of direct oral anticoagulants: a large population-based study. Gastroenterology. 2017;152:1014–22.CrossRef Abraham NS, Noseworthy PA, Yao X, et al. Gastrointestinal safety of direct oral anticoagulants: a large population-based study. Gastroenterology. 2017;152:1014–22.CrossRef
48.
Zurück zum Zitat Mazurek M, Lip GYH. Gastrointestinal bleeding and direct oral anticoagulants amongst pati ents with atrial fibrillation in the “Real World.” Gastroenterology. 2017;152:932–4.CrossRef Mazurek M, Lip GYH. Gastrointestinal bleeding and direct oral anticoagulants amongst pati ents with atrial fibrillation in the “Real World.” Gastroenterology. 2017;152:932–4.CrossRef
49.
Zurück zum Zitat Rutherford OW, Jonasson C, Ghanima W, et al. Comparison of dabigatran, rivaroxaban, and apixaban for effectiveness and safety in atrial fibrillation: a nationwide cohort study. Eur Heart J Cardiovasc Pharmacother. 2020;6:75–85.CrossRef Rutherford OW, Jonasson C, Ghanima W, et al. Comparison of dabigatran, rivaroxaban, and apixaban for effectiveness and safety in atrial fibrillation: a nationwide cohort study. Eur Heart J Cardiovasc Pharmacother. 2020;6:75–85.CrossRef
50.
Zurück zum Zitat Ingason AB, Hreinsson JP, Ágústsson AS, et al. Rivaroxaban is associated with higher rates of gastrointestinal bleeding than other direct oral anticoagulants: a Nationwide Propensity Score-weighted study. Ann Intern Med. 2021;174:1493–502.CrossRef Ingason AB, Hreinsson JP, Ágústsson AS, et al. Rivaroxaban is associated with higher rates of gastrointestinal bleeding than other direct oral anticoagulants: a Nationwide Propensity Score-weighted study. Ann Intern Med. 2021;174:1493–502.CrossRef
51.
Zurück zum Zitat Lip GYH, Keshishian AV, Zhang Y, et al. Oral anticoagulants for nonvalvular atrial fibrillation in patients with high risk of gastrointestinal bleeding. JAMA Netw Open. 2021;4: e2120064.CrossRef Lip GYH, Keshishian AV, Zhang Y, et al. Oral anticoagulants for nonvalvular atrial fibrillation in patients with high risk of gastrointestinal bleeding. JAMA Netw Open. 2021;4: e2120064.CrossRef
52.
Zurück zum Zitat Gu ZC, Wei AH, Zhang C, et al. Risk of major gastrointestinal bleeding with new vs conventional oral anticoagulants: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2020;18:792-799.e61.CrossRef Gu ZC, Wei AH, Zhang C, et al. Risk of major gastrointestinal bleeding with new vs conventional oral anticoagulants: a systematic review and meta-analysis. Clin Gastroenterol Hepatol. 2020;18:792-799.e61.CrossRef
53.
Zurück zum Zitat Grymonprez M, Steurbaut S, De Backer TL, et al. Effectiveness and safety of oral anticoagulants in older patients with atrial fibrillation: a systematic review and meta-analysis. Front Pharmacol. 2020;11: 583311.CrossRef Grymonprez M, Steurbaut S, De Backer TL, et al. Effectiveness and safety of oral anticoagulants in older patients with atrial fibrillation: a systematic review and meta-analysis. Front Pharmacol. 2020;11: 583311.CrossRef
54.
Zurück zum Zitat Dans AL, Connolly SJ, Wallentin L, et al. Concomitant use of antiplatelet therapy with dabigatran or warfarin in the randomized evaluation of longterm anticoagulation therapy (RE-LY) trial. Circulation. 2013;127:634–40.CrossRef Dans AL, Connolly SJ, Wallentin L, et al. Concomitant use of antiplatelet therapy with dabigatran or warfarin in the randomized evaluation of longterm anticoagulation therapy (RE-LY) trial. Circulation. 2013;127:634–40.CrossRef
55.
Zurück zum Zitat Ray WA, Chung CP, Murray KT, et al. Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding. JAMA. 2018;320:2221–30.CrossRef Ray WA, Chung CP, Murray KT, et al. Association of oral anticoagulants and proton pump inhibitor cotherapy with hospitalization for upper gastrointestinal tract bleeding. JAMA. 2018;320:2221–30.CrossRef
56.
Zurück zum Zitat Komen J, Pottegård A, Hjemdahl P, et al. Non-vitamin K antagonist oral anticoagulants, proton pump inhibitors and gastrointestinal bleeds. Heart. 2022;108:613–8.CrossRef Komen J, Pottegård A, Hjemdahl P, et al. Non-vitamin K antagonist oral anticoagulants, proton pump inhibitors and gastrointestinal bleeds. Heart. 2022;108:613–8.CrossRef
57.
Zurück zum Zitat Kato ET, Goto S, Giugliano RP. Overview of oral antithrombotic treatment in elderly patients with atrial fibrillation. Ageing Res Rev. 2019;49:115–24.CrossRef Kato ET, Goto S, Giugliano RP. Overview of oral antithrombotic treatment in elderly patients with atrial fibrillation. Ageing Res Rev. 2019;49:115–24.CrossRef
58.
Zurück zum Zitat Malik AH, Yandrapalli S, Aronow WS, Panza JA, Cooper HA. Meta-analysis of direct-acting oral anticoagulants compared with warfarin in patients >75 years of age. Am J Cardiol. 2019;123:2051–7.CrossRef Malik AH, Yandrapalli S, Aronow WS, Panza JA, Cooper HA. Meta-analysis of direct-acting oral anticoagulants compared with warfarin in patients >75 years of age. Am J Cardiol. 2019;123:2051–7.CrossRef
59.
Zurück zum Zitat Ray WA, Chung CP, Stein CM, et al. Association of rivaroxaban vs apixaban with major ischemic or hemorrhagic events in patients with atrial fibrillation. JAMA. 2021;326:2395–404.CrossRef Ray WA, Chung CP, Stein CM, et al. Association of rivaroxaban vs apixaban with major ischemic or hemorrhagic events in patients with atrial fibrillation. JAMA. 2021;326:2395–404.CrossRef
60.
Zurück zum Zitat Richter D, Guasti L, Walker D, et al. Frailty in cardiology: definition, assessment and clinical implications for general cardiology. A consensus document of the Council for Cardiology Practice (CCP), Association for Acute Cardio Vascular Care (ACVC), Association of Cardiovascular Nursing and Allied Professions (ACNAP), European Association of Preventive Cardiology (EAPC), European Heart Rhythm Association (EHRA), Council on Valvular Heart Diseases (VHD), Council on Hypertension (CHT), Council of Cardio-Oncology (CCO), Working Group (WG) Aorta and Peripheral Vascular Diseases, WG e-Cardiology, WG Thrombosis, of the European Society of Cardiology, European Primary Care Cardiology Society (EPCCS). Eur J Prev Cardiol. 2022;29:216–27.CrossRef Richter D, Guasti L, Walker D, et al. Frailty in cardiology: definition, assessment and clinical implications for general cardiology. A consensus document of the Council for Cardiology Practice (CCP), Association for Acute Cardio Vascular Care (ACVC), Association of Cardiovascular Nursing and Allied Professions (ACNAP), European Association of Preventive Cardiology (EAPC), European Heart Rhythm Association (EHRA), Council on Valvular Heart Diseases (VHD), Council on Hypertension (CHT), Council of Cardio-Oncology (CCO), Working Group (WG) Aorta and Peripheral Vascular Diseases, WG e-Cardiology, WG Thrombosis, of the European Society of Cardiology, European Primary Care Cardiology Society (EPCCS). Eur J Prev Cardiol. 2022;29:216–27.CrossRef
63.
Zurück zum Zitat Kim DH, Pawar A, Gagne JJ, Bessette LG, Lee H, Glynn RJ, Schneeweiss S. Frailty and clinical outcomes of direct oral anticoagulants versus warfarin in older adults with atrial fibrillation: a cohort study. Ann Intern Med. 2021;174:1214–23. https://doi.org/10.7326/M20-7141.CrossRef Kim DH, Pawar A, Gagne JJ, Bessette LG, Lee H, Glynn RJ, Schneeweiss S. Frailty and clinical outcomes of direct oral anticoagulants versus warfarin in older adults with atrial fibrillation: a cohort study. Ann Intern Med. 2021;174:1214–23. https://​doi.​org/​10.​7326/​M20-7141.CrossRef
64.
Zurück zum Zitat Steffel J, Giugliano RP, Braunwald E, et al. Edoxaban versus warfarin in atrial fibrillation patients at risk of falling: ENGAGE AF-TIMI 48 analysis. J Am Coll Cardiol. 2016;68:1169–78.CrossRef Steffel J, Giugliano RP, Braunwald E, et al. Edoxaban versus warfarin in atrial fibrillation patients at risk of falling: ENGAGE AF-TIMI 48 analysis. J Am Coll Cardiol. 2016;68:1169–78.CrossRef
65.
Zurück zum Zitat Rao MP, Vinereanu D, Wojdyla DM, et al. Clinical outcomes and history of fall in patients with atrial fibrillation treated with oral anticoagulation: insights from the ARISTOTLE trial. Am J Med. 2018;131:269-75e2.CrossRef Rao MP, Vinereanu D, Wojdyla DM, et al. Clinical outcomes and history of fall in patients with atrial fibrillation treated with oral anticoagulation: insights from the ARISTOTLE trial. Am J Med. 2018;131:269-75e2.CrossRef
66.
Zurück zum Zitat Giugliano RP. Non-vitamin K antagonist oral anticoagulants in older and frail patients with atrial fibrillation. Eur Heart J Suppl. 2022;24:A1–10.CrossRef Giugliano RP. Non-vitamin K antagonist oral anticoagulants in older and frail patients with atrial fibrillation. Eur Heart J Suppl. 2022;24:A1–10.CrossRef
67.
Zurück zum Zitat Gross PL, Chan NC. Thromboembolism in older adults. Front Med. 2021;7: 470016.CrossRef Gross PL, Chan NC. Thromboembolism in older adults. Front Med. 2021;7: 470016.CrossRef
68.
Zurück zum Zitat Zhou B, Wu H, Wang C, Lou B, She J. Impact of age, sex, and renal function on the efficacy and safety of direct oral anticoagulants vs. vitamin k antagonists for the treatment of acute venous thromboembolism: a meta-analysis of 22,040 patients. Front Cardiovasc Med. 2021;8:700740.CrossRef Zhou B, Wu H, Wang C, Lou B, She J. Impact of age, sex, and renal function on the efficacy and safety of direct oral anticoagulants vs. vitamin k antagonists for the treatment of acute venous thromboembolism: a meta-analysis of 22,040 patients. Front Cardiovasc Med. 2021;8:700740.CrossRef
70.
Zurück zum Zitat Wang SY, Giugliano RP. Non-vitamin K antagonist oral anticoagulant for atrial fibrillation in obese patients. Am J Cardiol. 2020;127:176–83.CrossRef Wang SY, Giugliano RP. Non-vitamin K antagonist oral anticoagulant for atrial fibrillation in obese patients. Am J Cardiol. 2020;127:176–83.CrossRef
71.
Zurück zum Zitat Boriani G, Ruff CT, Kuder JF, et al. Relationship between body mass index and outcomes in patients with atrial fibrillation treated with edoxaban or warfarin in the ENGAGE AF-TIMI 48 trial. Eur Heart J. 2019;40:1541–50.CrossRef Boriani G, Ruff CT, Kuder JF, et al. Relationship between body mass index and outcomes in patients with atrial fibrillation treated with edoxaban or warfarin in the ENGAGE AF-TIMI 48 trial. Eur Heart J. 2019;40:1541–50.CrossRef
72.
Zurück zum Zitat Hohnloser SH, Fudim M, Alexander JH, et al. Efficacy and safety of apixaban versus warfarin in patients with atrial fibrillation and extremes in body weight. Circulation. 2019;139:2292–300.CrossRef Hohnloser SH, Fudim M, Alexander JH, et al. Efficacy and safety of apixaban versus warfarin in patients with atrial fibrillation and extremes in body weight. Circulation. 2019;139:2292–300.CrossRef
73.
Zurück zum Zitat Boriani G, Ruff CT, Kuder JF, et al. Edoxaban versus warfarin in patients with atrial fibrillation at the extremes of body weight: an analysis from the ENGAGE AF-TIMI 48 trial. Thromb Haemost. 2021;121:140–9.CrossRef Boriani G, Ruff CT, Kuder JF, et al. Edoxaban versus warfarin in patients with atrial fibrillation at the extremes of body weight: an analysis from the ENGAGE AF-TIMI 48 trial. Thromb Haemost. 2021;121:140–9.CrossRef
74.
Zurück zum Zitat Martin AC, Thomas W, Mahir Z, et al. Direct oral anticoagulant concentrations in obese and high body weight patients: a cohort study. Thromb Haemost. 2021;121:224–33.CrossRef Martin AC, Thomas W, Mahir Z, et al. Direct oral anticoagulant concentrations in obese and high body weight patients: a cohort study. Thromb Haemost. 2021;121:224–33.CrossRef
75.
Zurück zum Zitat Costa OS, Beyer-Westendorf J, Ashton V, et al. Effectiveness and safety of rivaroxaban versus warfarin in obese nonvalvular atrial fibrillation patients: analysis of electronic health record data. Curr Med Res Opin. 2020;36:1081–8.CrossRef Costa OS, Beyer-Westendorf J, Ashton V, et al. Effectiveness and safety of rivaroxaban versus warfarin in obese nonvalvular atrial fibrillation patients: analysis of electronic health record data. Curr Med Res Opin. 2020;36:1081–8.CrossRef
76.
Zurück zum Zitat Berger JS, Laliberté F, Kharat A, Lejeune D, Moore KT, Jung Y, Lefebvre P, Ashton V. Real-world effectiveness and safety of rivaroxaban versus warfarin among non-valvular atrial fibrillation patients with obesity in a US population. Curr Med Res Opin. 2021;37:881–90.CrossRef Berger JS, Laliberté F, Kharat A, Lejeune D, Moore KT, Jung Y, Lefebvre P, Ashton V. Real-world effectiveness and safety of rivaroxaban versus warfarin among non-valvular atrial fibrillation patients with obesity in a US population. Curr Med Res Opin. 2021;37:881–90.CrossRef
77.
Zurück zum Zitat Deitelzweig S, Keshishian A, Kang A, et al. Effectiveness and safety of oral anticoagulants among nvaf patients with obesity: insights from the ARISTOPHANES study. J Clin Med. 2020;9:1633.CrossRef Deitelzweig S, Keshishian A, Kang A, et al. Effectiveness and safety of oral anticoagulants among nvaf patients with obesity: insights from the ARISTOPHANES study. J Clin Med. 2020;9:1633.CrossRef
78.
Zurück zum Zitat Martin KA, Beyer-Westendorf J, Davidson BL, Huisman MV, Sandset PM, Moll S. Use of direct oral anticoagulants in patients with obesity for treatment and prevention of venous thromboembolism: updated communication from the ISTH SSC Subcommittee on control of anticoagulation. J Thromb Haemost. 2021;19:1874–82.CrossRef Martin KA, Beyer-Westendorf J, Davidson BL, Huisman MV, Sandset PM, Moll S. Use of direct oral anticoagulants in patients with obesity for treatment and prevention of venous thromboembolism: updated communication from the ISTH SSC Subcommittee on control of anticoagulation. J Thromb Haemost. 2021;19:1874–82.CrossRef
79.
Zurück zum Zitat Cohen AT, Pan S, Byon W, Ilyas BS, Taylor T, Lee TC. Efficacy, safety, and exposure of apixaban in patients with high body weight or obesity and venous thromboembolism: insights from AMPLIFY. Adv Ther. 2021;38:3003–18.CrossRef Cohen AT, Pan S, Byon W, Ilyas BS, Taylor T, Lee TC. Efficacy, safety, and exposure of apixaban in patients with high body weight or obesity and venous thromboembolism: insights from AMPLIFY. Adv Ther. 2021;38:3003–18.CrossRef
80.
Zurück zum Zitat Crouch A, Ng TH, Kelley D, Knight T, Edwin S, ASCEND-HIGHER, Giuliano C. Multi-center retrospective study evaluating the efficacy and safety of apixaban versus warfarin for treatment of venous thromboembolism in patients with severe obesity. Pharmacotherapy. 2022;42:119–33.CrossRef Crouch A, Ng TH, Kelley D, Knight T, Edwin S, ASCEND-HIGHER, Giuliano C. Multi-center retrospective study evaluating the efficacy and safety of apixaban versus warfarin for treatment of venous thromboembolism in patients with severe obesity. Pharmacotherapy. 2022;42:119–33.CrossRef
81.
Zurück zum Zitat Pandey AK, Eikelboom JW. Direct oral anticoagulant dosing in extremes of body weight: time to revisit the guidelines? Thromb Haemost. 2021;121:118–20.CrossRef Pandey AK, Eikelboom JW. Direct oral anticoagulant dosing in extremes of body weight: time to revisit the guidelines? Thromb Haemost. 2021;121:118–20.CrossRef
82.
Zurück zum Zitat Ruff CT, Giugliano RP, Braunwald E, et al. Association between edoxaban dose, concentration, anti-factor Xa activity, and outcomes: an analysis of data from the randomised, double-blind ENGAGE AFTIMI 48 trial. Lancet. 2015;385:2288–95.CrossRef Ruff CT, Giugliano RP, Braunwald E, et al. Association between edoxaban dose, concentration, anti-factor Xa activity, and outcomes: an analysis of data from the randomised, double-blind ENGAGE AFTIMI 48 trial. Lancet. 2015;385:2288–95.CrossRef
83.
Zurück zum Zitat Covert K, Branam DL. Direct-acting oral anticoagulant use at extremes of body weight: Literature review and recommendations. Am J Health Syst Pharm. 2020;77:865–76.CrossRef Covert K, Branam DL. Direct-acting oral anticoagulant use at extremes of body weight: Literature review and recommendations. Am J Health Syst Pharm. 2020;77:865–76.CrossRef
85.
Zurück zum Zitat Chen A, Stecker E, Warden BA. Direct oral anticoagulant use: a practical guide to common clinical challenges. J Am Heart Assoc. 2020;9:e017559.CrossRef Chen A, Stecker E, Warden BA. Direct oral anticoagulant use: a practical guide to common clinical challenges. J Am Heart Assoc. 2020;9:e017559.CrossRef
86.
Zurück zum Zitat Kreutz R, Camm AJ, Rossing P. Concomitant diabetes with atrial fibrillation and anticoagulation management considerations. Eur Heart J Suppl. 2020;22:O78–86.CrossRef Kreutz R, Camm AJ, Rossing P. Concomitant diabetes with atrial fibrillation and anticoagulation management considerations. Eur Heart J Suppl. 2020;22:O78–86.CrossRef
87.
Zurück zum Zitat Plitt A, Zelniker TA, Park JG, et al. Patients with diabetes mellitus and atrial fibrillation treated with non-vitamin K antagonist oral anticoagulants: meta-analysis of eight outcomes in 58 634 patients across four randomized controlled trials. Eur Heart J Cardiovasc Pharmacother. 2021;7:f40–9.CrossRef Plitt A, Zelniker TA, Park JG, et al. Patients with diabetes mellitus and atrial fibrillation treated with non-vitamin K antagonist oral anticoagulants: meta-analysis of eight outcomes in 58 634 patients across four randomized controlled trials. Eur Heart J Cardiovasc Pharmacother. 2021;7:f40–9.CrossRef
88.
Zurück zum Zitat Cao B, Yao X, Zhang L, Hu X, Chen M, Shen M, Xu L. Efficacy and safety of direct oral anticoagulants in patients with diabetes and nonvalvular atrial fibrillation: meta-analysis of observational studies. Cardiovasc Ther. 2021;2021:5520027.CrossRef Cao B, Yao X, Zhang L, Hu X, Chen M, Shen M, Xu L. Efficacy and safety of direct oral anticoagulants in patients with diabetes and nonvalvular atrial fibrillation: meta-analysis of observational studies. Cardiovasc Ther. 2021;2021:5520027.CrossRef
89.
Zurück zum Zitat Huang HK, Liu PP, Lin SM, Hsu JY, Yeh JI, Lai EC, Peng CC, Munir KM, Loh CH, Tu YK. Diabetes-related complications and mortality in patients with atrial fibrillation receiving different oral anticoagulants: a nationwide analysis. Ann Intern Med. 2022;175:490–8.CrossRef Huang HK, Liu PP, Lin SM, Hsu JY, Yeh JI, Lai EC, Peng CC, Munir KM, Loh CH, Tu YK. Diabetes-related complications and mortality in patients with atrial fibrillation receiving different oral anticoagulants: a nationwide analysis. Ann Intern Med. 2022;175:490–8.CrossRef
90.
Zurück zum Zitat Kumar S, Lim E, Covic A, Verhamme P, Gale CP, Camm AJ, Goldsmith D. Anticoagulation in concomitant chronic kidney disease and atrial fibrillation: JACC review topic of the week. J Am Coll Cardiol. 2019;74:2204–15.CrossRef Kumar S, Lim E, Covic A, Verhamme P, Gale CP, Camm AJ, Goldsmith D. Anticoagulation in concomitant chronic kidney disease and atrial fibrillation: JACC review topic of the week. J Am Coll Cardiol. 2019;74:2204–15.CrossRef
91.
Zurück zum Zitat Weitz JI, Prandoni P, Verhamme P. Anticoagulation for patients with venous thromboembolism: when is extended treatment required? TH Open. 2020;4:e446–56.CrossRef Weitz JI, Prandoni P, Verhamme P. Anticoagulation for patients with venous thromboembolism: when is extended treatment required? TH Open. 2020;4:e446–56.CrossRef
92.
Zurück zum Zitat Catella J, Bertoletti L, Mismetti P, et al. Severe renal impairment and risk of bleeding during anticoagulation for venous thromboembolism. J Thromb Haemost. 2020;18:1728–37.CrossRef Catella J, Bertoletti L, Mismetti P, et al. Severe renal impairment and risk of bleeding during anticoagulation for venous thromboembolism. J Thromb Haemost. 2020;18:1728–37.CrossRef
93.
Zurück zum Zitat Ando G, Capranzano P. Non-vitamin K antagonist oral anticoagulants in atrial fibrillation patients with chronic kidney disease: a systematic review and network meta-analysis. Int J Cardiol. 2017;231:162–9.CrossRef Ando G, Capranzano P. Non-vitamin K antagonist oral anticoagulants in atrial fibrillation patients with chronic kidney disease: a systematic review and network meta-analysis. Int J Cardiol. 2017;231:162–9.CrossRef
94.
Zurück zum Zitat Cheung CYS, Parikh J, Farrell A, Lefebvre M, Summa-Sorgini C, Battistella M. Direct oral anticoagulant use in chronic kidney disease and dialysis patients with venous thromboembolism: a systematic review of thrombosis and bleeding outcomes. Ann Pharmacother. 2021;55:711–22.CrossRef Cheung CYS, Parikh J, Farrell A, Lefebvre M, Summa-Sorgini C, Battistella M. Direct oral anticoagulant use in chronic kidney disease and dialysis patients with venous thromboembolism: a systematic review of thrombosis and bleeding outcomes. Ann Pharmacother. 2021;55:711–22.CrossRef
96.
Zurück zum Zitat Sy J, Hsiung JT, Edgett D, Kalantar-Zadeh K, Streja E, Lau WL. Cardiovascular and bleeding outcomes with anticoagulants across kidney disease stages: analysis of a national US cohort. Am J Nephrol. 2021;52:199–208.CrossRef Sy J, Hsiung JT, Edgett D, Kalantar-Zadeh K, Streja E, Lau WL. Cardiovascular and bleeding outcomes with anticoagulants across kidney disease stages: analysis of a national US cohort. Am J Nephrol. 2021;52:199–208.CrossRef
97.
Zurück zum Zitat Chokesuwattanaskul R, Thongprayoon C, Tanawuttiwat T, Kaewput W, Pachariyanon P, Cheungpasitporn W. Comparative safety and efficacy of apixaban versus warfarin in patients with end stage renal disease: meta-analysis. Pacing Clin Electrophysiol. 2018;41:627–34.CrossRef Chokesuwattanaskul R, Thongprayoon C, Tanawuttiwat T, Kaewput W, Pachariyanon P, Cheungpasitporn W. Comparative safety and efficacy of apixaban versus warfarin in patients with end stage renal disease: meta-analysis. Pacing Clin Electrophysiol. 2018;41:627–34.CrossRef
98.
Zurück zum Zitat Malhotra K, Ishfaq MF, Goyal N, et al. Oral anticoagulation in patients with chronic kidney disease: a systematic review and meta-analysis. Neurology. 2019;92:e2421–31.CrossRef Malhotra K, Ishfaq MF, Goyal N, et al. Oral anticoagulation in patients with chronic kidney disease: a systematic review and meta-analysis. Neurology. 2019;92:e2421–31.CrossRef
99.
Zurück zum Zitat Ballestri S, Capitelli M, Fontana MC, Arioli D, Romagnoli E, Graziosi C, Lonardo A, Marietta M, Dentali F, Cioni G. Direct oral anticoagulants in patients with liver disease in the era of non-alcoholic fatty liver disease global epidemic: a narrative review. Adv Ther. 2020;37:1910–32. https://doi.org/10.1007/s12325-020-01307-z.CrossRef Ballestri S, Capitelli M, Fontana MC, Arioli D, Romagnoli E, Graziosi C, Lonardo A, Marietta M, Dentali F, Cioni G. Direct oral anticoagulants in patients with liver disease in the era of non-alcoholic fatty liver disease global epidemic: a narrative review. Adv Ther. 2020;37:1910–32. https://​doi.​org/​10.​1007/​s12325-020-01307-z.CrossRef
100.
Zurück zum Zitat Violi F, Loffredo L, Pastori D. Anticoagulation in patients with advanced liver disease: an open issue. Intern Emerg Med. 2021;16:61–71.CrossRef Violi F, Loffredo L, Pastori D. Anticoagulation in patients with advanced liver disease: an open issue. Intern Emerg Med. 2021;16:61–71.CrossRef
103.
Zurück zum Zitat Violi F, Vestri A, Menichelli D, Di Rocco A, Pastori D, Pignatelli P. Direct oral anticoagulants in patients with atrial fibrillation and advanced liver disease: an exploratory meta-analysis. Hepatol Commun. 2020;4:1034–40.CrossRef Violi F, Vestri A, Menichelli D, Di Rocco A, Pastori D, Pignatelli P. Direct oral anticoagulants in patients with atrial fibrillation and advanced liver disease: an exploratory meta-analysis. Hepatol Commun. 2020;4:1034–40.CrossRef
104.
Zurück zum Zitat Marietta M, Coluccio V, Luppi M. COVID-19, coagulopathy and venous thromboembolism: more questions than answers. Intern Emerg Med. 2020;15:1375–87.CrossRef Marietta M, Coluccio V, Luppi M. COVID-19, coagulopathy and venous thromboembolism: more questions than answers. Intern Emerg Med. 2020;15:1375–87.CrossRef
105.
Zurück zum Zitat Giannis D, Douketis JD, Spyropoulos AC. Anticoagulant therapy for COVID-19: what we have learned and what are the unanswered questions? Eur J Intern Med. 2022;96:13–6.CrossRef Giannis D, Douketis JD, Spyropoulos AC. Anticoagulant therapy for COVID-19: what we have learned and what are the unanswered questions? Eur J Intern Med. 2022;96:13–6.CrossRef
106.
Zurück zum Zitat Marietta M, Ageno W, Artoni A, De Candia E, Gresele P, Marchetti M, Marcucci R, Tripodi A. COVID-19 and haemostasis: a position paper from Italian Society on Thrombosis and Haemostasis (SISET). Blood Transfus. 2020;18:167–9. Marietta M, Ageno W, Artoni A, De Candia E, Gresele P, Marchetti M, Marcucci R, Tripodi A. COVID-19 and haemostasis: a position paper from Italian Society on Thrombosis and Haemostasis (SISET). Blood Transfus. 2020;18:167–9.
107.
Zurück zum Zitat Al-Samkari H, Connors JM. Managing the competing risks of thrombosis, bleeding, and anticoagulation in patients with malignancy. Blood Adv. 2019;3:3770–9.CrossRef Al-Samkari H, Connors JM. Managing the competing risks of thrombosis, bleeding, and anticoagulation in patients with malignancy. Blood Adv. 2019;3:3770–9.CrossRef
108.
Zurück zum Zitat Cosmi B. An update on the efficacy and safety of novel anticoagulants for cancer associated thrombosis. Expert Opin Pharmacother. 2021;22:583–94.CrossRef Cosmi B. An update on the efficacy and safety of novel anticoagulants for cancer associated thrombosis. Expert Opin Pharmacother. 2021;22:583–94.CrossRef
109.
Zurück zum Zitat Yun JP, Choi EK, Han KD, et al. Risk of atrial fibrillation according to cancer type: a nationwide population-based study. JACC CardioOncol. 2021;3:221–32.CrossRef Yun JP, Choi EK, Han KD, et al. Risk of atrial fibrillation according to cancer type: a nationwide population-based study. JACC CardioOncol. 2021;3:221–32.CrossRef
110.
Zurück zum Zitat Davis MK, Lim H, Lee AYY. Direct oral anticoagulants in patients with cancer and nonvalvular atrial fibrillation. JACC CardioOncol. 2021;3:425–7.CrossRef Davis MK, Lim H, Lee AYY. Direct oral anticoagulants in patients with cancer and nonvalvular atrial fibrillation. JACC CardioOncol. 2021;3:425–7.CrossRef
111.
Zurück zum Zitat Mariani MV, Magnocavallo M, Straito M, et al. Direct oral anticoagulants versus vitamin K antagonists in patients with atrial fibrillation and cancer a meta-analysis. J Thromb Thrombolysis. 2021;51:419–29.CrossRef Mariani MV, Magnocavallo M, Straito M, et al. Direct oral anticoagulants versus vitamin K antagonists in patients with atrial fibrillation and cancer a meta-analysis. J Thromb Thrombolysis. 2021;51:419–29.CrossRef
112.
Zurück zum Zitat Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest. 2016;149:315–52.CrossRef Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST guideline and expert panel report. Chest. 2016;149:315–52.CrossRef
113.
Zurück zum Zitat Ageno W, Donadini M. Breadth of complications of long-term oral anticoagulant care. Hematol Am Soc Hematol Educ Progr. 2018;2018:432–8.CrossRef Ageno W, Donadini M. Breadth of complications of long-term oral anticoagulant care. Hematol Am Soc Hematol Educ Progr. 2018;2018:432–8.CrossRef
114.
Zurück zum Zitat Clemens A, Strack A, Noack H, et al. Anticoagulant-related gastrointestinal bleeding—could this facilitate early detection of benign or malignant gastrointestinal lesions? Ann Med. 2014;46:672–8.CrossRef Clemens A, Strack A, Noack H, et al. Anticoagulant-related gastrointestinal bleeding—could this facilitate early detection of benign or malignant gastrointestinal lesions? Ann Med. 2014;46:672–8.CrossRef
116.
Zurück zum Zitat Raposeiras Roubín S, Abu Assi E, Barreiro Pardal C, et al. New cancer diagnosis after bleeding in anticoagulated patients with atrial fibrillation. J Am Heart Assoc. 2020;9: e016836.CrossRef Raposeiras Roubín S, Abu Assi E, Barreiro Pardal C, et al. New cancer diagnosis after bleeding in anticoagulated patients with atrial fibrillation. J Am Heart Assoc. 2020;9: e016836.CrossRef
117.
Zurück zum Zitat Beyth RJ, Quinn LM, Landefeld CS. Prospective evaluation of an index for predicting the risk of major bleeding in outpatients treated with warfarin. Am J Med. 1998;105:91–9.CrossRef Beyth RJ, Quinn LM, Landefeld CS. Prospective evaluation of an index for predicting the risk of major bleeding in outpatients treated with warfarin. Am J Med. 1998;105:91–9.CrossRef
119.
Zurück zum Zitat Fang MC, Go AS, Chang Y, et al. A new risk scheme to predict warfarin-associated hemorrhage: the ATRIA (Anticoagulation and Risk Factors in Atrial Fibrillation) study. J Am Coll Cardiol. 2011;58:395–401.CrossRef Fang MC, Go AS, Chang Y, et al. A new risk scheme to predict warfarin-associated hemorrhage: the ATRIA (Anticoagulation and Risk Factors in Atrial Fibrillation) study. J Am Coll Cardiol. 2011;58:395–401.CrossRef
120.
Zurück zum Zitat Pisters R, Lane DA, Nieuwlaat R, de Vos CB, Crijns HJ, Lip GY. A novel user-friendly score (HAS-BLED) to assess 1-year risk of major bleeding in patients with atrial fibrillation: the Euro Heart Survey. Chest. 2010;138:1093–100.CrossRef Pisters R, Lane DA, Nieuwlaat R, de Vos CB, Crijns HJ, Lip GY. A novel user-friendly score (HAS-BLED) to assess 1-year risk of major bleeding in patients with atrial fibrillation: the Euro Heart Survey. Chest. 2010;138:1093–100.CrossRef
121.
Zurück zum Zitat O’Brien EC, Simon DN, Thomas LE, et al. The ORBIT bleeding score: a simple bedside score to assess bleeding risk in atrial fibrillation. Eur Heart J. 2015;36:3258–64. O’Brien EC, Simon DN, Thomas LE, et al. The ORBIT bleeding score: a simple bedside score to assess bleeding risk in atrial fibrillation. Eur Heart J. 2015;36:3258–64.
122.
Zurück zum Zitat Hijazi Z, Oldgren J, Lindbäck J, et al. The novel biomarker-based ABC (age, biomarkers, clinical history)-bleeding risk score for patients with atrial fibrillation: a derivation and validation study. Lancet. 2016;387:2302–11.CrossRef Hijazi Z, Oldgren J, Lindbäck J, et al. The novel biomarker-based ABC (age, biomarkers, clinical history)-bleeding risk score for patients with atrial fibrillation: a derivation and validation study. Lancet. 2016;387:2302–11.CrossRef
123.
Zurück zum Zitat Barnett-Griness O, Stein N, Kotler A, Saliba W, Gronich N. Novel bleeding prediction model in atrial fibrillation patients on new oral anticoagulants. Heart. 2022;108:266–73.CrossRef Barnett-Griness O, Stein N, Kotler A, Saliba W, Gronich N. Novel bleeding prediction model in atrial fibrillation patients on new oral anticoagulants. Heart. 2022;108:266–73.CrossRef
124.
Zurück zum Zitat Nieuwenhuis HK, Albada J, Banga JD, Sixma JJ. Identification of risk factors for bleeding during treatment of acute venous thromboembolism with heparin or low molecular weight heparin. Blood. 1991;78:2337–43.CrossRef Nieuwenhuis HK, Albada J, Banga JD, Sixma JJ. Identification of risk factors for bleeding during treatment of acute venous thromboembolism with heparin or low molecular weight heparin. Blood. 1991;78:2337–43.CrossRef
125.
Zurück zum Zitat Kuijer PM, Hutten BA, Prins MH, Büller HR. Prediction of the risk of bleeding during anticoagulant treatment for venous thromboembolism. Arch Intern Med. 1999;159:457–60.CrossRef Kuijer PM, Hutten BA, Prins MH, Büller HR. Prediction of the risk of bleeding during anticoagulant treatment for venous thromboembolism. Arch Intern Med. 1999;159:457–60.CrossRef
126.
Zurück zum Zitat Ruíz-Giménez N, Suárez C, González R, et al. Predictive variables for major bleeding events in patients presenting with documented acute venous thromboembolism. Findings from the RIETE Registry. Thromb Haemost. 2008;100:26–31.CrossRef Ruíz-Giménez N, Suárez C, González R, et al. Predictive variables for major bleeding events in patients presenting with documented acute venous thromboembolism. Findings from the RIETE Registry. Thromb Haemost. 2008;100:26–31.CrossRef
127.
Zurück zum Zitat Kearon C, Akl EA, Comerota AJ, et al. Antithrombotic therapy for VTE disease: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141:e419S-e496S.CrossRef Kearon C, Akl EA, Comerota AJ, et al. Antithrombotic therapy for VTE disease: antithrombotic therapy and prevention of thrombosis, 9th ed: American College of Chest Physicians evidence-based clinical practice guidelines. Chest. 2012;141:e419S-e496S.CrossRef
128.
Zurück zum Zitat Klok FA, Hösel V, Clemens A, Yollo WD, Tilke C, Schulman S, Lankeit M, Konstantinides SV. Prediction of bleeding events in patients with venous thromboembolism on stable anticoagulation treatment. Eur Respir J. 2016;48:1369–76.CrossRef Klok FA, Hösel V, Clemens A, Yollo WD, Tilke C, Schulman S, Lankeit M, Konstantinides SV. Prediction of bleeding events in patients with venous thromboembolism on stable anticoagulation treatment. Eur Respir J. 2016;48:1369–76.CrossRef
129.
Zurück zum Zitat Klok FA, Huisman MV. How I assess and manage the risk of bleeding in patients treated for venous thromboembolism. Blood. 2020;135:724–34.CrossRef Klok FA, Huisman MV. How I assess and manage the risk of bleeding in patients treated for venous thromboembolism. Blood. 2020;135:724–34.CrossRef
130.
Zurück zum Zitat Marques Antunes M, Alves M, Pinto FJ, Agnelli G, Caldeira D. The high-risk bleeding category of different scores in patients with venous thromboembolism: systematic review and meta-analysis. Eur J Intern Med. 2021;94:45–55.CrossRef Marques Antunes M, Alves M, Pinto FJ, Agnelli G, Caldeira D. The high-risk bleeding category of different scores in patients with venous thromboembolism: systematic review and meta-analysis. Eur J Intern Med. 2021;94:45–55.CrossRef
131.
Zurück zum Zitat Hellenbart EL, Faulkenberg KD, Finks SW. Evaluation of bleeding in patients receiving direct oral anticoagulants. Vasc Health Risk Manag. 2017;13:325–42.CrossRef Hellenbart EL, Faulkenberg KD, Finks SW. Evaluation of bleeding in patients receiving direct oral anticoagulants. Vasc Health Risk Manag. 2017;13:325–42.CrossRef
132.
Zurück zum Zitat Tomaselli GF, Mahaffey KW, Cuker A, et al. 2020 ACC expert consensus decision pathway on management of bleeding in patients on oral anticoagulants: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2020;76:594–622.CrossRef Tomaselli GF, Mahaffey KW, Cuker A, et al. 2020 ACC expert consensus decision pathway on management of bleeding in patients on oral anticoagulants: a report of the American College of Cardiology Solution Set Oversight Committee. J Am Coll Cardiol. 2020;76:594–622.CrossRef
133.
Zurück zum Zitat Hemphill JC III, Greenberg SM, Anderson CS, Council on Clinical Cardiolog, et al. Guidelines for the management of spontaneous intracerebral hemorrhage: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2015;46:2032–60.CrossRef Hemphill JC III, Greenberg SM, Anderson CS, Council on Clinical Cardiolog, et al. Guidelines for the management of spontaneous intracerebral hemorrhage: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2015;46:2032–60.CrossRef
134.
Zurück zum Zitat Dionne JC, Oczkowski SJW, Hunt BJ, Antonelli M, Wijnberge M, Raasveld SJ, et al. Tranexamic acid in gastrointestinal bleeding: a systematic review and meta-analysis. Crit Care Med. 2022;50:e313–9.CrossRef Dionne JC, Oczkowski SJW, Hunt BJ, Antonelli M, Wijnberge M, Raasveld SJ, et al. Tranexamic acid in gastrointestinal bleeding: a systematic review and meta-analysis. Crit Care Med. 2022;50:e313–9.CrossRef
135.
Zurück zum Zitat King B, Milling T, Gajewski B, et al. Restarting and timing of oral anticoagulation after traumatic intracranial hemorrhage: a review and summary of ongoing and planned prospective randomized clinical trials. Trauma Surg Acute Care Open. 2020;5: e000605.CrossRef King B, Milling T, Gajewski B, et al. Restarting and timing of oral anticoagulation after traumatic intracranial hemorrhage: a review and summary of ongoing and planned prospective randomized clinical trials. Trauma Surg Acute Care Open. 2020;5: e000605.CrossRef
136.
Zurück zum Zitat Nielsen PB, Skjøth F, Søgaard M, et al. Non-vitamin K antagonist oral anticoagulants versus warfarin in atrial fibrillation patients with intracerebral hemorrhage. Stroke. 2019;50(4):939–46.CrossRef Nielsen PB, Skjøth F, Søgaard M, et al. Non-vitamin K antagonist oral anticoagulants versus warfarin in atrial fibrillation patients with intracerebral hemorrhage. Stroke. 2019;50(4):939–46.CrossRef
137.
Zurück zum Zitat Poli D, Antonucci E, Vignini E, et al. Anticoagulation resumption after intracranial hemorrhage in patients treated with VKA and DOACs. Eur J Intern Med. 2020;80:73–7.CrossRef Poli D, Antonucci E, Vignini E, et al. Anticoagulation resumption after intracranial hemorrhage in patients treated with VKA and DOACs. Eur J Intern Med. 2020;80:73–7.CrossRef
138.
Zurück zum Zitat Botto GL, Ameri P, De Caterina R. Many good reasons to switch from vitamin K antagonists to non-vitamin K antagonists in patients with non-valvular atrial fibrillation. J Clin Med. 2021;10:2866.CrossRef Botto GL, Ameri P, De Caterina R. Many good reasons to switch from vitamin K antagonists to non-vitamin K antagonists in patients with non-valvular atrial fibrillation. J Clin Med. 2021;10:2866.CrossRef
139.
Zurück zum Zitat Hawkes MA, Rabinstein AA. Anticoagulation for atrial fibrillation after intracranial hemorrhage: a systematic review. Neurol Clin Pract. 2018;8:48–57.CrossRef Hawkes MA, Rabinstein AA. Anticoagulation for atrial fibrillation after intracranial hemorrhage: a systematic review. Neurol Clin Pract. 2018;8:48–57.CrossRef
141.
Zurück zum Zitat Steiner T, Al-Shahi Salman R, Beer R, et al. European stroke organisation (ESO) guidelines for the management of spontaneous intracerebral hemorrhage. Int J Stroke. 2014;9:840–55.CrossRef Steiner T, Al-Shahi Salman R, Beer R, et al. European stroke organisation (ESO) guidelines for the management of spontaneous intracerebral hemorrhage. Int J Stroke. 2014;9:840–55.CrossRef
142.
Zurück zum Zitat Wiegele M, Schöchl H, Haushofer A, et al. Diagnostic and therapeutic approach in adult patients with traumatic brain injury receiving oral anticoagulant therapy: an Austrian interdisciplinary consensus statement. Crit Care. 2019;23:62.CrossRef Wiegele M, Schöchl H, Haushofer A, et al. Diagnostic and therapeutic approach in adult patients with traumatic brain injury receiving oral anticoagulant therapy: an Austrian interdisciplinary consensus statement. Crit Care. 2019;23:62.CrossRef
143.
Zurück zum Zitat Chai-Adisaksopha C, Hillis C, Monreal M, Witt DM, Crowther M. Thromboembolic events, recurrent bleeding and mortality after resuming anticoagulant following gastrointestinal bleeding. a meta-analysis. Thromb Haemost. 2015;114:819–25.CrossRef Chai-Adisaksopha C, Hillis C, Monreal M, Witt DM, Crowther M. Thromboembolic events, recurrent bleeding and mortality after resuming anticoagulant following gastrointestinal bleeding. a meta-analysis. Thromb Haemost. 2015;114:819–25.CrossRef
144.
Zurück zum Zitat Tapaskar N, Pang A, Werner DA, Sengupta N. Resuming anticoagulation following hospitalization for gastrointestinal bleeding is associated with reduced thromboembolic events and improved mortality: results from a systematic review and meta-analysis. Dig Dis Sci. 2021;66:554–66.CrossRef Tapaskar N, Pang A, Werner DA, Sengupta N. Resuming anticoagulation following hospitalization for gastrointestinal bleeding is associated with reduced thromboembolic events and improved mortality: results from a systematic review and meta-analysis. Dig Dis Sci. 2021;66:554–66.CrossRef
145.
Zurück zum Zitat Witt DM. What to do after the bleed: resuming anticoagulation after major bleeding. Hematol Am Soc Hematol Educ Progr. 2016;2016:620–4.CrossRef Witt DM. What to do after the bleed: resuming anticoagulation after major bleeding. Hematol Am Soc Hematol Educ Progr. 2016;2016:620–4.CrossRef
146.
Zurück zum Zitat Scott MJ, Veitch A, Thachil J. Reintroduction of anti-thrombotic therapy after a gastrointestinal haemorrhage: if and when? Br J Haematol. 2017;177:185–97.CrossRef Scott MJ, Veitch A, Thachil J. Reintroduction of anti-thrombotic therapy after a gastrointestinal haemorrhage: if and when? Br J Haematol. 2017;177:185–97.CrossRef
147.
Zurück zum Zitat Candeloro M, van Es N, Cantor N, et al. Recurrent bleeding and thrombotic events after resumption of oral anticoagulants following gastrointestinal bleeding: communication from the ISTH SSC Subcommittee on Control of Anticoagulation. J Thromb Haemost. 2021;19:2618–28.CrossRef Candeloro M, van Es N, Cantor N, et al. Recurrent bleeding and thrombotic events after resumption of oral anticoagulants following gastrointestinal bleeding: communication from the ISTH SSC Subcommittee on Control of Anticoagulation. J Thromb Haemost. 2021;19:2618–28.CrossRef
148.
Zurück zum Zitat Lip GYH, Lane DA. Matching the NOAC to the patient: remember the modifiable bleeding risk factors. J Am Coll Cardiol. 2015;66:2282–4.CrossRef Lip GYH, Lane DA. Matching the NOAC to the patient: remember the modifiable bleeding risk factors. J Am Coll Cardiol. 2015;66:2282–4.CrossRef
149.
Zurück zum Zitat Schaefer JK, McBane RD, Wysokinski WE. How to choose appropriate direct oral anticoagulant for patient with nonvalvular atrial fibrillation. Ann Hematol. 2016;95:437–49.CrossRef Schaefer JK, McBane RD, Wysokinski WE. How to choose appropriate direct oral anticoagulant for patient with nonvalvular atrial fibrillation. Ann Hematol. 2016;95:437–49.CrossRef
150.
Zurück zum Zitat Diener HC, Aisenberg J, Ansell J, et al. Choosing a particular oral anticoagulant and dose for stroke prevention in individual patients with non-valvular atrial fibrillation: part 2. Eur Heart J. 2017;38:860–8. Diener HC, Aisenberg J, Ansell J, et al. Choosing a particular oral anticoagulant and dose for stroke prevention in individual patients with non-valvular atrial fibrillation: part 2. Eur Heart J. 2017;38:860–8.
151.
Zurück zum Zitat Geldhof V, Vandenbriele C, Verhamme P, Vanassche T. Venous thromboembolism in the elderly: efficacy and safety of non-VKA oral anticoagulants. Thromb J. 2014;13(12):21.CrossRef Geldhof V, Vandenbriele C, Verhamme P, Vanassche T. Venous thromboembolism in the elderly: efficacy and safety of non-VKA oral anticoagulants. Thromb J. 2014;13(12):21.CrossRef
152.
Zurück zum Zitat Huisman MV, Ferreira M, Feuring M, Fraessdorf M, Klok FA. Less abnormal uterine bleeding with dabigatran than warfarin in women treated for acute venous thromboembolism. J Thromb Haemost. 2018;16:1775–8.CrossRef Huisman MV, Ferreira M, Feuring M, Fraessdorf M, Klok FA. Less abnormal uterine bleeding with dabigatran than warfarin in women treated for acute venous thromboembolism. J Thromb Haemost. 2018;16:1775–8.CrossRef
Metadaten
Titel
Risk and Management of Bleeding Complications with Direct Oral Anticoagulants in Patients with Atrial Fibrillation and Venous Thromboembolism: a Narrative Review
verfasst von
Stefano Ballestri
Elisa Romagnoli
Dimitriy Arioli
Valeria Coluccio
Alessandra Marrazzo
Afroditi Athanasiou
Maria Di Girolamo
Cinzia Cappi
Marco Marietta
Mariano Capitelli
Publikationsdatum
16.10.2022
Verlag
Springer Healthcare
Erschienen in
Advances in Therapy / Ausgabe 1/2023
Print ISSN: 0741-238X
Elektronische ISSN: 1865-8652
DOI
https://doi.org/10.1007/s12325-022-02333-9

Weitere Artikel der Ausgabe 1/2023

Advances in Therapy 1/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.