Hamostaseologie 2020; 40(02): 184-200
DOI: 10.1055/a-1113-0655
Review Article
Georg Thieme Verlag KG Stuttgart · New York

Does One Dose Really Fit All? On the Monitoring of Direct Oral Anticoagulants: A Review of the Literature

Thomas Moner-Banet
1   Department of Internal Medicine, Riviera-Chablais Hospital, Rennaz, Switzerland
,
Lorenzo Alberio
2   Division of Haematology and Central Haematology Laboratory, CHUV Lausanne University Hospital, University of Lausanne (UNIL), Lausanne, Switzerland
,
Pierre-Alexandre Bart
3   Department of Internal Medicine, CHUV Lausanne University Hospital, University of Lausanne (UNIL), Lausanne, Switzerland
› Author Affiliations
Further Information

Address for correspondence

Lorenzo Alberio, MD
Division of Haematology and Central Haematology Laboratory, Centre Hospitalier Universitaire Vaudois (CHUV)
Rue du Bugnon 46, 1005 Lausanne
Switzerland   

Publication History

20 October 2019

06 February 2020

Publication Date:
26 May 2020 (online)

 

Abstract

Background There is an increasing amount of literature on direct oral anticoagulant (DOAC) laboratory monitoring. The aims of the present review were to evaluate published data on monitoring DOACs, to provide clinical guidance on how to interpret results, and to summarize why, when, and how to monitor DOACs.

Methods The publications screened for this review were obtained through a PubMed search for articles published in English or French before April 2019 that had the following as their main themes: DOAC monitoring, DOAC exposure–effect relationship, DOAC drug interactions, and pharmacokinetics and pharmacodynamics of DOACs.

Results DOACs show important inter- and intrapersonal concentration variability and a significant exposure–effect relationship. Concentrations out of the expected range have been shown to lead to an increased adverse event rate and a lower efficacy. No definitive therapeutic range exists for DOACs except for dabigatran for which trough levels of 40 to 200 ng/mL seem to be the consensus. Indications to monitor include suspected drug accumulation in special patient populations, suspected drug failure, and acute situations such as hemorrhagic or thrombotic events.

Conclusion There is a likely benefit to monitor DOACs in order to improve their safety and efficacy but randomized controlled trials are required to determine the therapeutic range of these drugs and evaluate whether DOAC monitoring can improve outcomes in a clinical setting.


#

Introduction

Since the 20th century, oral anticoagulation has been performed using vitamin K antagonists (VKAs), which require regular laboratory monitoring due to their unpredictable pharmacokinetics and pharmacodynamics. This sparked an interest for better molecules that would be simpler to use and less variable in their efficacy. A few years ago, the direct oral anticoagulants (DOACs) hit the market. Their key features are the simplified posology (marketed as “one dose fits all”) and the lack of a need for monitoring. Their place in the therapeutic arsenal is in rapid expansion as an increasing number of studies are conducted on different patient populations.

Since their approval, DOACs have been subject to controversy regarding whether or not they should be monitored; this debate continues to this day. Since “one dose” DOACs have shown similar, if not better, efficacy and safety as VKAs, little research has been performed to study the possible benefits of individual dose tailoring for these drugs. However, DOACs have been shown to display considerable variation in their plasma levels and out-of-target concentrations could lead to an increased risk of adverse events such as bleeding or thromboembolism.

Currently, it is still unclear whether DOACs should be monitored. Therefore, we propose in this article a review of the literature on this topic, showing if, when, and how DOACs should be monitored.


#

Research Strategy

The publications screened for this review were obtained through a PubMed search for articles published in English or in French before April 2019 that had the following as their main themes: DOAC monitoring, DOAC exposure–effect relationship, DOAC drug interactions, and pharmacokinetics and pharmacodynamics of DOACs.

The following keywords were used either alone or in combinations: “direct oral anticoagulant,” “DOAC,” “novel oral anticoagulant,” “NACO,” “Dabigatran,” “Rivaroxaban,” “Apixaban,” “Edoxaban,” “monitoring,” “concentration,” “plasma level,” “laboratory,” “safety,” “bleeding,” “ischemic event,” “stroke,” “thromboembolism,” “therapeutic range,” “interaction,” and “dose tailoring.”

The articles were compiled, reviewed, and selected if they presented either a review of the literature, a meta-analysis, or original clinical data on DOAC monitoring. Then their bibliographies were reviewed and articles were selected among them following the same principles.

Official product information and European Medicines Agency and U.S. Food and Drug Administration (FDA) documentation for mentioned DOACs were also reviewed.


#

Why Should We Monitor DOACs?

DOACs meet most of the usual criteria for requiring some form of dose tailoring or therapeutic drug monitoring.[1] They notably show important intra- and interpersonal variability in concentrations and new data suggest that out-of-target concentrations are linked to more frequent adverse events. DOAC characteristics are summarized in [Table 1].

Table 1

Summary of DOAC characteristics

DOAC

Dabigatran (Pradaxa)

Rivaroxaban (Xarelto)

Apixaban (Eliquis)

Edoxaban (Lixiana)

References

[12] [13] [96] [108] [109]

[22] [23] [24] [105] [110] [111]

[41] [42] [112] [113]

[48] [114] [115] [116] [117]

Target

Factor IIa

Factor Xa

Factor Xa

Factor Xa

Approved indications

VTE treatment

Stroke prevention in AF (with restrictions)

VTE prevention in orthopaedics

VTE treatment

Stroke prevention in AF

VTE prevention in orthopaedics

VTE treatment

Stroke prevention in AF

VTE treatment

Stroke prevention in AF

Posology

VTE prophylaxis

10 mg qd

2.5 mg bid

VTE treatment

150 mg bid

15 mg bid for 3 wk then 20 mg qd

10 mg bid for 7 d then 5 mg bid

60 mg qd

Stroke prevention

150 mg bid

20 mg qd

5 mg bid

60 mg qd

Posology adaptation

110 mg bid if GFR 30–49 mL/min or >80 y old

15 mg od if GFR < 49 mL/min

2.5 mg bid if 2/3:

• > 80 years old

• < 60 kg

• Cr >133 µmol/L

30 mg od if GFR 15–49 mL/min or <60 kg or P-gp inhibitor

Not indicated

GFR < 30 mL/min

Child A–C cirrhosis

GFR < 15 mL/min

Child C cirrhosis

Should be used with caution if GFR < 30 mL/min

GFR < 15 mL/min

Child C cirrhosis

GFR < 15 mL/min

Child C cirrhosis

Bioavailability

3–7%

80–100%

50%

62%

Protein fixation

35%

95%

87%

40–60%

Time to peak

0.5–2 h

2–4 h

3–4 h

0.5–3 h

Metabolism

UGT (20%)

CYP 3A4/3A5/2J2

CYP 3A4/3A5

CYP 3A4 (minimal)

Elimination

80% renal, active form

20% renal and biliary, metabolites

36% renal, active form

32% renal metabolites

32% biliary metabolites

30% renal, active form

45% biliary, active form

25% renal and biliary metabolites

50% renal, active form

40% biliary, active form

10% renal and biliary metabolites

Half-life

12–14 h

5–13 h

8–15 h

10–14 h

Abbreviations: AF, atrial fibrillation; bid, twice daily; DOAC, direct oral anticoagulant; GFR, glomerular filtration rate; od, once a day; PE, pulmonary embolism; qd, every day; VTE, venous thromboembolism.


The absence of initial routine monitoring guidelines from licensing authorities seems in part to be the result of direct comparison to VKA instead of safety and efficacy optimization. The consensus being that monitoring would be superfluous since “one dose” DOACs have shown noninferior, if not better, efficacy and safety as VKAs.[2] [3] [4] [5]

In the next sections, we will review the available data on concentration variability and exposure–effect relationship for each DOAC.


#

Dabigatran

The RE-LY trial,[2] a study that compared two dabigatran doses with warfarin in atrial fibrillation (AF), was further analyzed by Reilly et al.[6] They evaluated the rate of bleeding events in relation to dabigatran trough levels. They found that the trough levels of dabigatran were subject to important variations with a fivefold increase between the 10th and 90th percentiles.

Age and creatinine clearance were the principal modifiers. The variable bioavailability of dabigatran (3–7%) could also be implicated, as suggested by Powell.[7] Genetic single nucleotide polymorphism (SNP) variants might also increase variability, as discussed further in this article.[8]

Reilly et al. showed a 55% trough level increase in patients with major bleeding events. The rate of ischemic events was increased among patients with low trough concentrations. The authors concluded that there was no optimal dose that would work for all patients and that a subset of patients could benefit from individualized dose tailoring by monitoring of dabigatran trough concentrations.

Boehringer Ingelheim, the manufacturer of dabigatran, was accused of withholding information regarding the potential benefits of monitoring dabigatran. A British Medical Journal (BMJ) investigation revealed that during U.S. litigation the company was forced to release internal correspondence and documentation.[9] The BMJ reported that in these documents an optimal benefit–risk ratio within 40 to 215 ng/mL for dabigatran trough concentrations was indicated. Moreover, in these internal communications concerns were voiced about not being able to defend the “no monitoring” attitude to health authorities after Reilly et al.'s study.

A Boehringer Ingelheim internal simulation study of the RE-LY data with dose tailoring showed a better safety profile if patients treated by dabigatran 150 mg with trough levels of >90 ng/mL after 1 week had a dose switch to 110 or 75 mg/day.[10] This dose titration showed comparable ischemic event rates versus no titration (risk ratio [RR] = 1.06, confidence interval [CI] 90%: 0.76–1.5), but the risk of major bleeding was significantly reduced (RR = 0.8, CI 90%: 0.66–0.97). These data were not shared with regulation authorities. As early as 2015, Safouris et al. studied the idea of an algorithm to adjust dabigatran doses first in regard to the patient characteristics and then depending on a plasma concentration measurement made in certain situations.[11] They suggested that doses should be adjusted if trough concentrations were out of the 48 to 200 ng/mL range.

Pradaxa FDA approval papers directly mention the relationship between trough levels and bleeding risk using data from the RE-LY trial.[12] Even with these data, the FDA did not, at the time, require any form of routine monitoring as their conclusions were that the 110 and 150 mg doses were respectively better and equivalent to warfarin in terms of bleeding risk.[2] [12] Product information of dabigatran presents thresholds for increased bleeding risk (trough levels >200 ng/mL in therapeutic indications, or >2 times the upper limit of normal activated partial thromboplastin time).[13]

A study by Šinigoj et al. (n = 44) showed that bleeding patients had significantly higher dabigatran trough levels (93 ± 36 vs. 72 ± 62 ng/mL, p = 0.02); however, they found no association between peak levels and bleeding risk.[14] Albaladejo et al. in a study including patients hospitalized for bleeding events while taking DOACs found that the median concentration of dabigatran was 162 ng/mL (range: 3–3,500 ng/mL; median time of measurement: 8.5 hours (0.8–29 hours) after the last dose, n = 123), which is a value in the peak range measured at almost trough time.[15] Volbers et al. found in a study measuring dabigatran levels in acute cerebrovascular events (n = 19) that patients with intracranial hemorrhage had significantly higher plasma concentrations in comparison to patients with an acute ischemic event (p < 0.05).[16] The BISTRO II (n = 351) randomized trial, which compared enoxaparin to dabigatran for orthopaedic prophylaxis showed a correlation between dabigatran peak concentrations and deep venous thrombosis and bleeding events using a logistic regression analysis. They did not analyze trough levels.[17] Testa et al. (n = 565) associated patients with low dabigatran trough levels and high CHA2DS2-VASc scores with thromboembolic events.[18] Chin et al. in a simulation study using data from the RE-LY trial and Reilly et al.'s follow-up study estimated that a trough concentration of 30 to 130 ng/mL was optimal and proposed the thrombin time (TT) as a screening assay (trough TT target of 130–300 seconds, with a normal range in their laboratory of 18–28 seconds).[19] Recently Chaussade et al. associated trough levels of >243.9 ng/mL to higher bleeding rates in a prospective study realized in a geriatric setting. This cutoff had a sensitivity of 54% and a specificity of 98% (n = 68). They found that patients who bled during the 1-year follow-up had higher dabigatran trough and peak levels (p = 0.01 for both).[20]

Another substudy of the RE-LY trial showed that some SNPs had an impact on bleeding risk. One of which had 32% prevalence in the white European ancestry population and significantly reduced dabigatran exposure and bleeding risk; the authors suggested genetic screening before treatment in such patients.[8] Another approach could be to use laboratory monitoring with dose-tailoring strategies in these patients.

Overall, these studies demonstrate that out-of-expected range concentrations of dabigatran can lead to a higher adverse event rate and that there is potential for some form of therapeutic drug monitoring to improve outcomes for patients.


#

Rivaroxaban

Despite its rather constant bioavailability, rivaroxaban also exhibits multiple-fold plasma trough and peak level variations.[21] [22] [23] [24] Gulilat et al. even reported 60-fold variations in random plasma levels (n = 94).[25] Genetic variants have been identified that could explain part of this important variability.[26]

The dose-exposure effect of rivaroxaban is well known, with higher doses leading to significantly more bleeding.[27] The FDA approval papers for Xarelto mention an increased bleeding risk as the area under the curve (AUC) increases, a twofold increase resulting in 50% more major bleeds.[23]

A Japanese study by Nakano et al. analyzing prothrombin time (PT) measurements made with the HemosIL RecombiPlasTin assay (normal range: 8–12 seconds) found an increased risk of bleeding in patients with peak PT >20 seconds (62.5 vs. 22.7%, p = 0.022). In this study, the trough PT was not associated with bleeding events (likely due to the lack of sensitivity of PT).[28] Woodruff et al. compared PT levels measured within 24 hours of taking rivaroxaban in a retrospective study (n = 199) and were able to conclude that patients with PT >30 seconds (twice the upper limit of the normal PT of the assay used in the study) had a threefold higher bleeding risk compared with those with PT <30 seconds (p = 0.006).[29] In a recent study involving 94 patients, Sakaguchi et al. showed that the rivaroxaban peak concentration was an independent predictive bleeding risk factor (p = 0.012). Trough levels were not associated with bleeding in this study.[30] In a prospective study (n = 156), Wada et al. showed that the rivaroxaban peak concentration was an independent predictive variable for the risk of bleeding (p < 0.01). In the same study, higher trough levels showed a nonsignificant trend toward higher bleeding risk.[31] Albaladejo et al. in a study including patients taking rivaroxaban hospitalized for bleeding events (n = 285) found that the median concentration of rivaroxaban was 124 ng/mL (range: 0–1,245; median time of measurement: 14 hours [0.2–62 hours] after the last dose), which is a high value for such timing.[15] Krause et al. were able to show in a prospective study involving 212 young venous thromboembolism patients that a weight-adjusted dose (mg/kg) was correlated with bleeding rates (p < 0.01) and found that bleeders showed a trend toward higher trough levels (p = 0.08).[32]

A study performed by Seiffge et al. on stroke patients (n = 241) did not find a difference between the plasma levels of ischemic and hemorrhagic stroke patients at the time of the event.[33] This may be due to the fact that they included patients who had peak concentrations as well as patients who had trough concentrations. Similarly, in a small study (n = 23), Zalewski et al. found no relation between trough levels and bleeding rates.[34]

Much circumstantial data on the rivaroxaban exposure–effect relationship come from case reports.[35] [36] However, case reports have also shown that despite overdoses of rivaroxaban and apixaban (showing very high anti-Xa activity), sometimes no bleeding occurred.[37] [38] An observational study reporting rivaroxaban and apixaban overdoses described a 7% bleeding rate; although anti-Xa values were not measured, these data support the fact that a high plasma level does not necessarily lead to a bleeding episode.[39] This may suggest that the anticoagulant effect of a given rivaroxaban concentration varies as a function of the individual prothrombotic state and that acutely elevated rivaroxaban trough levels are less predictive of bleeding than long-lasting overexposure.

These studies seem to show that rivaroxaban concentrations can affect outcomes such as bleeding or thrombosis and that interindividual variation is to be expected regarding the efficacy of the drug. However, evidence is less clear than for dabigatran and data on possible therapeutic ranges are still lacking. Peak levels seemed to be more correlated to adverse events than trough levels, which could be due to the low sample size and small effect size of variations in trough levels in the mentioned studies.


#

Apixaban

Apixaban trough levels vary significantly, as for dabigatran and rivaroxaban.[40] Early pharmacokinetics studies showed a variance of around 30% for the AUC of apixaban concentration, which is less variable than other DOACs.[41] Eliquis product information shows a four- to sixfold variation in trough levels across most dosages but mentions that no clinically relevant information for a single patient can be extrapolated from these data alone.[42] Gulilat et al. reported a 50-fold variation in random plasma levels.[25] Ueshima et al. found in a genomic study involving Japanese patients that there were genotypes that could affect significantly apixaban plasma levels.[43]

Apixaban FDA approval papers show that increased AUCs at a steady state lead to higher bleeding rates (p = 0.02). A twofold increase in AUC meant that the risk of major bleeding over 1 year increased from 1.79 to 3.11% (70% increase).[44] In a prospective study involving 169 patients taking apixaban, Wada et al. showed that both trough and peak levels were independent predictors of bleeding risk (p < 0.01).[31] Bhagirath et al. using data from the AVERROES trial found a significant correlation between apixaban trough levels and minor bleeding (p < 0.01) and that patients in the lowest trough level decile had an increased stroke risk (p = 0.013).[45] In a study of patients taking DOACs hospitalized for bleeding events (n = 34), Albaladejo et al. found that the median concentration of apixaban was 111 ng/mL (range: 18–537; median time of measurement 11 hours [2.6–87 hours] after the last dose), which is a highly elevated value at almost trough time.[15] However, in a simulation study using data from clinical trials and analyzing the exposure–response relation for apixaban, Byon et al. found no significant relation between trough levels and bleeding events.[46]

Overall, the data for apixaban also seem to suggest that a significant exposure–effect relationship exists and that dose tailoring could help improve outcomes.


#

Edoxaban

Edoxaban is also known for multiple-fold inter- and intrapersonal variability.[47]

Edoxaban FDA approval papers specifically mention relationship between trough levels, renal function, and bleeding risk, a twofold increase in trough levels meaning a doubling of bleeding rates.[48]

Weitz et al. in a phase II study (n = 1146) that compared edoxaban to warfarin in AF were able to show that increased exposure was correlated with bleeding rates, with trough levels being the most predictive parameter (p = 0.01 for major bleeds, as calculated by Giugliano).[49] [50] Salazar et al. showed in an exposure–outcome modeling analysis of phase I and II trials of edoxaban that plasma levels seemed to be correlated to bleeding, trough levels being again the most predictive factor (p < 0.001).[51] Ruff et al. in a subanalysis of the ENGAGE trial, comparing edoxaban to warfarin, showed that there was a similar safety with dose-tailored edoxaban and classic warfarin therapy. The dose tailoring was done on clinical factors alone. In this study higher trough concentrations led to more bleeding events and lower trough concentrations were linked to increased thromboembolic events.[52] Yin et al. also analyzed data from the ENGAGE trial and found that a higher percentage of inhibition of endogenous factor Xa (FXa) activity (obtained using a Russell's viper venom test) was correlated with both major bleeding and thromboembolic and stroke events when levels of inhibition were high and low, respectively (p < 0.001). They also identified a threshold at which the inhibition of FXa activity was capped (440 ng/mL).[53] Chao et al. compared Asians to non-Asians in regard to the safety profile of edoxaban in the ENGAGE trial and found that there was a better safety profile for Asians, which they explained with lower mean trough levels.[54]

In summary, the exposure–effect relationship for edoxaban seems well defined with trough levels being the most predictive parameter but precise therapeutic intervals are still lacking.


#

Is There a Known Therapeutic Range for DOACs?

Although many studies have measured peak and trough levels while on treatment, a therapeutic range cannot be extrapolated from that data alone. We summarized observed plasma concentration data for multiples indications for each DOAC in [Table 2].

Table 2

Observed peak and trough concentrations for different indications and DOACs

DOAC

Population

Peak levels

Ref.

Trough levels

Ref.

Dabigatran 150 mg bid

Stroke prevention in patients with AF

175 (74, 383)[a]

184 (64, 443)[b]

159 (±83)[c]

[6]

[104]

[14]

91 (40, 215)[a]

90 (31, 225)[b]

69 (±40)[c]

[6]

[104]

[14]

Dabigatran 110 mg bid

Stroke prevention in patients with AF and GFR 30–49 mL/min or >80 y old

187 (±122)[c]

[14]

90 (±71)[c]

[14]

Dabigatran 150 mg bid

Treatment of DVT/PE

175 (117, 275)[d]

[118]

60 (39, 95)[d]

[118]

Rivaroxaban 20 mg qd

Treatment of DVT/PE

270 (189, 419)[e]

215 (22, 535)[a]

[22]

[105]

26 (6, 87)[e]

32 (6, 239)[a]

[22]

[105]

Rivaroxaban 20 mg qd

Stroke prevention in patients with AF

249 (184, 343)[e]

[22]

44 (12, 137)[e]

[22]

Rivaroxaban 15 mg qd

Stroke prevention in patients with AF and GFR 30–49 mL/min

229 (178, 313)[a]

[22]

57 (18, 136)[a]

[22]

Rivaroxaban 10 mg qd

VTE prevention after orthopaedic surgery

125 (91, 196)[b]

101 (7, 273)[a]

149 (108, 209)[b]

[22]

[105]

[21]

9 (1, 38)[b]

14 (4, 51)[a]

17 (8, 50)[b]

[22]

[105]

[21]

Apixaban 10 mg bid

Treatment of DVT/PE for the first 7 d

251 (111, 572)[b]

[42]

120 (41, 335)[b]

[42]

Apixaban 5 mg bid

Treatment of DVT/PE after 7 d

132 (59, 302)[b]

[42]

63 (22, 177)[b]

[42]

Apixaban 5 mg bid

Stroke prevention in patients with AF

171 (91, 321)[b]

[42]

103 (41, 230)[b]

[42]

Apixaban 2.5 mg bid

VTE prevention in elective orthopaedic surgery

77 (41, 146)[b]

[42]

51 (23, 109)[b]

[42]

Apixaban 2.5 mg bid

Stroke prevention in patients with AF if 2/3:

• 80 years old

• < 60 kg

• Creatinine > 133 µmol/L

123 (69, 221)[b]

[42]

79 (34, 162)[b]

[42]

Apixaban 2.5 mg bid

Prevention of DVT/PE after 6 mo of therapeutic treatment

67 (30, 153)[b]

[42]

32 (11, 90)[b]

[42]

Edoxaban 60 mg qd

Treatment of DVT/PE

234 (149, 317)[d]

[107]

19 (10, 39)[d]

[107]

Edoxaban 60 mg qd

Stroke prevention in patients with AF

170 (120, 245)[f]

301 (60, 569)[g]

[49]

[47]

36 (19, 62)[d]

39 (13, 110)[g]

[52]

[47]

Edoxaban 30 mg qd

Treatment of DVT/PE in patients with >1/3:

• GFR 30–49 mL/min

• ≤ 60 kg

• p-gp inhibitor

164 (99, 225)[d]

[107]

16 (8, 32)[d]

[107]

Edoxaban 30 mg qd

Stroke prevention in patients with AF with >1/3:

• GFR 30–49 mL/min

• ≤ 60 kg

• p-gp inhibitor

85 (55, 115)[f]

169 (10, 400)[g]

[49]

[47]

27 (15, 45)[d]

38 (7, 147)[g]

[52]

[47]

Abbreviations: AF, atrial fibrillation; bid, twice daily; DOAC, direct oral anticoagulant; DVT, deep venous thrombosis; GFR, glomerular filtration rate; qd, every day; VTE, venous thromboembolism.


a Mean (10th, 90th percentiles).


b Median (5th, 95th percentiles).


c Mean (SD).


d Median (IQR).


e Mean (5th, 95th percentiles).


f Median (1.5 × IQR), obtained from box plots.


g Mean (min, max).


Data suggest that there could be an optimal risk–benefit range in which DOAC concentrations can be stabilized. However, no study has validated a therapeutic interval for any DOAC as of yet and the precise levels for each DOAC at which the risk of thromboembolism or bleeding increases are not yet known. [Fig. 1] summarizes cut-off values and suggested therapeutic ranges, if they exist. It is worth noting that the quality of the data used to create these ranges is low and that care should be taken when interpreting results.

Zoom Image
Fig. 1 Proposed DOACs through level interpretation. In green: estimated median concentration target. In yellow: estimated target range. In orange: increased risk of adverse event. In red: highly increased risk of adverse event. DOAC, direct oral anticoagulant.

#

Methods of Monitoring

Standard DOAC monitoring is not easy considering that clinicians are not as familiar with it as with VKA and international normalized ratio. Most easily accessible laboratory assays are unspecific and/or unable to cover the whole range of concentrations that would be needed for efficient monitoring and more specialized assays (anti-IIa and anti-Xa assays) are only available in some laboratories today.

[Table 3] summarizes current monitoring guidelines for DOACs and gives recommendations for interpretation of standard coagulation tests.

Table 3

DOAC monitoring methods (based on various studies[119] [120] [121] [122])

Coagulation test

Dabigatran

Rivaroxaban

Apixaban

Edoxaban

PT

Less sensitive than APTT. Not usable

Normal PT likely excludes excess plasma concentrations but does not exclude concentrations within the therapeutic range. Variable between laboratories

Not sensitive enough to be used

Normal PT likely excludes excess plasma concentrations but does not exclude concentrations within the therapeutic range.Variable between laboratories

INR

International sensitivity ratio not available for dabigatran. Not usable

International sensitivity ratio not available for rivaroxaban. Not usable

International sensitivity ratio not available for apixaban. Not usable

International sensitivity ratio not available for edoxaban. Not usable

APTT

Normal APTT likely excludes excess plasma concentrations but does not exclude concentrations within the therapeutic range. Variable between laboratories

Less sensitive and more variable than PT

Less sensitive than PT. Not usable

Less sensitive than PT. Not usable

TT

Normal TT excludes clinically relevant dabigatran presence. Too sensitive for quantitative measurement

Anti-Xa (calibrated)

Sensitive, linear across clinical ranges. Quantitative assay

Sensitive, linear across clinical ranges. Quantitative assay

Sensitive, linear across clinical range. Quantitative assay

Anti-IIa (calibrated)

Sensitive, linear across clinical ranges. Quantitative assay

Recommendation

TT to exclude drug presence. Calibrated Anti-IIa for quantitative measurement

PT to exclude excess drug concentration. Calibrated Anti-Xa for quantitative measurement

Calibrated anti-Xa for quantitative measurement

PT to exclude excess drug concentration. Calibrated anti-Xa for quantitative measurement

A normal PT cannot exclude the presence of anti-Xa DOACs

Abbreviations: APTT, activated partial thromboplastin time; DOAC, direct oral anticoagulant; INR, international normalized ratio; PT, prothrombin time; TT, thrombin time.



#

Indications for Monitoring

While the lack of data for the clinical outcomes of monitoring is an obstacle to clear indications, guidelines for laboratory and clinical monitoring in specific situations are appearing.[55] [56] In the next section we address which patient populations should be considered for monitoring.


#

Off-Label Doses

Studies have shown that approximately 25% of patients on DOACs are inappropriately dosed.[57] [58] This is likely due to factors outside approved dosing criteria, making clinicians cautious to prescribe the full dosage and perhaps a bias in the perception of bleeding risk, which is overestimated by clinicians.[59] [60] Underdosing was shown to increase thromboembolic event and stroke rates.[61] Similarly, inappropriately highly dosed patients had a higher bleeding rate.[62]

While some instances of these inappropriately dosed patients are likely due to lack of awareness of the recommendations for DOAC prescription, patients with unique characteristics for whom the risk–benefit ratio of various doses is uncertain are potential targets for DOAC monitoring to confirm the maintained efficacy of an off-label dose.


#

Renal Insufficiency

All DOACs are at least partially eliminated by the kidneys, and the low glomerular filtration rate (GFR) has been associated with higher AUCs and higher bleeding rates across most dosages for most DOACs, thus justifying the use of adjusted doses in patients with <50 mL/min GFR.[63] Patients with chronic kidney disease (CKD), notably due to uremic toxins, are at a higher risk of bleeding and of thromboembolic events.[64] This increases the risk of adverse events when out-of-target range excursions of plasma concentrations of DOAC occur, possibly justifying a closer monitoring of their plasma levels to ensure optimal safety.

DOACs showed a similar, if not better, efficacy and safety profile in renal failure patients when compared with VKA.[65] [66] [67] Rivaroxaban also showed lower rates of stroke and systemic embolism without a change in bleeding rates in patients with at least one episode of >20% decrease in renal function during follow-up in a re-analysis of the ROCKET-AF trial which compared it to warfarin.[68]

Despite initial and once per year renal function work-up, renal function can often deteriorate acutely and lead to retention of DOACs, especially in elderly patients. This is confirmed by a recent study that associated variation of renal function over time and major bleeding events in DOAC-treated AF patients.[69] Therefore, patients with suspected or confirmed acute impaired renal function should be considered for DOAC laboratory monitoring.

Overall, while patients with CKD could benefit from plasma level monitoring, studies still need to be conducted to attest the usefulness of such monitoring.


#

Elderly Patients

Old age was associated with more adverse events in DOAC trials. However, DOACs still mostly presented a similar or better safety profile in the elderly when compared with warfarin.[70] [71] [72] Lower rates of bleeding were observed in >75-year-old patients taking reduced dose edoxaban in the ENGAGE trial, showing the impact of dose reduction.[70] Dabigatran 150 twice daily (bid) and rivaroxaban 20 mg once daily were shown to increase risk of major bleeding when compared with warfarin in the elderly but older elderly patients (>75 years old) were those who benefited the most from the treatment (lowest number needed to treat).[73] Of note, the deterioration of renal function is highly correlated with age and could explain by itself the higher rates of bleeding in elderly patients.

A study performed by Bando et al. showed that there was no increased adverse event rates when compared with younger patients with adequate dose reduction of rivaroxaban in elderly patients with AF.[74] Nissan et al. showed that above-expected range trough and peak plasma levels of apixaban are more frequent in elderly patients, even on the right dosage.[75] In a study performed by Khan et al. on over 75-year-old AF patients taking DOACs, those with major bleeding also presented an acute deterioration of renal function. This underlines the importance of GFR monitoring in the elderly taking DOACs.[76] Finally, hypoalbuminemia, a frequent condition in the elderly, has also been associated with higher rates of bleeding in rivaroxaban-taking patients.[29]

Elderly patients are a fragile population in which bleeding events could be more frequent for those taking DOACs. Severity and consequences of bleeding events are likely to be worse in older patients due to frailty and comorbidities. DOAC concentration fluctuations out of the target range are likely to be more frequent due to GFR variation, hypoalbuminemia, and polymedication. This perhaps warrants some form of monitoring in this population to ensure an optimal safety profile. However, studies are needed to verify the usefulness of this approach and determine if old age is an independent risk factor for adverse events in the elderly taking DOACs.


#

Thromboembolic and Bleeding Events

A thrombotic or hemorrhagic event under well-conducted DOAC treatment should always raise the question of out-of-target drug levels versus real treatment failure (i.e., side effect occurring despite an adequate drug level). Monitoring should be performed in such circumstances in the emergency department especially now that antidotes are on the market. Plasma levels can help determine when these expensive drugs are really needed and help follow the reversal of the anticoagulation when they are given. A threshold of >50 ng/mL is usually used to consider an antidote administration in severely bleeding patients.[77] [78]


#

Dosing Errors

Dosing errors (overdoses most often) are good indications for DOAC monitoring. Although not all overdoses lead to bleeding and the necessity of reversing anticoagulation as mentioned before,[37] [38] knowing the drug levels can be very useful in the clinical setting.


#

Surgical Procedures and Emergency Invasive Procedures

DOACs should not be routinely interrupted before minor surgical procedures.[79] They should be stopped at least 2 to 3 days before any surgical procedure with a high bleeding risk or even sooner should renal function be altered and resumed 48 hours later. For low bleeding risk procedures, DOACs should be stopped 1 to 2 days before or sooner depending on renal function, DOACs should then be resumed after 24 hours.[80] [81] [82] Therefore, for the majority of elective patients, DOAC monitoring is not needed. However, in CKD patients and in patients taking interacting drugs, monitoring cold be useful.[83]

In cases where the appropriate stopping period cannot be easily assessed or followed, such as emergency procedures, drug monitoring can give critical information to the clinician. Guidelines exist regarding the thresholds that should be used[84] but little data support these values and a lot of debates exist regarding whether monitoring should be used while we lack precise cut-offs. A summary of existing thresholds can be found in [Table 4]. These values might be too conservative as they represent more or less the usual trough levels in healthy patients and very little data are available on the individual anticoagulant potential of a given concentration.[85] When possible, surgery should be delayed until DOAC activity in plasma is below these thresholds, otherwise reversal should be considered when the procedure cannot be delayed and plasma concentrations of DOACs are significantly higher than >30 ng/mL.[77] [78]

Table 4

Recommended thresholds for interventions and thrombolysis

Any DOAC

Erdoes et al. (2018)[123]

International consensus statement on the perioperative management of direct oral anticoagulants in cardiac surgery

<30 ng/mL is safe for high-risk cardiac operations

<50 ng/mL is the recommended threshold if the operation is urgent and has low bleeding risk

The authors suggest monitoring of DOACs in cardiac surgery with impaired renal or hepatic function or if bridging to heparins is needed

Dabigatran

Pernod et al. (2013)[84]

Recommendations of a French working group on perioperative hemostasis

<30 ng/mL to operate without increased bleeding risk

30–200 ng/mL: delay intervention if possible by 12 h then re-measure

200–400 ng/mL: delay intervention if possible by 12–24 h then re-measure

>400 ng/mL: high risk of hemorrhage, overdose

The 30 ng/mL threshold comes from the expected plasma concentrations with the stopping time of 24–72 h in the elective surgery protocol of the RELY trial. The other thresholds are expert opinions

Steiner et al. (2013)[124]

Recommendations for the emergency management of DOAC-related complications

<50 ng/mL to consider thrombolysis in ischemic stroke

<50 ng/mL to consider intervention in SAH patients

Level of evidence IV: expert opinion for both thresholds

Albaladejo et al. (2018)[78]

Management of bleeding and emergency invasive procedures in patients on dabigatran: updated guidelines from the French Working Group on Perioperative Haemostasis (GIHP), September 2016

<30 ng/mL is safe to operate in very high hemorrhagic risk patients (neurosurgery, liver surgery)

<30 ng/mL for perimedullar anesthesia or deep nerve block

<50 ng/mL is safe to operate in high hemorrhage risk patients where hemostasis is controllable and low risk patients

Steps to be taken when the thresholds are not met depending on the urgency of the procedure are detailed in the article. They mainly include reversal through antidotes and waiting

Rivaroxaban

Pernod et al. (2013)[84]

Recommendations of the French Working Group on Perioperative Haemostasis

<30 ng/mL to operate without increased bleeding risk

30–200 ng/mL: delay intervention if possible by 12 h then re-measure

200–400 ng/mL: delay intervention if possible by 12–24 h then re-measure

>400 ng/mL: high risk of hemorrhage, overdose

The 30 ng/mL threshold comes from the expected plasma concentrations with the stopping time of 48 h in the elective surgery protocol of the ROCKET-AF trial. The other thresholds are expert opinions

Steiner et al. (2013)[124]

Recommendations for the emergency management of DOAC-related complications

<100 ng/mL to consider thrombolysis in ischemic stroke

<100 ng/mL to consider intervention in SAH patients

Level of evidence IV: expert opinion for both thresholds

Douketis et al. (2017)[82]

The Perioperative Anticoagulant Use for Surgery Evaluation (PAUSE) study for patients on a direct oral anticoagulant who need an elective surgery or procedure: design and rationale

<50 ng/mL as a safe threshold for invasive procedures

Decided based on pharmacokinetic studies by expert consensus

Apixaban

Steiner et al. (2013)[124]

Recommendations for the emergency management of DOAC-related complications

<10 ng/mL to consider thrombolysis in ischemic stroke

<10 ng/mL to consider intervention in SAH patients

Level of evidence IV: expert opinion for both thresholds. This is possibly a typographical error since 10-fold higher thresholds are mentioned in the same paper for rivaroxaban while both DOACs do not exhibit such a difference in expected concentrations

Douketis et al. (2017)[82]

The Perioperative Anticoagulant Use for Surgery Evaluation (PAUSE) study for patients on a direct oral anticoagulant who need an elective surgery or procedure: design and rationale

<50 ng/mL as a safe threshold for invasive procedures

Decided based on pharmacokinetic studies by expert consensus

Abbreviations: DOAC, direct oral anticoagulant; SAH, subarachnoid hemorrhage.


When thrombolytic treatment for stroke is needed, DOAC levels should be monitored to ensure the safety of the procedure. Certain thresholds have been defined to consider a patient safe for thrombolysis; these can be found in [Table 4].

DOAC monitoring in the perioperative setting probably has a place since it can greatly influence the clinician's decisions; however, more studies are required to clearly identify safe thresholds for various procedures and when reversal is needed.


#

Liver Insufficiency

Patients with liver insufficiency have often been excluded from phase III DOAC trials and therefore little data on how this affects drug levels have been produced. DOACs are all partly eliminated by the liver and therefore caution should be exerted in such cases. Therefore, DOACs are currently contraindicated in severe cirrhosis, especially if there is associated coagulopathy. The AUC of rivaroxaban doubles in patients with Child-B cirrhosis[86]; a doubling in AUC has been shown to increase by 50% the risk of major bleeding in patients taking rivaroxaban.[23] A meta-analysis on the subject of DOACs in cirrhosis patients showed that DOACs have a similar safety profile as VKA and low-molecular-weight heparin.[87]

Bleeding and thromboembolic events are more frequent in cirrhosis.[88] In addition to DOAC accumulation due to liver failure, hypoalbuminemia, a common factor in cirrhosis, is associated with higher bleeding rates in rivaroxaban.[29] Therefore, specific care should be taken in cirrhotic patients receiving DOACs, especially since hepatic vein thrombosis is starting to be treated with DOACs off label.[89]

Monitoring DOAC concentrations in this patient population could lead to better safety and efficacy but studies are needed to confirm the usefulness of it.


#

Drug Interactions

DOACs are subject to many clinically important interactions (see [Table 5]) with significant variations in their concentrations. Most of these drugs are not formally contraindicated and caution measures, such as monitoring DOAC levels, could be taken when it is necessary to treat the patient with both drugs.

Table 5

DOAC interactions that affect plasma levels

DOAC

Ref.

Drugs at risk of interaction[a]

Increases significantly plasma levels

Lowers significantly plasma levels

Dabigatran

[13] [96]

Potent P-gp inhibitors/inducers

P-gp/CYP3A4 dual inhibitors/inducers

Ketoconazole and Itraconazole [ b ]

Dronedarone [ b ]

Ciclosporin [ b ]

Tacrolimus

Ritonavir and protease inhibitors [ b ]

Verapamil[ d ]

Amiodarone[ d ]

Quinidine [ b ]

Macrolides

Ticagrelor

Posaconazole

Rifampicin[c]

St John's Wort[c]

Carbamazepine[c]

Phenytoin[c]

Magnesium and aluminum based antacids[ d ]

Rivaroxaban

[22] [105] [110] [125]

Potent P-gp/CYP 3A4 dual inhibitors/inducers

Ketoconazole, posaconazole, and itraconazole [e]

Ritonavir and protease inhibitors [e]

Macrolides[f]

Diltiazem[f]

Verapamil[f]

Rifampicin[c]

St John's-Woth[c]

Carbamazepine[c]

Phenytoin[c]

Apixaban

[42] [112]

Potent P-gp/CYP 3A4 dual inhibitors/inducers

Ketoconazole, posaconazole, and itraconazole [e]

Ritonavir and protease inhibitors [e]

Macrolides[f]

Diltiazem[f]

Rifampicin [c]

St John's-Woth [c]

Carbamazepine [c]

Phenytoin [c]

Edoxaban

[114] [115] [126]

Potent P-gp inhibitors

Ketoconazole[g]

Erythromycin[g]

Ciclosporin[g]

Protease inhibitors

Verapamil[g]

Dronedarone[g]

Quinidine[g]

Rifampicin

Abbreviation: DOAC, direct oral anticoagulant.


Note: In italics: contraindicated; in bold: not recommended or to be avoided.


a Cytostatic agents and other anticancer drugs could also be at risk of interaction and caution should be exerted in these cases since pharmacokinetic as well as pharmacodynamic interactions have been suggested.[127]


b Increased risk of hemorrhage, contraindicated.


c Concomitant use should be avoided, risk of important lowering of efficacy.


d Should be taken at least 2 hours after Pradaxa, clinicians should be cautious.


e To be avoided, increased hemorrhagic risk, if unavoidable surveillance is warranted.


f ROCKET-AF data showed no increased risk of hemorrhage if GFR >30 mL/min.[110]


g Lead to higher concentrations and higher bleeding risk, dose adaptation to 30 mg once daily is needed.


A study performed by Chang et al. (n = 91,330) assessed the bleeding risk of patients treated by DOACs (dabigatran, rivaroxaban, and apixaban) and concomitant drugs. They showed that drugs with interaction risks are frequently prescribed with DOACs. Amiodarone and fluconazole were associated with a significantly higher bleeding rate. They paradoxically found that atorvastatin reduced the bleeding rate, which they associated with the lower incidence of hemorrhagic transformation of ischemic strokes. They did not find an increased bleeding risk in patients treated with digoxin, verapamil, cyclosporine, and macrolides.[90]


#

Extreme-Weight Patients

Extreme-weight patients (<50 kg and >120 kg) can be exposed to higher and lower concentrations respectively and sometimes out-of-target drug levels.[91] Some studies have shown variability in DOAC AUC with extreme weight but the effect was not deemed clinically significant.[92] [93] Safety and efficacy were shown to be similar to those of VKA in this population.[94] However, it is worth mentioning that a meta-analysis of phase III trial data for DOACs showed a similar safety profile in extreme-weight patients but these studies did not include many patients weighing over 100 kg, which may be too low to show an effect.[95] A recent in vivo study on obese patients showed that rivaroxaban concentrations above 80 ng/mL efficiently inhibited thrombin generation, an effect that disappeared at concentrations below 50 ng/mL.[85] For dabigatran, product information mentions an increase in plasma levels with low body weight (<50 kg).[96] It has been suggested that in the very obese with supratherapeutic creatinine clearance, dabigatran concentrations could be subtherapeutic.[97]

As few trials have studied the safety of usual DOAC doses in extreme-weight patients, it is safe to monitor such patients until further evidence is provided.[98] [99] [100]


#

High Bleeding Risk Patients

In patients whose clinical scores such as the “HAS-BLED score” show important hemorrhagic risk and who are still being treated with DOAC, it could be safe to monitor drug levels. It should be safe to ensure that patients with clinical conditions that could favor bleeding do not have high DOAC through plasma levels.


#

Adherence Monitoring

Doubts on patient compliance could be in part alleviated by drug-level monitoring. Hu et al. found in a study dedicated to compliance assessment in dabigatran therapy for AF that 10.7% of patients were noncompliant and that dabigatran monitoring was a way of identifying such patients.[101] A study conducted by Keita et al. showed that only 50 to 67.5% of patients taking DOACs had a high adherence rate as defined by the MMAS-8 adherence score.[102]

A barrier to DOAC adherence monitoring is that their concentration at trough is not dependent on adherence; trough levels reached after the first pill are the same after the next. Therefore, DOAC monitoring cannot verify compliance on the long term; it can just confirm that the last dose has been taken. Peak levels are too variable to be used in this way and the peak time is too narrow for clinical use. D-dimers could be used for adherence monitoring as they are lowered by efficient anticoagulation.[103]


#

A Practical Approach to DOAC Monitoring: A Case Study

We present here a case vignette describing the practical approach to DOAC monitoring and results' interpretation using the algorithm depicted in [Fig. 2].

Zoom Image
Fig. 2 Proposed DOAC monitoring algorithm. In green: estimated median concentration target. In yellow: estimated target range. In orange: increased risk of adverse event. In red: highly increased risk of adverse event (see [Fig. 1] for references). DOAC, direct oral anticoagulant.

An 86-year-old female patient known for diabetes and AF treated by metformin and apixaban 5 mg bid presents to the emergency department (ED) in the afternoon, shortly after a fall from her height in the street without witness. She cannot tell whether she hit her head or had loss of consciousness and has no specific complaints besides a slight headache. The review of systems is without particularity. She recalls having taken apixaban 5 mg in the morning. The clinical exam reveals hypovolemia with signs of dehydration and a frontal hematoma without neurological focal signs. Her blood panels show an acute renal insufficiency with a creatinine clearance of 35 mL/min/1.783 m2 and an inflammatory syndrome. While the patient was being watched in the ED, she developed an altered mental state with disorientation. A cerebral CT-scan showed an acute right-sided subdural hematoma.

As discussed above, bleeds (even if provoked) while on anticoagulant therapy should prompt monitoring of the DOAC plasma levels. A quantitative measurement at admission will help in guiding the reversal strategy. In this case, an initial apixaban concentration >600 ng/mL justified the administration of high-dose (i.e., 50 IU/kg body weight) 4-factor prothrombin concentrate in addition to tranexamic acid. At follow-up, after correction of the hypovolemia and improvement of the renal function, the patient's apixaban trough level was measured at 170 ng/mL, a value which is at the higher side of the observed concentration range and associated with higher bleeding rates (see [Figs. 1] and [2]).[31] [32] [33] [34] [35] [36] [37] [38] [39] [40] [41] [42] Despite no formal indication, the patient's dose was lowered to 2.5 ng/mL and a later trough level was found to be 100 ng/mL, which allowed for continuation of the latter dosage.


#

Conclusion

Many questions still exist regarding whether DOACs should be monitored. However, an increasing amount of evidence is showing that high or low plasma levels can lead to increased adverse events. Therefore, improving the efficacy and safety of DOACs could be possible through plasma level monitoring. Precise therapeutic intervals are still lacking as well as threshold for increased adverse event risk and more studies are required to identify populations in which monitoring would be useful.

Randomized controlled clinical trials investigating the effect of DOAC plasma level monitoring and dose tailoring are the next step to determine the therapeutic range of these drugs and to evaluate whether DOAC monitoring can be used effectively to improve their usage.

For the time being, we can only consider the observed trough level ranges as an approximation of drug targets and monitor populations which are likely to benefit most from this testing, for instance, to exclude drug accumulation, when drug failure is suspected, or in acute situations such as hemorrhagic or thrombotic events.


#
#

Conflict of Interest

Thomas Moner-Banet does not declare any conflict of interest. Pierre-Alexandre Bart does not declare any conflict of interest. Lorenzo Alberio declares to have received grants/research support from Bayer, support for the CHUV Haemophilia Nurses Program from Bayer, honoraria for participating in scientific advisory boards: Bayer, Boehringer Ingelheim, Daiichi Sankyo, and Pfizer, and honoraria as a consultant/speaker: Bayer and Boehringer Ingelheim.

  • References

  • 1 Mismetti P, Laporte S. New oral antithrombotics: a need for laboratory monitoring. For. J Thromb Haemost 2010; 8 (04) 621-626
  • 2 Connolly SJ, Ezekowitz MD, Yusuf S. , et al. RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 2009; 361 (12) 1139-1151
  • 3 Giugliano RP, Ruff CT, Braunwald E. , et al. ENGAGE AF-TIMI 48 Investigators. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2013; 369 (22) 2093-2104
  • 4 Patel MR, Mahaffey KW, Garg J. , et al. ROCKET AF Investigators. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011; 365 (10) 883-891
  • 5 Granger CB, Alexander JH, McMurray JJV. , et al. ARISTOTLE Committees and Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2011; 365 (11) 981-992
  • 6 Reilly PA, Lehr T, Haertter S. , et al. RE-LY Investigators. The effect of dabigatran plasma concentrations and patient characteristics on the frequency of ischemic stroke and major bleeding in atrial fibrillation patients: the RE-LY Trial (Randomized Evaluation of Long-Term Anticoagulation Therapy). J Am Coll Cardiol 2014; 63 (04) 321-328
  • 7 Powell JR. Are new oral anticoagulant dosing recommendations optimal for all patients?. JAMA 2015; 313 (10) 1013-1014
  • 8 Paré G, Eriksson N, Lehr T. , et al. Genetic determinants of dabigatran plasma levels and their relation to bleeding. Circulation 2013; 127 (13) 1404-1412
  • 9 Cohen D. Dabigatran: how the drug company withheld important analyses. BMJ 2014; 349: g4670
  • 10 An idea for a mid to long term strategy for Pradaxa [Internet]. Boehringer Ingelheim International. Available at: https://www.bmj.com/investigation/dabigatran . Accessed February 26, 2019
  • 11 Safouris A, Triantafyllou N, Parissis J, Tsivgoulis G. The case for dosing dabigatran: how tailoring dose to patient renal function, weight and age could improve the benefit-risk ratio. Ther Adv Neurol Disorder 2015; 8 (06) 245-254
  • 12 FDA. Pradaxa clinical pharmacology and biopharmaceutics review(s) [Internet]. 2011 [cited 2019 Feb 25]. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2010/022512Orig1s000ClinPharmR_Corrrected%203.11.2011.pdf . Accessed February 25, 2019
  • 13 Infromation professionnelle du Compendium Suisse des Médicaments: Pradaxa [Internet]. Compendium Suisse des Médicaments. Available at: https://compendium.ch/mpro/mnr/23436/html/fr . Accessed March 5, 2019
  • 14 Šinigoj P, Malmström RE, Vene N. , et al. Dabigatran concentration: variability and potential bleeding prediction in “real-life” patients with atrial fibrillation. Basic Clin Pharmacol Toxicol 2015; 117 (05) 323-329
  • 15 Albaladejo P, Samama C-M, Sié P. , et al. GIHP-NACO Study Group. Management of severe bleeding in patients treated with direct oral anticoagulants: an observational registry analysis. Anesthesiology 2017; 127 (01) 111-120
  • 16 Volbers B, Köhrmann M, Kallmünzer B. , et al. Dabigatran plasma levels in acute cerebrovascular events. J Stroke Cerebrovasc Dis 2016; 25 (04) 877-882
  • 17 Eriksson BI, Dahl OE, Büller HR. , et al. BISTRO II Study Group. A new oral direct thrombin inhibitor, dabigatran etexilate, compared with enoxaparin for prevention of thromboembolic events following total hip or knee replacement: the BISTRO II randomized trial. J Thromb Haemost 2005; 3 (01) 103-111
  • 18 Testa S, Paoletti O, Legnani C. , et al. Low drug levels and thrombotic complications in high-risk atrial fibrillation patients treated with direct oral anticoagulants. J Thromb Haemost 2018; 16 (05) 842-848
  • 19 Chin PK, Wright DF, Patterson DM, Doogue MP, Begg EJ. A proposal for dose-adjustment of dabigatran etexilate in atrial fibrillation guided by thrombin time. Br J Clin Pharmacol 2014; 78 (03) 599-609
  • 20 Chaussade E, Hanon O, Boully C. , et al. Real-life peak and trough dabigatran plasma measurements over time in hospitalized geriatric patients with atrial fibrillation. J Nutr Health Aging 2018; 22 (01) 165-173
  • 21 Schellings MW, Boonen K, Schmitz EM. , et al. Determination of dabigatran and rivaroxaban by ultra-performance liquid chromatography-tandem mass spectrometry and coagulation assays after major orthopaedic surgery. Thromb Res 2016; 139: 128-134
  • 22 Mueck W, Stampfuss J, Kubitza D, Becka M. Clinical pharmacokinetic and pharmacodynamic profile of rivaroxaban. Clin Pharmacokinet 2014; 53 (01) 1-16
  • 23 FDA. Xarelto – clinical pharmacology & biopharmaceutical review [Internet]. FDA; 2009. . Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022406Orig1s000ClinPharmR.pdf . Accessed February 25, 2019
  • 24 Girgis IG, Patel MR, Peters GR. , et al. Population pharmacokinetics and pharmacodynamics of rivaroxaban in patients with non-valvular atrial fibrillation: results from ROCKET AF. J Clin Pharmacol 2014; 54 (08) 917-927
  • 25 Gulilat M, Tang A, Gryn SE. , et al. Interpatient variation in rivaroxaban and apixaban plasma concentrations in routine care. Can J Cardiol 2017; 33 (08) 1036-1043
  • 26 Kanuri SH, Kreutz RP. Pharmacogenomics of novel direct oral anticoagulants: newly identified genes and genetic variants. J Pers Med 2019; 9 (01) E7
  • 27 Kubitza D, Berkowitz SD, Misselwitz F. Evidence-based development and rationale for once-daily rivaroxaban dosing regimens across multiple indications. Clin Appl Thromb Hemost 2016; 22 (05) 412-422
  • 28 Nakano Y, Kondo T, Osanai H. , et al. Clinical usefulness of measuring prothrombin time and soluble fibrin levels in Japanese patients with atrial fibrillation receiving rivaroxaban. J Cardiol 2015; 65 (03) 185-190
  • 29 Woodruff AE, Wovkulich MM, Mogle BT, Hassan AK. Association between prothrombin time and bleeding in hospitalized patients receiving rivaroxaban. Am J Health Syst Pharm 2018; 75 (22) 1783-1789
  • 30 Sakaguchi T, Osanai H, Murase Y. , et al. Monitoring of anti-Xa activity and factors related to bleeding events: A study in Japanese patients with nonvalvular atrial fibrillation receiving rivaroxaban. J Cardiol 2017; 70 (03) 244-249
  • 31 Wada S, Toyoda K, Sato S. , et al. Anti-Xa activity and event risk in patients with direct factor Xa inhibitors initiated early after stroke. Circ J 2018; 82 (11) 2872-2879
  • 32 Krause M, Henningsen A, Torge A. , et al. Impact of gender on safety and efficacy of Rivaroxaban in adolescents & young adults with venous thromboembolism. Thromb Res 2016; 148: 145-151
  • 33 Seiffge DJ, Kägi G, Michel P. , et al. Novel Oral Anticoagulants in Stroke Patients study group. Rivaroxaban plasma levels in acute ischemic stroke and intracerebral hemorrhage. Ann Neurol 2018; 83 (03) 451-459
  • 34 Zalewski J, Rychlak R, Góralczyk T, Undas A. Rivaroxaban concentration in patients with deep vein thrombosis who reported thrombus progression or minor hemorrhagic complications: first Polish experience. Pol Arch Med Wewn 2014; 124 (10) 553-555
  • 35 Romano SL, Chiarugi P, Casini M, Pellegrini G, Ruocco L. A low rivaroxaban plasma level may indicate anticoagulation undertreatment. Eur J Case Rep Intern Med 2018; 5 (11) 000937
  • 36 Ing Lorenzini K, Daali Y, Fontana P, Desmeules J, Samer C. Rivaroxaban-induced hemorrhage associated with ABCB1 genetic defect. Front Pharmacol 2016; 7: 494
  • 37 Barton J, Wong A, Graudins A. Anti-Xa activity in apixaban overdose: a case report. Clin Toxicol (Phila) 2016; 54 (09) 871-873
  • 38 Repplinger DJ, Hoffman RS, Nelson LS, Hines EQ, Howland M, Su MK. Lack of significant bleeding despite large acute rivaroxaban overdose confirmed with whole blood concentrations. Clin Toxicol (Phila) 2016; 54 (08) 647-649
  • 39 Spiller HA, Mowry JB, Aleguas Jr A. , et al. An observational study of the factor Xa inhibitors rivaroxaban and apixaban as reported to eight poison centers. Ann Emerg Med 2016; 67 (02) 189-195
  • 40 Becker RC, Alexander JH, Newby LK. , et al. Effect of apixaban, an oral and direct factor Xa inhibitor, on coagulation activity biomarkers following acute coronary syndrome. Thromb Haemost 2010; 104 (05) 976-983
  • 41 Frost C, Wang J, Nepal S. , et al. Apixaban, an oral, direct factor Xa inhibitor: single dose safety, pharmacokinetics, pharmacodynamics and food effect in healthy subjects. Br J Clin Pharmacol 2013; 75 (02) 476-487
  • 42 Eliquis: Summary of product characteristics [Internet]. European Medicines Agency. Available at: https://www.ema.europa.eu/en/documents/product-information/eliquis-epar-product-information_en.pdf . Accessed March 4, 2019
  • 43 Ueshima S, Hira D, Fujii R. , et al. Impact of ABCB1, ABCG2, and CYP3A5 polymorphisms on plasma trough concentrations of apixaban in Japanese patients with atrial fibrillation. Pharmacogenet Genomics 2017; 27 (09) 329-336
  • 44 FDA. Apixaban clinical pharmacology/biopharmaceutics review [Internet]. FDA; 2012. . Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/202155Orig1s000ClinPharmR.pdf . Accessed February 25, 2019
  • 45 Bhagirath VC, Eikelboom JW, Hirsh J. , et al. Apixaban-calibrated anti-FXa activity in relation to outcome events and clinical characteristics in patients with atrial fibrillation: results from the AVERROES trial. TH Open 2017; 1 (02) e139-e145
  • 46 Byon W, Sweeney K, Frost C, Boyd RA. Population pharmacokinetics, pharmacodynamics, and exploratory exposure-response analyses of apixaban in subjects treated for venous thromboembolism. CPT Pharmacometrics Syst Pharmacol 2017; 6 (05) 340-349
  • 47 Testa S, Dellanoce C, Paoletti O. , et al. Edoxaban plasma levels in patients with non-valvular atrial fibrillation: Inter and intra-individual variability, correlation with coagulation screening test and renal function. Thromb Res 2019; 175: 61-67
  • 48 FDA. Edoxaban clinical pharmacology and biopharmaceutics review [Internet]. FDA; 2014 . Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/206316Orig1Orig2s000ClinPharmR.pdf . Accessed February 25, 2019
  • 49 Weitz JI, Connolly SJ, Patel I. , et al. Randomised, parallel-group, multicentre, multinational phase 2 study comparing edoxaban, an oral factor Xa inhibitor, with warfarin for stroke prevention in patients with atrial fibrillation. Thromb Haemost 2010; 104 (03) 633-641
  • 50 Giugliano RP. Compilation of oral presentations: OC-WE-003 The relationship between oral factor Xa (FXa) inhibitor du-176b pharmacokinetics (PK) and the probability of bleeding events (BE) in patients with atrial fibrillation (AF). Boston, MA: International Society on Thrombosis and Haemostasis; 2009
  • 51 Salazar DE, Mendell J, Kastrissios H. , et al. Modelling and simulation of edoxaban exposure and response relationships in patients with atrial fibrillation. Thromb Haemost 2012; 107 (05) 925-936
  • 52 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 AF-TIMI 48 trial. Lancet 2015; 385 (9984): 2288-2295
  • 53 Yin OQP, Antman EM, Braunwald E. , et al. Linking endogenous factor Xa activity, a biologically relevant pharmacodynamic marker, to edoxaban plasma concentrations and clinical outcomes in the ENGAGE AF-TIMI 48 trial. Circulation 2018; 138 (18) 1963-1973
  • 54 Chao T-F, Chen S-A, Ruff CT. , et al. Clinical outcomes, edoxaban concentration, and anti-factor Xa activity of Asian patients with atrial fibrillation compared with non-Asians in the ENGAGE AF-TIMI 48 trial. Eur Heart J 2019; 40 (19) 1518-1527
  • 55 Conway SE, Hwang AY, Ponte CD, Gums JG. Laboratory and clinical monitoring of direct acting oral anticoagulants: what clinicians need to know. Pharmacotherapy 2017; 37 (02) 236-248
  • 56 Tamigniau A, Douxfils J, Nicolas J-B. , et al. Why, when and how to monitor new oral anticoagulants [in French]. Rev Med Suisse 2014; 10 (416) 326-333
  • 57 Moudallel S, Steurbaut S, Cornu P, Dupont A. Appropriateness of DOAC prescribing before and during hospital admission and analysis of determinants for inappropriate prescribing. Front Pharmacol 2018; 9: 1220
  • 58 Gupta M, Singh N, Tsigoulis M. , et al. Underuse of full dose factor Xa inhibition in atrial fibrillation: insight from the SPRINT-AF registry. J Am Coll Cardiol 2015; 65 (10) A348
  • 59 Wehbe RM, Yadlapati A. Underuse of oral anticoagulants for nonvalvular atrial fibrillation: past, present, and future. Tex Heart Inst J 2016; 43 (04) 287-290
  • 60 Avorn J. The psychology of clinical decision making - implications for medication use. N Engl J Med 2018; 378 (08) 689-691
  • 61 Dillinger J-G, Aleil B, Cheggour S. , et al. Dosing issues with non-vitamin K antagonist oral anticoagulants for the treatment of non-valvular atrial fibrillation: why we should not underdose our patients. Arch Cardiovasc Dis 2018; 111 (02) 85-94
  • 62 Shrestha S, Baser O, Kwong WJ. Effect of renal function on dosing of non-vitamin K antagonist direct oral anticoagulants among patients with nonvalvular atrial fibrillation. Ann Pharmacother 2018; 52 (02) 147-153
  • 63 Turpie AGG, Purdham D, Ciaccia A. Nonvitamin K antagonist oral anticoagulant use in patients with renal impairment. Ther Adv Cardiovasc Dis 2017; 11 (09) 243-256
  • 64 Lutz J, Jurk K, Schinzel H. Direct oral anticoagulants in patients with chronic kidney disease: patient selection and special considerations. Int J Nephrol Renovasc Dis 2017; 10: 135-143
  • 65 Barrios V, Górriz JL. Atrial fibrillation and chronic kidney disease: focus on rivaroxaban. J Comp Eff Res 2015; 4 (06) 651-664
  • 66 Andò 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: 162-169
  • 67 Feldberg J, Patel P, Farrell A. , et al. A systematic review of direct oral anticoagulant use in chronic kidney disease and dialysis patients with atrial fibrillation. Nephrol Dial Transplant 2019; 34 (02) 265-277
  • 68 Greenblatt DJ, Patel M, Harmatz JS, Nicholson WT, Rubino CM, Chow CR. Impaired rivaroxaban clearance in mild renal insufficiency with verapamil coadministration: potential implications for bleeding risk and dose selection. J Clin Pharmacol 2018; 58 (04) 533-540
  • 69 Becattini C, Giustozzi M, Ranalli MG. , et al. Variation of renal function over time is associated with major bleeding in patients treated with direct oral anticoagulants for atrial fibrillation. J Thromb Haemost 2018; 16 (05) 833-841
  • 70 Min M, Sibicky S. Concerns for bleeding in the elderly with the use of direct oral anticoagulants. Consult Pharm 2018; 33 (05) 262-267
  • 71 Ruff CT, Giugliano RP, Braunwald E. , et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet 2014; 383 (9921): 955-962
  • 72 Eikelboom JW, Wallentin L, Connolly SJ. , et al. Risk of bleeding with 2 doses of dabigatran compared with warfarin in older and younger patients with atrial fibrillation: an analysis of the randomized evaluation of long-term anticoagulant therapy (RE-LY) trial. Circulation 2011; 123 (21) 2363-2372
  • 73 Kailas SD, Thambuluru SR. Efficacy and safety of direct oral anticoagulants compared to warfarin in prevention of thromboembolic events among elderly patients with atrial fibrillation. Cureus 2016; 8 (10) e836
  • 74 Bando S, Nishikado A, Hiura N. , et al. Efficacy and safety of rivaroxaban in extreme elderly patients with atrial fibrillation: analysis of the Shikoku Rivaroxaban Registry Trial (SRRT). J Cardiol 2018; 71 (02) 197-201
  • 75 Nissan R, Spectre G, Hershkovitz A. , et al. Apixaban levels in octogenarian patients with non-valvular atrial fibrillation. Drugs Aging 2019; 36 (02) 165-177
  • 76 Khan F, Huang H, Datta YH. Direct oral anticoagulant use and the incidence of bleeding in the very elderly with atrial fibrillation. J Thromb Thrombolysis 2016; 42 (04) 573-578
  • 77 Levy JH, Ageno W, Chan NC, Crowther M, Verhamme P, Weitz JI. ; Subcommittee on Control of Anticoagulation. When and how to use antidotes for the reversal of direct oral anticoagulants: guidance from the SSC of the ISTH. J Thromb Haemost 2016; 14 (03) 623-627
  • 78 Albaladejo P, Pernod G, Godier A. , et al. Members of the French Working Group on Perioperative Haemostasis. Management of bleeding and emergency invasive procedures in patients on dabigatran: updated guidelines from the French Working Group on Perioperative Haemostasis (GIHP) - September 2016. Anaesth Crit Care Pain Med 2018; 37 (04) 391-399
  • 79 Steffel J, Verhamme P, Potpara TS. , et al. ESC Scientific Document Group. The 2018 European Heart Rhythm Association Practical Guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation. Eur Heart J 2018; 39 (16) 1330-1393
  • 80 Kaatz S, Mahan CE, Nakhle A. , et al. Management of elective surgery and emergent bleeding with direct oral anticoagulants. Curr Cardiol Rep 2017; 19 (12) 124
  • 81 Prisco D, Ageno W, Becattini C. , et al. SIMI (Italian Society of Internal Medicine); FADOI (Federation of Associations of Hospital Doctors on Internal Medicine); SISET (Italian Society for the Study of Haemostasis and Thrombosis). Italian intersociety consensus on DOAC use in internal medicine. Intern Emerg Med 2017; 12 (03) 387-406
  • 82 Douketis JD, Spyropoulos AC, Anderson JM. , et al. The Perioperative Anticoagulant Use for Surgery Evaluation (PAUSE) study for patients on a direct oral anticoagulant who need an elective surgery or procedure: design and rationale. Thromb Haemost 2017; 117 (12) 2415-2424
  • 83 Godier A, Dincq A-S, Martin A-C. , et al. Predictors of pre-procedural concentrations of direct oral anticoagulants: a prospective multicentre study. Eur Heart J 2017; 38 (31) 2431-2439
  • 84 Pernod G, Albaladejo P, Godier A. , et al. Management of major bleeding complications and emergency surgery in patients on long-term treatment with direct oral anticoagulants, thrombin or factor-Xa inhibitors. Proposals of the Working Group on Perioperative Haemostasis (GIHP) - March 2013 [in French]. Ann Fr Anesth Reanim 2013; 32 (10) 691-700
  • 85 Bertaggia-Calderara D, Kröll D, Gerschheimer C. , et al. Effect of rivaroxaban on thrombin generation in vivo. A study in obese patients. Int J Lab Hematol 2018; 40 (01) e11-e14
  • 86 Graff J, Harder S. Anticoagulant therapy with the oral direct factor Xa inhibitors rivaroxaban, apixaban and edoxaban and the thrombin inhibitor dabigatran etexilate in patients with hepatic impairment. Clin Pharmacokinet 2013; 52 (04) 243-254
  • 87 Hoolwerf EW, Kraaijpoel N, Büller HR, van Es N. Direct oral anticoagulants in patients with liver cirrhosis: a systematic review. Thromb Res 2018; 170: 102-108
  • 88 Ha NB, Regal RE. Anticoagulation in patients with cirrhosis: caught between a rock-liver and a hard place. Ann Pharmacother 2016; 50 (05) 402-409
  • 89 Priyanka P, Kupec JT, Krafft M, Shah NA, Reynolds GJ. Newer oral anticoagulants in the treatment of acute portal vein thrombosis in patients with and without cirrhosis. Int J Hepatol 2018; 2018: 8432781
  • 90 Chang SH, Chou IJ, Yeh YH. , et al. Association between use of non-vitamin K oral anticoagulants with and without concurrent medications and risk of major bleeding in nonvalvular atrial fibrillation. JAMA 2017; 318 (13) 1250-1259
  • 91 Patel JP, Roberts LN, Arya R. Anticoagulating obese patients in the modern era. Br J Haematol 2011; 155 (02) 137-149
  • 92 Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Body weight has limited influence on the safety, tolerability, pharmacokinetics, or pharmacodynamics of rivaroxaban (BAY 59-7939) in healthy subjects. J Clin Pharmacol 2007; 47 (02) 218-226
  • 93 Upreti VV, Wang J, Barrett YC. , et al. Effect of extremes of body weight on the pharmacokinetics, pharmacodynamics, safety and tolerability of apixaban in healthy subjects. Br J Clin Pharmacol 2013; 76 (06) 908-916
  • 94 Di Minno MND, Lupoli R, Di Minno A, Ambrosino P, Scalera A, Dentali F. Effect of body weight on efficacy and safety of direct oral anticoagulants in the treatment of patients with acute venous thromboembolism: a meta-analysis of randomized controlled trials. Ann Med 2015; 47 (01) 61-68
  • 95 Boonyawat K, Caron F, Li A. , et al. Association of body weight with efficacy and safety outcomes in phase III randomized controlled trials of direct oral anticoagulants: a systematic review and meta-analysis. J Thromb Haemost 2017; 15 (07) 1322-1333
  • 96 Pradaxa: summary of product characteristics [Internet]. European Medicines Agency; Available at: https://www.ema.europa.eu/documents/product-information/pradaxa-epar-product-information_en.pdf . Accessed March 9, 2020
  • 97 Safouris A, Demulder A, Triantafyllou N, Tsivgoulis G. Rivaroxaban presents a better pharmacokinetic profile than dabigatran in an obese non-diabetic stroke patient. J Neurol Sci 2014; 346 (1–2): 366-367
  • 98 Güler E, Babur Güler G, Demir GG, Hatipoğlu S. A review of the fixed dose use of new oral anticoagulants in obese patients: Is it really enough?. Anatol J Cardiol 2015; 15 (12) 1020-1029
  • 99 Kröll D, Stirnimann G, Vogt A. , et al. Pharmacokinetics and pharmacodynamics of single doses of rivaroxaban in obese patients prior to and after bariatric surgery. Br J Clin Pharmacol 2017; 83 (07) 1466-1475
  • 100 Kröll D, Nett PC, Borbély YM. , et al. The effect of bariatric surgery on the direct oral anticoagulant rivaroxaban: the extension study. Surg Obes Relat Dis 2018; 14 (12) 1890-1896
  • 101 Hu YF, Liao JN, Chern CM. , et al. Identification and management of noncompliance in atrial fibrillation patients receiving dabigatran: the role of a drug monitor. Pacing Clin Electrophysiol 2015; 38 (04) 465-471
  • 102 Keita I, Aubin-Auger I, Lalanne C. , et al. Assessment of quality of life, satisfaction with anticoagulation therapy, and adherence to treatment in patients receiving long-course vitamin K antagonists or direct oral anticoagulants for venous thromboembolism. Patient Prefer Adherence 2017; 11: 1625-1634
  • 103 Ombandza-Moussa E, Samama MM, Horellou MH, Chatelier AL, Elalamy I, Conard J. Influence of oral anticoagulant treatment on D-dimers levels [in French]. Ann Biol Clin (Paris) 2001; 59 (05) 579-583
  • 104 van Ryn J, Stangier J, Haertter S. , et al. Dabigatran etexilate--a novel, reversible, oral direct thrombin inhibitor: interpretation of coagulation assays and reversal of anticoagulant activity. Thromb Haemost 2010; 103 (06) 1116-1127
  • 105 Xarelto: summary of product characteristics [Internet]. European Medicines Agency. Available at: https://www.ema.europa.eu/en/documents/product-information/xarelto-epar-product-information_en.pdf . Accessed March 5, 2019
  • 106 Beyer J, Trujillo T, Fisher S, Ko A, Lind SE, Kiser TH. Evaluation of a heparin-calibrated antifactor Xa assay for measuring the anticoagulant effect of oral direct Xa inhibitors. Clin Appl Thromb Hemost 2016; 22 (05) 423-428
  • 107 Verhamme P, Wells PS, Segers A. , et al. Dose reduction of edoxaban preserves efficacy and safety for the treatment of venous thromboembolism. An analysis of the randomised, double-blind HOKUSAI VTE trial. Thromb Haemost 2016; 116 (04) 747-753
  • 108 Liesenfeld KH, Lehr T, Dansirikul C. , et al. Population pharmacokinetic analysis of the oral thrombin inhibitor dabigatran etexilate in patients with non-valvular atrial fibrillation from the RE-LY trial. J Thromb Haemost 2011; 9 (11) 2168-2175
  • 109 Stangier J, Clemens A. Pharmacology, pharmacokinetics, and pharmacodynamics of dabigatran etexilate, an oral direct thrombin inhibitor. Clin Appl Thromb Hemost 2009; 15 (Suppl. 01) 9S-16S
  • 110 Information professionnelle du Compendium Suisse des Médicaments: Xarelto [Internet]. Compendium Suisse des Médicaments; 2018 . Available at: https://compendium.ch/mpro/mnr/20753/html/fr?Platform=Desktop#7600 . Accessed February 25, 2019
  • 111 Kreutz R. Pharmacodynamic and pharmacokinetic basics of rivaroxaban. Fundam Clin Pharmacol 2012; 26 (01) 27-32
  • 112 Information professionnelle du Compendium Suisse des Médicaments: Eliquis [Internet]. Compendium Suisse des Médicaments; 2019 . Available at: https://compendium.ch/mpro/mnr/22732/html/fr#7300 . Accessed March 19, 2019
  • 113 Kubisz P, Stanciakova L, Dobrotova M, Samos M, Mokan M, Stasko J. Apixaban - metabolism, pharmacologic properties and drug interactions. Curr Drug Metab 2017; 18 (07) 609-621
  • 114 Lixiana: summary of product characteristics [Internet]. European Medicines Agency. Available at: https://www.ema.europa.eu/en/documents/product-information/lixiana-epar-product-information_en.pdf . Accessed March 19, 2019
  • 115 Information professionnelle du Compendium Suisse des Médicaments: Lixiana [Internet]. Compendium Suisse des Médicaments; 2017 . Available at: https://compendium.ch/mpro/mnr/26503/html/fr#7300 . Accessed March 19, 2019
  • 116 Parasrampuria DA, Truitt KE. Pharmacokinetics and pharmacodynamics of edoxaban, a non-vitamin K antagonist oral anticoagulant that inhibits clotting factor Xa. Clin Pharmacokinet 2016; 55 (06) 641-655
  • 117 Bathala MS, Masumoto H, Oguma T, He L, Lowrie C, Mendell J. Pharmacokinetics, biotransformation, and mass balance of edoxaban, a selective, direct factor Xa inhibitor, in humans. Drug Metab Dispos 2012; 40 (12) 2250-2255
  • 118 Gosselin RC, Adcock DM, Bates SM. , et al. International Council for Standardization in Haematology (ICSH) recommendations for laboratory measurement of direct oral anticoagulants. Thromb Haemost 2018; 118 (03) 437-450
  • 119 Cuker A, Husseinzadeh H. Laboratory measurement of the anticoagulant activity of edoxaban: a systematic review. J Thromb Thrombolysis 2015; 39 (03) 288-294
  • 120 Dale BJ, Chan NC, Eikelboom JW. Laboratory measurement of the direct oral anticoagulants. Br J Haematol 2016; 172 (03) 315-336
  • 121 Samuelson BT, Cuker A, Siegal DM, Crowther M, Garcia DA. Laboratory assessment of the anticoagulant activity of direct oral anticoagulants: a systematic review. Chest 2017; 151 (01) 127-138
  • 122 Douxfils J, Ageno W, Samama C-M. , et al. Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians. J Thromb Haemost 2018; 16 (02) 209-219
  • 123 Erdoes G, Martinez Lopez De Arroyabe B, Bolliger D. , et al. International consensus statement on the peri-operative management of direct oral anticoagulants in cardiac surgery. Anaesthesia 2018; 73 (12) 1535-1545
  • 124 Steiner T, Böhm M, Dichgans M. , et al. Recommendations for the emergency management of complications associated with the new direct oral anticoagulants (DOACs), apixaban, dabigatran and rivaroxaban. Clin Res Cardiol 2013; 102 (06) 399-412
  • 125 Mueck W, Kubitza D, Becka M. Co-administration of rivaroxaban with drugs that share its elimination pathways: pharmacokinetic effects in healthy subjects. Br J Clin Pharmacol 2013; 76 (03) 455-466
  • 126 Mendell J, Zahir H, Matsushima N. , et al. Drug-drug interaction studies of cardiovascular drugs involving P-glycoprotein, an efflux transporter, on the pharmacokinetics of edoxaban, an oral factor Xa inhibitor. Am J Cardiovasc Drugs 2013; 13 (05) 331-342
  • 127 Riess H, Prandoni P, Harder S, Kreher S, Bauersachs R. Direct oral anticoagulants for the treatment of venous thromboembolism in cancer patients: potential for drug-drug interactions. Crit Rev Oncol Hematol 2018; 132: 169-179

Address for correspondence

Lorenzo Alberio, MD
Division of Haematology and Central Haematology Laboratory, Centre Hospitalier Universitaire Vaudois (CHUV)
Rue du Bugnon 46, 1005 Lausanne
Switzerland   

  • References

  • 1 Mismetti P, Laporte S. New oral antithrombotics: a need for laboratory monitoring. For. J Thromb Haemost 2010; 8 (04) 621-626
  • 2 Connolly SJ, Ezekowitz MD, Yusuf S. , et al. RE-LY Steering Committee and Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 2009; 361 (12) 1139-1151
  • 3 Giugliano RP, Ruff CT, Braunwald E. , et al. ENGAGE AF-TIMI 48 Investigators. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2013; 369 (22) 2093-2104
  • 4 Patel MR, Mahaffey KW, Garg J. , et al. ROCKET AF Investigators. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med 2011; 365 (10) 883-891
  • 5 Granger CB, Alexander JH, McMurray JJV. , et al. ARISTOTLE Committees and Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2011; 365 (11) 981-992
  • 6 Reilly PA, Lehr T, Haertter S. , et al. RE-LY Investigators. The effect of dabigatran plasma concentrations and patient characteristics on the frequency of ischemic stroke and major bleeding in atrial fibrillation patients: the RE-LY Trial (Randomized Evaluation of Long-Term Anticoagulation Therapy). J Am Coll Cardiol 2014; 63 (04) 321-328
  • 7 Powell JR. Are new oral anticoagulant dosing recommendations optimal for all patients?. JAMA 2015; 313 (10) 1013-1014
  • 8 Paré G, Eriksson N, Lehr T. , et al. Genetic determinants of dabigatran plasma levels and their relation to bleeding. Circulation 2013; 127 (13) 1404-1412
  • 9 Cohen D. Dabigatran: how the drug company withheld important analyses. BMJ 2014; 349: g4670
  • 10 An idea for a mid to long term strategy for Pradaxa [Internet]. Boehringer Ingelheim International. Available at: https://www.bmj.com/investigation/dabigatran . Accessed February 26, 2019
  • 11 Safouris A, Triantafyllou N, Parissis J, Tsivgoulis G. The case for dosing dabigatran: how tailoring dose to patient renal function, weight and age could improve the benefit-risk ratio. Ther Adv Neurol Disorder 2015; 8 (06) 245-254
  • 12 FDA. Pradaxa clinical pharmacology and biopharmaceutics review(s) [Internet]. 2011 [cited 2019 Feb 25]. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2010/022512Orig1s000ClinPharmR_Corrrected%203.11.2011.pdf . Accessed February 25, 2019
  • 13 Infromation professionnelle du Compendium Suisse des Médicaments: Pradaxa [Internet]. Compendium Suisse des Médicaments. Available at: https://compendium.ch/mpro/mnr/23436/html/fr . Accessed March 5, 2019
  • 14 Šinigoj P, Malmström RE, Vene N. , et al. Dabigatran concentration: variability and potential bleeding prediction in “real-life” patients with atrial fibrillation. Basic Clin Pharmacol Toxicol 2015; 117 (05) 323-329
  • 15 Albaladejo P, Samama C-M, Sié P. , et al. GIHP-NACO Study Group. Management of severe bleeding in patients treated with direct oral anticoagulants: an observational registry analysis. Anesthesiology 2017; 127 (01) 111-120
  • 16 Volbers B, Köhrmann M, Kallmünzer B. , et al. Dabigatran plasma levels in acute cerebrovascular events. J Stroke Cerebrovasc Dis 2016; 25 (04) 877-882
  • 17 Eriksson BI, Dahl OE, Büller HR. , et al. BISTRO II Study Group. A new oral direct thrombin inhibitor, dabigatran etexilate, compared with enoxaparin for prevention of thromboembolic events following total hip or knee replacement: the BISTRO II randomized trial. J Thromb Haemost 2005; 3 (01) 103-111
  • 18 Testa S, Paoletti O, Legnani C. , et al. Low drug levels and thrombotic complications in high-risk atrial fibrillation patients treated with direct oral anticoagulants. J Thromb Haemost 2018; 16 (05) 842-848
  • 19 Chin PK, Wright DF, Patterson DM, Doogue MP, Begg EJ. A proposal for dose-adjustment of dabigatran etexilate in atrial fibrillation guided by thrombin time. Br J Clin Pharmacol 2014; 78 (03) 599-609
  • 20 Chaussade E, Hanon O, Boully C. , et al. Real-life peak and trough dabigatran plasma measurements over time in hospitalized geriatric patients with atrial fibrillation. J Nutr Health Aging 2018; 22 (01) 165-173
  • 21 Schellings MW, Boonen K, Schmitz EM. , et al. Determination of dabigatran and rivaroxaban by ultra-performance liquid chromatography-tandem mass spectrometry and coagulation assays after major orthopaedic surgery. Thromb Res 2016; 139: 128-134
  • 22 Mueck W, Stampfuss J, Kubitza D, Becka M. Clinical pharmacokinetic and pharmacodynamic profile of rivaroxaban. Clin Pharmacokinet 2014; 53 (01) 1-16
  • 23 FDA. Xarelto – clinical pharmacology & biopharmaceutical review [Internet]. FDA; 2009. . Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/022406Orig1s000ClinPharmR.pdf . Accessed February 25, 2019
  • 24 Girgis IG, Patel MR, Peters GR. , et al. Population pharmacokinetics and pharmacodynamics of rivaroxaban in patients with non-valvular atrial fibrillation: results from ROCKET AF. J Clin Pharmacol 2014; 54 (08) 917-927
  • 25 Gulilat M, Tang A, Gryn SE. , et al. Interpatient variation in rivaroxaban and apixaban plasma concentrations in routine care. Can J Cardiol 2017; 33 (08) 1036-1043
  • 26 Kanuri SH, Kreutz RP. Pharmacogenomics of novel direct oral anticoagulants: newly identified genes and genetic variants. J Pers Med 2019; 9 (01) E7
  • 27 Kubitza D, Berkowitz SD, Misselwitz F. Evidence-based development and rationale for once-daily rivaroxaban dosing regimens across multiple indications. Clin Appl Thromb Hemost 2016; 22 (05) 412-422
  • 28 Nakano Y, Kondo T, Osanai H. , et al. Clinical usefulness of measuring prothrombin time and soluble fibrin levels in Japanese patients with atrial fibrillation receiving rivaroxaban. J Cardiol 2015; 65 (03) 185-190
  • 29 Woodruff AE, Wovkulich MM, Mogle BT, Hassan AK. Association between prothrombin time and bleeding in hospitalized patients receiving rivaroxaban. Am J Health Syst Pharm 2018; 75 (22) 1783-1789
  • 30 Sakaguchi T, Osanai H, Murase Y. , et al. Monitoring of anti-Xa activity and factors related to bleeding events: A study in Japanese patients with nonvalvular atrial fibrillation receiving rivaroxaban. J Cardiol 2017; 70 (03) 244-249
  • 31 Wada S, Toyoda K, Sato S. , et al. Anti-Xa activity and event risk in patients with direct factor Xa inhibitors initiated early after stroke. Circ J 2018; 82 (11) 2872-2879
  • 32 Krause M, Henningsen A, Torge A. , et al. Impact of gender on safety and efficacy of Rivaroxaban in adolescents & young adults with venous thromboembolism. Thromb Res 2016; 148: 145-151
  • 33 Seiffge DJ, Kägi G, Michel P. , et al. Novel Oral Anticoagulants in Stroke Patients study group. Rivaroxaban plasma levels in acute ischemic stroke and intracerebral hemorrhage. Ann Neurol 2018; 83 (03) 451-459
  • 34 Zalewski J, Rychlak R, Góralczyk T, Undas A. Rivaroxaban concentration in patients with deep vein thrombosis who reported thrombus progression or minor hemorrhagic complications: first Polish experience. Pol Arch Med Wewn 2014; 124 (10) 553-555
  • 35 Romano SL, Chiarugi P, Casini M, Pellegrini G, Ruocco L. A low rivaroxaban plasma level may indicate anticoagulation undertreatment. Eur J Case Rep Intern Med 2018; 5 (11) 000937
  • 36 Ing Lorenzini K, Daali Y, Fontana P, Desmeules J, Samer C. Rivaroxaban-induced hemorrhage associated with ABCB1 genetic defect. Front Pharmacol 2016; 7: 494
  • 37 Barton J, Wong A, Graudins A. Anti-Xa activity in apixaban overdose: a case report. Clin Toxicol (Phila) 2016; 54 (09) 871-873
  • 38 Repplinger DJ, Hoffman RS, Nelson LS, Hines EQ, Howland M, Su MK. Lack of significant bleeding despite large acute rivaroxaban overdose confirmed with whole blood concentrations. Clin Toxicol (Phila) 2016; 54 (08) 647-649
  • 39 Spiller HA, Mowry JB, Aleguas Jr A. , et al. An observational study of the factor Xa inhibitors rivaroxaban and apixaban as reported to eight poison centers. Ann Emerg Med 2016; 67 (02) 189-195
  • 40 Becker RC, Alexander JH, Newby LK. , et al. Effect of apixaban, an oral and direct factor Xa inhibitor, on coagulation activity biomarkers following acute coronary syndrome. Thromb Haemost 2010; 104 (05) 976-983
  • 41 Frost C, Wang J, Nepal S. , et al. Apixaban, an oral, direct factor Xa inhibitor: single dose safety, pharmacokinetics, pharmacodynamics and food effect in healthy subjects. Br J Clin Pharmacol 2013; 75 (02) 476-487
  • 42 Eliquis: Summary of product characteristics [Internet]. European Medicines Agency. Available at: https://www.ema.europa.eu/en/documents/product-information/eliquis-epar-product-information_en.pdf . Accessed March 4, 2019
  • 43 Ueshima S, Hira D, Fujii R. , et al. Impact of ABCB1, ABCG2, and CYP3A5 polymorphisms on plasma trough concentrations of apixaban in Japanese patients with atrial fibrillation. Pharmacogenet Genomics 2017; 27 (09) 329-336
  • 44 FDA. Apixaban clinical pharmacology/biopharmaceutics review [Internet]. FDA; 2012. . Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2012/202155Orig1s000ClinPharmR.pdf . Accessed February 25, 2019
  • 45 Bhagirath VC, Eikelboom JW, Hirsh J. , et al. Apixaban-calibrated anti-FXa activity in relation to outcome events and clinical characteristics in patients with atrial fibrillation: results from the AVERROES trial. TH Open 2017; 1 (02) e139-e145
  • 46 Byon W, Sweeney K, Frost C, Boyd RA. Population pharmacokinetics, pharmacodynamics, and exploratory exposure-response analyses of apixaban in subjects treated for venous thromboembolism. CPT Pharmacometrics Syst Pharmacol 2017; 6 (05) 340-349
  • 47 Testa S, Dellanoce C, Paoletti O. , et al. Edoxaban plasma levels in patients with non-valvular atrial fibrillation: Inter and intra-individual variability, correlation with coagulation screening test and renal function. Thromb Res 2019; 175: 61-67
  • 48 FDA. Edoxaban clinical pharmacology and biopharmaceutics review [Internet]. FDA; 2014 . Available at: https://www.accessdata.fda.gov/drugsatfda_docs/nda/2015/206316Orig1Orig2s000ClinPharmR.pdf . Accessed February 25, 2019
  • 49 Weitz JI, Connolly SJ, Patel I. , et al. Randomised, parallel-group, multicentre, multinational phase 2 study comparing edoxaban, an oral factor Xa inhibitor, with warfarin for stroke prevention in patients with atrial fibrillation. Thromb Haemost 2010; 104 (03) 633-641
  • 50 Giugliano RP. Compilation of oral presentations: OC-WE-003 The relationship between oral factor Xa (FXa) inhibitor du-176b pharmacokinetics (PK) and the probability of bleeding events (BE) in patients with atrial fibrillation (AF). Boston, MA: International Society on Thrombosis and Haemostasis; 2009
  • 51 Salazar DE, Mendell J, Kastrissios H. , et al. Modelling and simulation of edoxaban exposure and response relationships in patients with atrial fibrillation. Thromb Haemost 2012; 107 (05) 925-936
  • 52 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 AF-TIMI 48 trial. Lancet 2015; 385 (9984): 2288-2295
  • 53 Yin OQP, Antman EM, Braunwald E. , et al. Linking endogenous factor Xa activity, a biologically relevant pharmacodynamic marker, to edoxaban plasma concentrations and clinical outcomes in the ENGAGE AF-TIMI 48 trial. Circulation 2018; 138 (18) 1963-1973
  • 54 Chao T-F, Chen S-A, Ruff CT. , et al. Clinical outcomes, edoxaban concentration, and anti-factor Xa activity of Asian patients with atrial fibrillation compared with non-Asians in the ENGAGE AF-TIMI 48 trial. Eur Heart J 2019; 40 (19) 1518-1527
  • 55 Conway SE, Hwang AY, Ponte CD, Gums JG. Laboratory and clinical monitoring of direct acting oral anticoagulants: what clinicians need to know. Pharmacotherapy 2017; 37 (02) 236-248
  • 56 Tamigniau A, Douxfils J, Nicolas J-B. , et al. Why, when and how to monitor new oral anticoagulants [in French]. Rev Med Suisse 2014; 10 (416) 326-333
  • 57 Moudallel S, Steurbaut S, Cornu P, Dupont A. Appropriateness of DOAC prescribing before and during hospital admission and analysis of determinants for inappropriate prescribing. Front Pharmacol 2018; 9: 1220
  • 58 Gupta M, Singh N, Tsigoulis M. , et al. Underuse of full dose factor Xa inhibition in atrial fibrillation: insight from the SPRINT-AF registry. J Am Coll Cardiol 2015; 65 (10) A348
  • 59 Wehbe RM, Yadlapati A. Underuse of oral anticoagulants for nonvalvular atrial fibrillation: past, present, and future. Tex Heart Inst J 2016; 43 (04) 287-290
  • 60 Avorn J. The psychology of clinical decision making - implications for medication use. N Engl J Med 2018; 378 (08) 689-691
  • 61 Dillinger J-G, Aleil B, Cheggour S. , et al. Dosing issues with non-vitamin K antagonist oral anticoagulants for the treatment of non-valvular atrial fibrillation: why we should not underdose our patients. Arch Cardiovasc Dis 2018; 111 (02) 85-94
  • 62 Shrestha S, Baser O, Kwong WJ. Effect of renal function on dosing of non-vitamin K antagonist direct oral anticoagulants among patients with nonvalvular atrial fibrillation. Ann Pharmacother 2018; 52 (02) 147-153
  • 63 Turpie AGG, Purdham D, Ciaccia A. Nonvitamin K antagonist oral anticoagulant use in patients with renal impairment. Ther Adv Cardiovasc Dis 2017; 11 (09) 243-256
  • 64 Lutz J, Jurk K, Schinzel H. Direct oral anticoagulants in patients with chronic kidney disease: patient selection and special considerations. Int J Nephrol Renovasc Dis 2017; 10: 135-143
  • 65 Barrios V, Górriz JL. Atrial fibrillation and chronic kidney disease: focus on rivaroxaban. J Comp Eff Res 2015; 4 (06) 651-664
  • 66 Andò 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: 162-169
  • 67 Feldberg J, Patel P, Farrell A. , et al. A systematic review of direct oral anticoagulant use in chronic kidney disease and dialysis patients with atrial fibrillation. Nephrol Dial Transplant 2019; 34 (02) 265-277
  • 68 Greenblatt DJ, Patel M, Harmatz JS, Nicholson WT, Rubino CM, Chow CR. Impaired rivaroxaban clearance in mild renal insufficiency with verapamil coadministration: potential implications for bleeding risk and dose selection. J Clin Pharmacol 2018; 58 (04) 533-540
  • 69 Becattini C, Giustozzi M, Ranalli MG. , et al. Variation of renal function over time is associated with major bleeding in patients treated with direct oral anticoagulants for atrial fibrillation. J Thromb Haemost 2018; 16 (05) 833-841
  • 70 Min M, Sibicky S. Concerns for bleeding in the elderly with the use of direct oral anticoagulants. Consult Pharm 2018; 33 (05) 262-267
  • 71 Ruff CT, Giugliano RP, Braunwald E. , et al. Comparison of the efficacy and safety of new oral anticoagulants with warfarin in patients with atrial fibrillation: a meta-analysis of randomised trials. Lancet 2014; 383 (9921): 955-962
  • 72 Eikelboom JW, Wallentin L, Connolly SJ. , et al. Risk of bleeding with 2 doses of dabigatran compared with warfarin in older and younger patients with atrial fibrillation: an analysis of the randomized evaluation of long-term anticoagulant therapy (RE-LY) trial. Circulation 2011; 123 (21) 2363-2372
  • 73 Kailas SD, Thambuluru SR. Efficacy and safety of direct oral anticoagulants compared to warfarin in prevention of thromboembolic events among elderly patients with atrial fibrillation. Cureus 2016; 8 (10) e836
  • 74 Bando S, Nishikado A, Hiura N. , et al. Efficacy and safety of rivaroxaban in extreme elderly patients with atrial fibrillation: analysis of the Shikoku Rivaroxaban Registry Trial (SRRT). J Cardiol 2018; 71 (02) 197-201
  • 75 Nissan R, Spectre G, Hershkovitz A. , et al. Apixaban levels in octogenarian patients with non-valvular atrial fibrillation. Drugs Aging 2019; 36 (02) 165-177
  • 76 Khan F, Huang H, Datta YH. Direct oral anticoagulant use and the incidence of bleeding in the very elderly with atrial fibrillation. J Thromb Thrombolysis 2016; 42 (04) 573-578
  • 77 Levy JH, Ageno W, Chan NC, Crowther M, Verhamme P, Weitz JI. ; Subcommittee on Control of Anticoagulation. When and how to use antidotes for the reversal of direct oral anticoagulants: guidance from the SSC of the ISTH. J Thromb Haemost 2016; 14 (03) 623-627
  • 78 Albaladejo P, Pernod G, Godier A. , et al. Members of the French Working Group on Perioperative Haemostasis. Management of bleeding and emergency invasive procedures in patients on dabigatran: updated guidelines from the French Working Group on Perioperative Haemostasis (GIHP) - September 2016. Anaesth Crit Care Pain Med 2018; 37 (04) 391-399
  • 79 Steffel J, Verhamme P, Potpara TS. , et al. ESC Scientific Document Group. The 2018 European Heart Rhythm Association Practical Guide on the use of non-vitamin K antagonist oral anticoagulants in patients with atrial fibrillation. Eur Heart J 2018; 39 (16) 1330-1393
  • 80 Kaatz S, Mahan CE, Nakhle A. , et al. Management of elective surgery and emergent bleeding with direct oral anticoagulants. Curr Cardiol Rep 2017; 19 (12) 124
  • 81 Prisco D, Ageno W, Becattini C. , et al. SIMI (Italian Society of Internal Medicine); FADOI (Federation of Associations of Hospital Doctors on Internal Medicine); SISET (Italian Society for the Study of Haemostasis and Thrombosis). Italian intersociety consensus on DOAC use in internal medicine. Intern Emerg Med 2017; 12 (03) 387-406
  • 82 Douketis JD, Spyropoulos AC, Anderson JM. , et al. The Perioperative Anticoagulant Use for Surgery Evaluation (PAUSE) study for patients on a direct oral anticoagulant who need an elective surgery or procedure: design and rationale. Thromb Haemost 2017; 117 (12) 2415-2424
  • 83 Godier A, Dincq A-S, Martin A-C. , et al. Predictors of pre-procedural concentrations of direct oral anticoagulants: a prospective multicentre study. Eur Heart J 2017; 38 (31) 2431-2439
  • 84 Pernod G, Albaladejo P, Godier A. , et al. Management of major bleeding complications and emergency surgery in patients on long-term treatment with direct oral anticoagulants, thrombin or factor-Xa inhibitors. Proposals of the Working Group on Perioperative Haemostasis (GIHP) - March 2013 [in French]. Ann Fr Anesth Reanim 2013; 32 (10) 691-700
  • 85 Bertaggia-Calderara D, Kröll D, Gerschheimer C. , et al. Effect of rivaroxaban on thrombin generation in vivo. A study in obese patients. Int J Lab Hematol 2018; 40 (01) e11-e14
  • 86 Graff J, Harder S. Anticoagulant therapy with the oral direct factor Xa inhibitors rivaroxaban, apixaban and edoxaban and the thrombin inhibitor dabigatran etexilate in patients with hepatic impairment. Clin Pharmacokinet 2013; 52 (04) 243-254
  • 87 Hoolwerf EW, Kraaijpoel N, Büller HR, van Es N. Direct oral anticoagulants in patients with liver cirrhosis: a systematic review. Thromb Res 2018; 170: 102-108
  • 88 Ha NB, Regal RE. Anticoagulation in patients with cirrhosis: caught between a rock-liver and a hard place. Ann Pharmacother 2016; 50 (05) 402-409
  • 89 Priyanka P, Kupec JT, Krafft M, Shah NA, Reynolds GJ. Newer oral anticoagulants in the treatment of acute portal vein thrombosis in patients with and without cirrhosis. Int J Hepatol 2018; 2018: 8432781
  • 90 Chang SH, Chou IJ, Yeh YH. , et al. Association between use of non-vitamin K oral anticoagulants with and without concurrent medications and risk of major bleeding in nonvalvular atrial fibrillation. JAMA 2017; 318 (13) 1250-1259
  • 91 Patel JP, Roberts LN, Arya R. Anticoagulating obese patients in the modern era. Br J Haematol 2011; 155 (02) 137-149
  • 92 Kubitza D, Becka M, Zuehlsdorf M, Mueck W. Body weight has limited influence on the safety, tolerability, pharmacokinetics, or pharmacodynamics of rivaroxaban (BAY 59-7939) in healthy subjects. J Clin Pharmacol 2007; 47 (02) 218-226
  • 93 Upreti VV, Wang J, Barrett YC. , et al. Effect of extremes of body weight on the pharmacokinetics, pharmacodynamics, safety and tolerability of apixaban in healthy subjects. Br J Clin Pharmacol 2013; 76 (06) 908-916
  • 94 Di Minno MND, Lupoli R, Di Minno A, Ambrosino P, Scalera A, Dentali F. Effect of body weight on efficacy and safety of direct oral anticoagulants in the treatment of patients with acute venous thromboembolism: a meta-analysis of randomized controlled trials. Ann Med 2015; 47 (01) 61-68
  • 95 Boonyawat K, Caron F, Li A. , et al. Association of body weight with efficacy and safety outcomes in phase III randomized controlled trials of direct oral anticoagulants: a systematic review and meta-analysis. J Thromb Haemost 2017; 15 (07) 1322-1333
  • 96 Pradaxa: summary of product characteristics [Internet]. European Medicines Agency; Available at: https://www.ema.europa.eu/documents/product-information/pradaxa-epar-product-information_en.pdf . Accessed March 9, 2020
  • 97 Safouris A, Demulder A, Triantafyllou N, Tsivgoulis G. Rivaroxaban presents a better pharmacokinetic profile than dabigatran in an obese non-diabetic stroke patient. J Neurol Sci 2014; 346 (1–2): 366-367
  • 98 Güler E, Babur Güler G, Demir GG, Hatipoğlu S. A review of the fixed dose use of new oral anticoagulants in obese patients: Is it really enough?. Anatol J Cardiol 2015; 15 (12) 1020-1029
  • 99 Kröll D, Stirnimann G, Vogt A. , et al. Pharmacokinetics and pharmacodynamics of single doses of rivaroxaban in obese patients prior to and after bariatric surgery. Br J Clin Pharmacol 2017; 83 (07) 1466-1475
  • 100 Kröll D, Nett PC, Borbély YM. , et al. The effect of bariatric surgery on the direct oral anticoagulant rivaroxaban: the extension study. Surg Obes Relat Dis 2018; 14 (12) 1890-1896
  • 101 Hu YF, Liao JN, Chern CM. , et al. Identification and management of noncompliance in atrial fibrillation patients receiving dabigatran: the role of a drug monitor. Pacing Clin Electrophysiol 2015; 38 (04) 465-471
  • 102 Keita I, Aubin-Auger I, Lalanne C. , et al. Assessment of quality of life, satisfaction with anticoagulation therapy, and adherence to treatment in patients receiving long-course vitamin K antagonists or direct oral anticoagulants for venous thromboembolism. Patient Prefer Adherence 2017; 11: 1625-1634
  • 103 Ombandza-Moussa E, Samama MM, Horellou MH, Chatelier AL, Elalamy I, Conard J. Influence of oral anticoagulant treatment on D-dimers levels [in French]. Ann Biol Clin (Paris) 2001; 59 (05) 579-583
  • 104 van Ryn J, Stangier J, Haertter S. , et al. Dabigatran etexilate--a novel, reversible, oral direct thrombin inhibitor: interpretation of coagulation assays and reversal of anticoagulant activity. Thromb Haemost 2010; 103 (06) 1116-1127
  • 105 Xarelto: summary of product characteristics [Internet]. European Medicines Agency. Available at: https://www.ema.europa.eu/en/documents/product-information/xarelto-epar-product-information_en.pdf . Accessed March 5, 2019
  • 106 Beyer J, Trujillo T, Fisher S, Ko A, Lind SE, Kiser TH. Evaluation of a heparin-calibrated antifactor Xa assay for measuring the anticoagulant effect of oral direct Xa inhibitors. Clin Appl Thromb Hemost 2016; 22 (05) 423-428
  • 107 Verhamme P, Wells PS, Segers A. , et al. Dose reduction of edoxaban preserves efficacy and safety for the treatment of venous thromboembolism. An analysis of the randomised, double-blind HOKUSAI VTE trial. Thromb Haemost 2016; 116 (04) 747-753
  • 108 Liesenfeld KH, Lehr T, Dansirikul C. , et al. Population pharmacokinetic analysis of the oral thrombin inhibitor dabigatran etexilate in patients with non-valvular atrial fibrillation from the RE-LY trial. J Thromb Haemost 2011; 9 (11) 2168-2175
  • 109 Stangier J, Clemens A. Pharmacology, pharmacokinetics, and pharmacodynamics of dabigatran etexilate, an oral direct thrombin inhibitor. Clin Appl Thromb Hemost 2009; 15 (Suppl. 01) 9S-16S
  • 110 Information professionnelle du Compendium Suisse des Médicaments: Xarelto [Internet]. Compendium Suisse des Médicaments; 2018 . Available at: https://compendium.ch/mpro/mnr/20753/html/fr?Platform=Desktop#7600 . Accessed February 25, 2019
  • 111 Kreutz R. Pharmacodynamic and pharmacokinetic basics of rivaroxaban. Fundam Clin Pharmacol 2012; 26 (01) 27-32
  • 112 Information professionnelle du Compendium Suisse des Médicaments: Eliquis [Internet]. Compendium Suisse des Médicaments; 2019 . Available at: https://compendium.ch/mpro/mnr/22732/html/fr#7300 . Accessed March 19, 2019
  • 113 Kubisz P, Stanciakova L, Dobrotova M, Samos M, Mokan M, Stasko J. Apixaban - metabolism, pharmacologic properties and drug interactions. Curr Drug Metab 2017; 18 (07) 609-621
  • 114 Lixiana: summary of product characteristics [Internet]. European Medicines Agency. Available at: https://www.ema.europa.eu/en/documents/product-information/lixiana-epar-product-information_en.pdf . Accessed March 19, 2019
  • 115 Information professionnelle du Compendium Suisse des Médicaments: Lixiana [Internet]. Compendium Suisse des Médicaments; 2017 . Available at: https://compendium.ch/mpro/mnr/26503/html/fr#7300 . Accessed March 19, 2019
  • 116 Parasrampuria DA, Truitt KE. Pharmacokinetics and pharmacodynamics of edoxaban, a non-vitamin K antagonist oral anticoagulant that inhibits clotting factor Xa. Clin Pharmacokinet 2016; 55 (06) 641-655
  • 117 Bathala MS, Masumoto H, Oguma T, He L, Lowrie C, Mendell J. Pharmacokinetics, biotransformation, and mass balance of edoxaban, a selective, direct factor Xa inhibitor, in humans. Drug Metab Dispos 2012; 40 (12) 2250-2255
  • 118 Gosselin RC, Adcock DM, Bates SM. , et al. International Council for Standardization in Haematology (ICSH) recommendations for laboratory measurement of direct oral anticoagulants. Thromb Haemost 2018; 118 (03) 437-450
  • 119 Cuker A, Husseinzadeh H. Laboratory measurement of the anticoagulant activity of edoxaban: a systematic review. J Thromb Thrombolysis 2015; 39 (03) 288-294
  • 120 Dale BJ, Chan NC, Eikelboom JW. Laboratory measurement of the direct oral anticoagulants. Br J Haematol 2016; 172 (03) 315-336
  • 121 Samuelson BT, Cuker A, Siegal DM, Crowther M, Garcia DA. Laboratory assessment of the anticoagulant activity of direct oral anticoagulants: a systematic review. Chest 2017; 151 (01) 127-138
  • 122 Douxfils J, Ageno W, Samama C-M. , et al. Laboratory testing in patients treated with direct oral anticoagulants: a practical guide for clinicians. J Thromb Haemost 2018; 16 (02) 209-219
  • 123 Erdoes G, Martinez Lopez De Arroyabe B, Bolliger D. , et al. International consensus statement on the peri-operative management of direct oral anticoagulants in cardiac surgery. Anaesthesia 2018; 73 (12) 1535-1545
  • 124 Steiner T, Böhm M, Dichgans M. , et al. Recommendations for the emergency management of complications associated with the new direct oral anticoagulants (DOACs), apixaban, dabigatran and rivaroxaban. Clin Res Cardiol 2013; 102 (06) 399-412
  • 125 Mueck W, Kubitza D, Becka M. Co-administration of rivaroxaban with drugs that share its elimination pathways: pharmacokinetic effects in healthy subjects. Br J Clin Pharmacol 2013; 76 (03) 455-466
  • 126 Mendell J, Zahir H, Matsushima N. , et al. Drug-drug interaction studies of cardiovascular drugs involving P-glycoprotein, an efflux transporter, on the pharmacokinetics of edoxaban, an oral factor Xa inhibitor. Am J Cardiovasc Drugs 2013; 13 (05) 331-342
  • 127 Riess H, Prandoni P, Harder S, Kreher S, Bauersachs R. Direct oral anticoagulants for the treatment of venous thromboembolism in cancer patients: potential for drug-drug interactions. Crit Rev Oncol Hematol 2018; 132: 169-179

Zoom Image
Fig. 1 Proposed DOACs through level interpretation. In green: estimated median concentration target. In yellow: estimated target range. In orange: increased risk of adverse event. In red: highly increased risk of adverse event. DOAC, direct oral anticoagulant.
Zoom Image
Fig. 2 Proposed DOAC monitoring algorithm. In green: estimated median concentration target. In yellow: estimated target range. In orange: increased risk of adverse event. In red: highly increased risk of adverse event (see [Fig. 1] for references). DOAC, direct oral anticoagulant.