Skip to main content
Erschienen in: BMC Cardiovascular Disorders 1/2020

Open Access 01.12.2020 | Research article

Characteristics and outcomes in patients with atrial fibrillation receiving direct oral anticoagulants in off-label doses

verfasst von: Alexandros Briasoulis, Yubo Gao, Chakradhari Inampudi, Paulino Alvarez, Rabea Asleh, Elizabeth Chrischilles, Enrique C. Leira, Mary Vaughan-Sarrazin

Erschienen in: BMC Cardiovascular Disorders | Ausgabe 1/2020

Abstract

Background

We evaluated adherence to dosing criteria for patients with atrial fibrillation (AF) taking dabigatran or rivaroxaban and the impact of off-label dosing on thromboembolic and bleeding risk.

Methods

We used data for a retrospective cohort from a large U.S. health plan for Medicare beneficiaries age > =65 years with AF who initiated dabigatran or rivaroxaban during 2010–2016. Stroke and major bleeding were quantified in patients who were eligible for low dose but received standard dose, and in patients who were eligible for standard dose but received low dose.

Results

We identified 8035 and 19,712 patients who initiated dabigatran or rivaroxaban, respectively. Overall, 1401 (17.4%) and 7820 (39.7%) patients who received dabigatran and rivaroxaban met criteria for low dose, respectively. Of those, 959 (68.5%) and 3904 (49.9%) received standard dose. In contrast, 1013 (15.3%) and 2551 (21.5%) of patients eligible for standard dose dabigatran and rivaroxaban received low dose. Mean follow-up for patients eligible for low and standard dose dabigatran and rivaroxaban were 13.9, 15.1, 10.1, and 12.3 months, respectively. In unadjusted analyses, patients eligible for low or standard dose dabigatran and rivaroxaban but receiving off-label dose, had no differences in the rates of ischemic stroke. Among patients who met criteria for standard dose direct oral anticoagulants (DOAC), use of low dose was associated with significantly higher risk of any major bleeding (Dabigatran: HR = 1.44; 95% CI 1.14–1.8, P = 0.002, Rivaroxaban HR 1.34, 95% CI 1.11–1.6, P = 0.002) and gastrointestinal bleeding (Dabigatran: HR = 1.48; 95% CI 1.08–2, P = 0.016). In patients who met criteria for low dose DOACs, there was lower risk of major bleeding (Dabigatran: HR = 0.59; 95% CI 0.43–0.8, P < 0.001), gastrointestinal (Rivaroxaban: HR 0.79; 95% CI 0.64–0.98, P = 0.03) and intracranial bleeding (Dabigatran: HR = 0.33; 95% CI 0.12–0.9, P = 0.001) with standard dosing. After propensity matching, use of off-label doses was not associated with stroke, major, gastrointestinal or intracranial bleeding for either dabigatran or rivaroxaban.

Conclusions

While a significant number of patients receive higher or lower dose of dabigatran and rivaroxaban than recommended, we found no evidence of significant impact on thromboembolic or hemorrhagic outcomes.
Hinweise

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12872-020-01340-4.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ACE
Angiotensin-converting-enzyme
AF
Atrial fibrillation
CI
Confidence intervals
CKD
Chronic kidney disease
CrCL
Creatinine clearance)
DOACs
Direct oral anticoagulants
eGFR
Estimated glomerular filtration rate
FDA
Food and Drug Administration
ICD-9-CM
International classification of diseases–ninth revision–clinical modification
ICH
Intracranial hemorrhage
MDRD
Modification of diet in renal disease
NKDEP
National Kidney Disease Education Program
P-gp
P-glycoprotein
P-gp-Cyp3A4
P-gp and cytochrome-3A4
RCT
Randomized controlled trials
RR
Relative rates
VKA
Vitamin K antagonist

Background

Patients with Atrial fibrillation (AF) have a higher risk for stroke or systemic embolism, death and disability [1]. Oral anticoagulants, either vitamin K antagonist (VKA) or direct oral anticoagulants (DOACs) reduce that thromboembolic risk by about two-thirds irrespective of baseline risk [2]. However, the use of anticoagulation is associated with increased risk of bleeding, with intracranial hemorrhage (ICH) being the most serious bleeding complication [3]. Randomized controlled trials (RCT) of DOACs [Dabigatran, Rivaroxaban, Apixaban, and Edoxaban] have demonstrated similar protection against ischemic stroke but lower rates of ICH compared with VKAs [47].
The RCTs of DOACs in AF patients used dose adjustments based on patient characteristics such age, weight, renal function and the use of concomitant medications. A reduced dose of 75 mg twice daily of dabigatran is recommended to decrease bleeding risk in patients with creatinine clearance (CrCl) 15–30 mL/minute, or co- administration of a strong P-glycoprotein [P-gp] inhibitor (e.g., dronedarone) in patients with CrCl 30–50 mL/minute [8, 9]. With regards to rivaroxaban, a dose reduction to 15 mg daily is recommended in patients with CrCl 15–50 mL/minute, and concomitant use of a dual P-gp and cytochrome-3A4 [P-gp-Cyp3A4] inhibitor should be avoided to prevent potential increased rivaroxaban concentration [10]. Administration of lower apixaban dose 2.5 mg twice daily is indicated if 2 of the following 3 criteria are met: age > 80 years, weight < than 60 kg, and serum Creatinine > 1.5 mg/dl [11].
Since the use of DOACs became widespread, deviations from the recommended dosing are not infrequent [12, 13]. Analysis of 5738 patients treated with DOACs from the ORBITA-AF II registry showed that 9.4% of patients were under-dosed and 3.4% were overdosed. Overdosing was associated with significantly increased all-cause mortality whereas under-dosing was associated with increased cardiovascular hospitalizations [14]. A subsequent analysis of 7925 AF patients treated with DOACs from the same registry showed that 16% of patients were on reduced doses with many of these doses adjustments (57%) not following the recommended doses [15]. Nevertheless, after risk- adjustment, the use of lower-than-recommended dose resulted in similar thromboembolic and bleeding risk compared to appropriately dosed DOAC use [15].
We hypothesized that a sizeable number of DOAC prescriptions do not adhere to the Food and Drug Administration (FDA) dosing criteria and may increase thromboembolic and bleeding events. The purpose of our study was to: 1) examine characteristics and predictors of low dose use among patients who meet FDA criteria for standard dose, or standard dose use among patients who meet FDA criteria for low dose, among patients who initiate dabigatran and rivaroxaban, ii) compare the risk of ischemic stroke and bleeding events in patients receiving off-label low dose or off-label standard dose to patients receiving FDA-recommended doses in a community-based sample of elderly Medicare beneficiaries with AF enrolled in a large U.S. health plan.

Methods

Data source

We designed a new user retrospective cohort study using data for Medicare beneficiaries enrolled in a large U.S. health plan with prescription drug coverage. Medical (inpatient visit, outpatient physician visits) and pharmacy claims with detailed prescription fill information from October 1, 2010 through December 31, 2016 were analyzed. In addition, the data also includes laboratory test results (such as serum creatinine) for Medicare beneficiaries enrolled in managed care plans. The study was non-human subject research by the University of Iowa institutional review board because it involved analysis of an existing database that was fully de-identified.

Patient population

We analyzed claims for Medicare beneficiaries age > = 65 years with newly diagnosed atrial fibrillation (AF) between 2010 and 2016 (dabigatran was approved by FDA in October 2010 followed by rivaroxaban approval in November 2011). We identified patients who initiated dabigatran 150 mg twice daily (standard dose) or 75 mg twice daily (low dose), or rivaroxaban 20 mg daily (standard dose) or 15 mg daily. We did not include patients receiving apixaban as information to assess dosing criteria such as patient weight was not available in our data, and we did not include patients receiving edoxaban due to relatively low use of this drug during our time frame. Patients were categorized into mutually exclusive groups according to the first DOAC and DOAC dose received.
Patients were excluded if they did not have a diagnosis of AF during the 12 months prior to initiating the DOAC, where AF was defined as International Classification of Diseases–Ninth Revision–Clinical Modification [ICD-9-CM] code 427.31 or ICD-Tenth Revision [ICD-10] code I48.0, I48.1, I48.2, I48.3, I48.4, or I48.91, as primary or secondary diagnosis. Additionally, we excluded patients who: i) were younger than 65 years at the time of diagnosis, ii) had a diagnosis indicating pulmonary embolism or deep vein thrombosis within 8 weeks prior to initiating the DOAC, iii) underwent hip surgery within 6 weeks prior to initiating the DOAC, or iv) were not enrolled in the health care plan for at least 1 year prior to initiating the DOAC.
Patients who initiated dabigatran or rivaroxaban were categorized to approximate dosing criteria, using the most recent estimated glomerular filtration rate (eGFR) available prior to initiating the DOAC as a proxy for creatinine clearance. eGFR was calculated based on Modification of Diet in Renal Disease (MDRD) Study equation or Chronic Kidney Disease Epidemiology Collaboration (CKD-EPI) equation [16, 17]. Patients who received dabigatran were deemed to meet criteria for low dose if they had severe renal disease (defined as eGFR < 30 mL/minute/ 1.73 m2) or had moderate renal disease and concurrent use of a p-gp inhibitor (where moderate renal disease was defined as eGFR 30–50 mL/minute/1.73 m2 and p-gp inhibitors included dronedarone, cyclosporine, itraconazole, tacrolimus, ketoconazole). Patients who received rivaroxaban were deemed to meet criteria for low dose if they had eGFR< 50 or concomitant use of a dual P-gp-Cyp3A4 inhibitor (including ketoconazole, fluconazole, itraconazole, cobicistat, conivaptan, indinavir, voriconazole, posaconazole, nefazodone HCL, ritonavir, saquinavir, telithromycin). Patients with no valid GFR for assessing renal function were excluded. Among patients eligible for low doses of dabigatran or rivaroxaban, 87.4 and 86.5% had available eGFR whereas among those eligible for standard dose dabigatran or rivaroxaban, 54.9 and 56% had available eGFR respectively. We performed sensitivity analysis for DOAC dose adjustments based on eGFR only, excluding drug interactions.

Covariates

Data on patient-level characteristics such as demographics, comorbid conditions, concurrent medication use, and prior health services utilization were extracted from health care plan enrollment data and inpatient, outpatient, and physician claims. Comorbid diseases were identified by ICD-9-CM/ICD-10 diagnoses on claims during the 12 months preceding the date of first DOAC fill. We used the Charlson Comorbidity Index to estimate patients’ overall comorbidity status [18]. We also identified all conditions in the CHA2DS2-VASc stroke risk score (congestive heart failure diagnosis, female sex, hypertension diagnosis, diabetes, age, prior stroke or transient ischemic attack, and vascular disease diagnosis). History of major bleeding was defined as any prior major bleeding, gastrointestinal bleeding, intracranial hemorrhage, or prior receipt of transfusion. Additional conditions included liver disease, alcohol abuse, obesity, chronic obstructive pulmonary disease, peripheral vascular disease, pulmonary circulation disease, heart valve disease, history of coronary revascularization, history of implantable cardiac device, and prior pulmonary embolism or deep vein thrombosis. We also extracted data on medication use at the time of DOAC initiation (p-glycoprotein inhibitors, cytochrome 3A4 inhibitors, insulin, statins, beta blockers, angiotensin-converting-enzyme (ACE) inhibitors, angiotensin-receptor blockers (ARBs), calcium channel blockers, prescription antiplatelets (e.g., clopidogrel), proton pump inhibitors and non-steroidal anti-inflammatory drugs. (A list of included drugs and ICD9/ICD10 codes for comorbid conditions is provided in Additional file 1: Table S1). Medications were considered concomitant if the patient had supply within 90 days from the DOAC prescription. We also identified patients with a history of warfarin use prior to initiating the DOAC.

Endpoints

We selected the following clinical endpoints: i) ischemic stroke, ii) any major bleeding, iii) gastrointestinal bleeding (GI), iv) intracranial bleeding based on the primary ICD-9-CM/ICD-10 diagnosis on inpatient claims for acute care stays (definitions provided in Additional file 1: Table S1). We also examined drug discontinuation, as defined by the date of last fill for the original DOAC and dose. Patients were followed from the date of the initial DOAC prescription until December 31, 2016 or lapse of health plan enrollment (due to death or other reason), or cessation of the initial DOAC dose.

Statistical analysis

Analyses were conducted separately for patients who initiated dabigatran and rivaroxaban, and by dose eligibility. For example, among patients who received dabigatran and met criteria for low dose dabigatran, we compared characteristics and outcomes among patients who received standard dose in contrast to dosing criteria vs. those who received the recommended low dose. Similarly, among patients who received dabigatran and met criteria for standard dose, we compared patients who received low dose vs. those who received the recommended standard dose. We compared demographic characteristics, comorbid diseases, and concurrent medication use among patients on different doses using the chi-square test.
We examined rates of ischemic stroke, any major bleeding, GI hemorrhage, and intracranial hemorrhage per patient-year of follow-up in the full sample and in propensity matched patients. Specifically, we performed 2-way nearest-neighbor propensity-matching to create groups of patients receiving low dose or standard dose that were balanced with respect to observed patient characteristics. Propensity matching was conducted separately for patients qualifying for low or standard dose, and for patients on dabigatran or rivaroxaban (i.e., four separate propensity matched samples). We assessed covariate balance in propensity-matched samples using standardized differences between patients receiving low or standard dose, where differences less than 10% indicate satisfactory balance. Because standardized differences remained greater than 10% for a small number of covariates, we further adjusted for unbalanced covariates if they were related to the outcome of interest using Cox proportional hazards regression models. We then calculated rates of each endpoint per patient year of follow-up in unmatched and propensity-matched samples. Statistical significance was assessed using Cox proportional hazards regression models that censored for end of follow-up (December 31, 2016), medication cessation, or disenrollment from the managed care plan for any reason. Results were reported as hazard ratios (HR) with 95% confidence intervals (CI) for each drug and recommended dose, where the HR reflects the outcome rate for off-label dosing relative to FDA-recommended dose. Finally, one sensitivity analysis was performed in which drug dosing criteria was based on GFR measurements only, without considering pharmacologic interactions reflected in FDA criteria.
All analyses were conducted using SAS with 2-tailed level of significance set at 0.05.

Results

Patient characteristic

We identified 8035 patients with valid GFR measurements including 6580 on standard dabigatran dose and 1455 patients on low dose (18.1%); 19,712 patients were rivaroxaban including 13,245 on standard dose rivaroxaban and 6467 on low dose (32.8%). We identified significant differences in baseline characteristics between different does of dabigatran and rivaroxaban before propensity matching (Tables 1 and 2). Among dabigatran patients, those taking low dose were more likely to be over 75 years (P < 0.001), female (P = 0.01), have CKD III or more advanced (P < 0.001) and higher burden of comorbidities as indicated by higher Charlson comorbidity index (low dose: 5.52 ± 3.75 vs. high dose: 4.17 ± 3.47, P < 0.001). Among rivaroxaban patients, low dose was more frequently prescribed to patients over 75 years (P < 0.001), females (P = 0.03), patients with CKD III or more advanced (P < 0.001) and patients with higher comorbidity burden (low dose: 5.05 ± 3.65 vs. high dose: 3.71 ± 3.34, P < 0.001).
Table 1
Characteristics of patients taking standard (150 mg) or reduced (75 mg) dose dabigatran
 
Reduced dose (75 mg twice daily)
Standard Dose (150 mg twice daily)
P-value
Total number of patients
1455
6580
 
Age Category, years
  
< 0.001
 65–69
7.1%
18.0%
 
 70 to 74
13.7%
26.8%
 
 75 to 79
18.8%
25.4%
 
 80 to 84
26.0%
18.1%
 
 85 to 89
16.8%
7.1%
 
 90 or over
17.5%
4.7%
 
Sex
  
< 0.001
 Female
56.3%
47.9%
 
 Male
43.7%
52.1%
 
Race Category
  
< 0.001
 White
84.1%
87.5%
 
 Black
10.2%
6.9%
 
 Hispanic
2.5%
1.7%
 
 Asian
1.4%
0.6%
 
 Other
1.9%
3.2%
 
 Smoking
14.0%
16.7%
0.01
 Alcohol use
2.0%
2.3%
< 0.001
Weight Category (based on ICD-9 and ICD-10 codes for BMI Category)
  
< 0.001
 Under-Weight
1.2%
0.6%
 
 Healthy or Overweight
10.1%
8.0%
 
 Obese or Severe Obese
20.1%
23.9%
 
 Not available
68.5%
67.5%
 
Comorbid Conditions
 Prior Stroke
26.8%
24.4%
0.06
 Prior major bleeding from Diagnosis
26.7%
24.5%
0.07
 Gastrointestinal bleeding
0
13.0%
0.24
 Cerebral bleeding
0
0.8%
0.7
 Diabetes
50.7%
48.9%
0.2
 Prior AMI
8.0%
5.2%
< 0.001
 Liver Disease
3.4%
2.6%
0.13
 Heart Failure
48.6%
31.3%
< 0.001
 Hypertension
96.0%
93.4%
< 0.001
 Ischemic cardiomyopathy
55.5%
48.0%
< 0.001
 Pulmonary
16.6%
11.4%
< 0.001
 COPD
36.4%
31.8%
< 0.001
 Transfusion from Procedure
3.8%
2.7%
0.02
 Revascularization
17.3%
15.0%
0.03
 Implantable Devices
16.9%
13.8%
0.002
 Valve Disease
44.7%
40.3%
0.002
Renal Disease (ICD-9 and ICD-10 codes)
  
< 0.001
 None or Mild (Stage I, II)
53.4%
81.7%
 
 Moderate (Stage III)
36.0%
16.5%
 
 Severe (Stage IV, V)
10.6%
1.7%
 
Concurrent Drugs (+/− 90 days of initiating DOAC)
 SSRI/SNRI
33.5%
31.0%
0.05
 Strong and moderate P-GP inhibitors
21.4%
22.7%
0.28
 P-GP inducers
16.4%
18.8%
0.03
 Strong P-GP and CYP3A4 dual inhibitors
22.7%
22.5%
0.8
 P-GP and CYP3A4 inducers
16.4%
18.8%
0.03
 ACE inhibitors
70.1%
68.0%
0.12
 Warfarin
20.8%
25.0%
< 0.001
 Angiotensin receptor blockers
41.0%
39.3%
0.2
 Beta blockers
92.1%
89.1%
< 0.001
 Calcium channel blockers
66.8%
63.8%
0.03
 Digoxin
31.1%
29.4%
0.2
 Proton pump inhibitors
60.5%
56.4%
0.004
 NSAIDs
49.6%
51.2%
0.29
 Antiplatelets
32.0%
26.5%
< 0.001
 Insulin
17.0%
12.9%
< 0.001
 Statins
79.6%
79.4%
0.86
 Antiarrhythmics
47.3%
48.0%
0.6
Estimated Glomerular Filtration rate (ml/min/1.73 m2)
  
< 0.001
  < 30
12.3%
2.5%
 
 30–60
62.2%
45.8%
 
 60–90
23.8%
46.0%
 
> = 90
1.8%
5.7%
 
CCI(Charlson Comorbodity Index), mean (standard deviation)
5.52 (3.75)
4.17(3.47)
< 0.001
Abbreviations: ACE Angiotensin converting enzyme, AMI Acute myocardial infarction, COPD Chronic obstructive pulmonary disease, ICD International classification of diseases, NSAIDS Non-steroidal antiinflammatory drugs, P-GP P-glycoprotein, CYP Cytochrome
Table 2
Characteristics of patients taking standard (20 mg) or reduced (15 mg) dose rivaroxaban
 
Reduced Dose
(15 mg daily)
Standard Dose
(20 mg daily)
P-value
Total number of patients
6467
13,245
 
Age Category, years
  
< 0.001
 65–69
7.9%
18.2%
 
 70 to 74
14.8%
29.0%
 
 75 to 79
20.7%
25.6%
 
 80 to 84
25.4%
17.0%
 
 85 to 89
19.8%
7.8%
 
 90 or over
11.4%
2.4%
 
Sex
  
< 0.001
 Female
57.1%
46.2%
 
 Male
42.9%
53.8%
 
Race Category
  
< 0.001
 White
85.2%
85.8%
 
 Black
9.0%
8.0%
 
 Hispanic
2.2%
1.6%
 
 Asian
0.8%
0.8%
 
 Other
2.8%
3.8%
 
 Smoking
20.2%
22.8%
< 0.001
 Alcohol
2.3%
3.2%
< 0.001
Weight Category (based on ICD-9 and ICD-10 codes for BMI Category)
  
< 0.001
 Under-Weight
2.3%
1.3%
 
 Healthy or Overweight
19.1%
14.3%
 
 Obese or Severe Obese
26.2%
31.2%
 
 Not available
52.4%
53.2%
 
Comorbid Conditions
 Prior Stroke
28.2%
22.4%
< 0.001
 Prior major bleeding from Diagnosis
26.6%
23.9%
< 0.001
 Gastrointestinal bleeding
15.2%
13.5%
0.002
 Cerebral bleeding
1.3%
0.8%
< 0.001
 Diabetes
51.0%
47.7%
< 0.001
 Prior AMI
9.0%
5.7%
< 0.001
 Liver Disease
2.9%
3.1%
0.35
 Heart Failure
35.6%
24.3%
< 0.001
 Hypertension
94.6%
92.1%
< 0.001
 Ischemic cardiomyopathy
47.7%
40.2%
< 0.001
 Pulmonary
15.9%
11.1%
< 0.001
 COPD
36.3%
31.4%
< 0.001
 Transfusion from Procedure
5.0%
2.3%
< 0.001
 Revascularization
16.5%
13.7%
< 0.001
 Implantable Devices
16.7%
12.5%
< 0.001
 Valve Disease
40.6%
36.4%
0.002
Renal Disease (ICD-9 and ICD-10 codes)
  
< 0.001
 None or Mild (Stage I, II)
54.8%
83.9%
 
 Moderate (Stage III)
37.5%
14.5%
 
 Severe (Stage IV, V)
7.7%
1.5%
 
Concurrent Drugs (+/− 90 days of initiating DOAC)
 SSRI/SNRI
33.7%
29.1%
< 0.001
 Strong and moderate P-GP inhibitors
18.3%
18.5%
0.63
 P-GP inducers
18.5%
16.5%
< 0.001
 Strong P-GP and CYP3A4 dual inhibitors
22.7%
20.8%
0.002
 P-GP and CYP3A4 inducers
18.5%
16.5%
< 0.001
 ACE inhibitors
67.4%
64.3%
< 0.001
 Warfarin
15.0%
16.1%
0.045
 Angiotensin receptor blockers
40.0%
36.7%
< 0.001
 Beta blockers
88.1%
86.5%
0.015
 Calcium channel blockers
64.5%
59.4%
< 0.001
 Digoxin
23.1%
20.8%
< 0.001
 Proton pump inhibitors
58.3%
52.3%
< 0.001
 NSAIDs
52.9%
50.1%
< 0.001
 Antiplatelets
30.2%
24.3%
< 0.001
 Insulin
14.3%
10.8%
< 0.001
 Statins
79.3%
77.6%
0.006
 Antiarrhythmics
40.4%
43.1%
< 0.001
Estimated Glomerular Filtration rate (ml/min/1.73 m2)
  
< 0.001
  < 30
7.5%
1.3%
 
 30–60
61.1%
35.2%
 
 60–90
28.8%
56.7%
 
  > =90
2.6%
6.8%
 
CCI(Charlson Comorbodity Index), mean (standard deviation)
5.05(3.65)
3.71(3.34)
< 0.001
Abbreviations: ACE Angiotensin converting enzyme, AMI Acute myocardial infarction, COPD Chronic obstructive pulmonary disease, ICD International classification of diseases, NSAIDS Non-steroidal antiinflammatory drugs, P-GP P-glycoprotein, CYP Cytochrome
As shown on Tables 3, 4, 5 and 6, 1401 (17.4%) and 7820 (39.7%) patients who received dabigatran and rivaroxaban met criteria for low dose, respectively. Of those, 959 (68.5%) and 3904 (49.9%) received standard dose of dabigatran and rivaroxaban respectively. In contrast, 1013 (15.3%) and 2551 (21.5%) of patients eligible for standard dose dabigatran and rivaroxaban received low dose. Patients older than 75 years, females, African Americans, and patients with history of major bleeding or heart failure were more likely to receive lower than recommended dose of dabigatran or rivaroxaban (Tables 3, 4, 5 and 6). Conversely, patients eligible for low dose dabigatran or rivaroxaban that received standard dose were more likely younger with lower rates of advanced CKD.
Table 3
Bivariable associations between low dose dabigatran eligible patients’ characteristics on low or standard dose of dabigatran
 
Low dose eligible that received low dose Dabigatran
(n = 442)
Low dose eligible that received standard dose Dabigatran
(n = 959)
P value
Before Matching
Standardized Difference
After Matching
(N = 366 vs 366)
Standardized Difference
Year
  
0.001
  
 2010–2012
57.7%
69.3%
 
0.244
0.228
 2013
16.1%
10.3%
 
0.170
0.289
 2014
10.9%
8.1%
 
0.093
0.047
 2015
7.7%
6.5%
 
0.048
0
 2016–2017
7.7%
5.7%
 
0.078
0.011
Region
  
0.1
  
 Midwest
15.4%
12.4%
 
0.086
0.008
 Northeast
0.5%
1.4%
 
0.096
0.141
 South
73.3%
76.7%
 
0.080
0
 West
10.9%
9.5%
 
0.045
0.035
Age Category
  
< 0.001
  
 65–69
7.9%
14.0%
 
0.195
0
 70 to 74
16.5%
24.2%
 
0.192
0
 75 to 79
18.6%
28.5%
 
0.235
0
 80 to 84
25.1%
19.3%
 
0.140
0
 85 to 89
13.1%
7.8%
 
0.174
0
 90 or over
18.8%
6.3%
 
0.385
0
Sex
  
0.2
  
 Female
57.9%
54.3%
 
0.072
0
 Male
42.1%
45.7%
   
 Race Category
     
 White
84.2%
85.0%
 
0.023
0
 Black
10.4%
8.9%
 
0.052
0
 Hispanic
2.5%
2.4%
 
0.006
0
 Other
    
0
Smoking
  
0.04
  
 Yes
11.5%
15.7%
 
0.123
0.055
 No
88.5%
84.3%
   
Weight Category (based on ICD-9 and ICD-10 codes for BMI Category)
  
0.02
  
 Underweight
1.6%
0.6%
 
0.092
0
 Healthy or Overweight
8.6%
6.3%
 
0.089
0.062
 Obese or Severe Obese
20.4%
26.3%
 
0.140
0.013
 Others
69.5%
66.8%
 
0.056
0.024
Comorbid Conditions
 Prior Stroke
26.7%
27.4%
0.8
0.016
0.042
 Prior major bleeding
29.0%
28.2%
0.8
0.018
0.006
 Diabetes
55.0%
54.0%
0.7
0.019
0.077
 Prior AMI
10.6%
6.2%
0.003
0.162
0.147
 Liver Disease
2.9%
3.2%
0.8
0.017
0
 Heart Failure
56.8%
40.7%
< 0.001
0.327
0.265
 Hypertension
98.2%
96.1%
0.04
0.124
0.082
 Ischemic cardiomyopathy
60.4%
57.2%
0.3
0.064
0.089
 Pulmonary
18.8%
13.3%
0.008
0.148
0.066
 COPD
37.8%
36.2%
0.6
0.033
0.034
 Transfusion from Procedure
5.4%
4.3%
0.3
0.054
0.051
 Revascularization
18.6%
17.8%
0.7
0.019
0.043
 Implantable Devices
20.1%
17.5%
0.2
0.067
0
 Valve Disease
45.9%
42.0%
0.2
0.079
0.099
Concurrent Drugs (+/− 90 days of initiating DOAC)
 SSRI/SNRI
32.6%
36.5%
0.15
0.083
0.023
 Strong and moderate p-gp inhibitors
53.5%
79.9%
< 0.001
0.580
0.068
 Warfarin
22.6%
31.0%
0.001
0.189
0.153
 Strong p-gp and cyp3a4 dual inhibitors
35.7%
47.5%
< 0.001
0.241
0.089
 ACE inhibitors
74.0%
72.7%
0.6
0.029
0.044
 Angiotensin receptor blockers
46.6%
45.9%
0.8
0.015
0.066
 Beta blockers
95.2%
92.9%
0.09
0.099
0.165
 Calcium channel blockers
69.7%
67.5%
0.4
0.048
0.098
 Digoxin
28.7%
33.8%
0.06
0.109
0.083
 Proton pump inhibitors
64.7%
63.2%
0.6
0.032
0.119
 NSAIDs
52.7%
55.4%
0.4
0.053
0.055
 Antiplatelets
35.3%
31.3%
0.13
0.085
0.099
 Insulin
22.9%
17.6%
0.02
0.130
0.206
 Statins
80.8%
83.3%
0.24
0.066
0.007
Renal Disease
 Moderate (GFR 30–60 ml/min/1.73 m2)
45.7%
74.9%
< 0.001
0.6245
0.0439
 Severe
(GFR < 30 ml/min/1.73 m2)
54.3%
25.1%
   
Abbreviations: ACE Angiotensin converting enzyme, AMI Acute myocardial infarction, COPD Chronic obstructive pulmonary disease, ICD International classification of diseases, NSAIDS Non-steroidal antiinflammatory drugs, P-GP P-glycoprotein, CYP Cytochrome
Table 4
Bivariable associations between standard dose dabigatran eligible patients’ characteristics on low or standard dose of dabigatran
 
Standard dose eligible that received low dose Dabigatran (n = 1013)
Standard dose eligible that received standard dose Dabigatran (n = 5621)
P value
Before Matching
Standardized Difference
After Matching (N = 1001 vs 1001)
Standardized Difference
Year
  
< 0.001
  
 2010–12
50.0%
55.7%
 
0.116
0.198
 2013
18.6%
13.6%
 
0.137
0.174
 2014
11.6%
11.4%
 
0.008
0.032
 2015
10.9%
10.1%
 
0.026
0.047
 2016–17
9.0%
9.3%
 
0.009
0.040
Region
  
0.8
  
 Midwest
12.3%
12.9%
 
0.017
0.024
 Northeast
1.7%
1.5%
 
0.018
0.016
 South
73.7%
74.3%
 
0.012
0.009
 West
12.2%
11.4%
 
0.027
0.006
Age Category
  
< 0.001
  
 65–69
6.7%
18.7%
 
0.365
0
 70 to 74
12.5%
27.3%
 
0.3755
0
 75 to 79
19.0%
24.9%
 
0.143
0
 80 to 84
26.4%
17.9%
 
0.206
0
 85 to 89
18.5%
6.9%
 
0.351
0
 90 or over
17.0%
4.4%
 
0.415
0
Sex
  
< 0.001
  
 Female
55.6%
46.8%
 
0.176
0
 Male
44.4%
53.2%
   
Race Category
  
< 0.001
  
 White
84.0%
87.9%
 
0.113
0
 Black
10.1%
6.6%
 
0.126
0
 Hispanic
2.5%
1.6%
 
0.059
0
 Asian
1.2%
0.6%
 
0.066
0
 Other
2.3%
3.3%
 
0.060
0
 Smoker
15.0%
16.8%
0.15
0.049
0.037
Weight Category (based on ICD-9/ ICD-10 codes for BMI)
  
0.003
  
 Under Weight
1.1%
0.6%
 
0.057
0.031
 Healthy or Overweight
10.8%
8.3%
 
0.082
0.013
 Obese or Severe Obese
20.0%
23.4%
 
0.084
0.054
 Others
68.1%
67.6%
 
0.010
0.061
Comorbid Conditions
 Prior Stroke
26.9%
23.9%
0.04
0.067
0.011
 Prior Major Bleeding
25.8%
23.9%
0.2
0.044
0.042
 Diabetes
48.9%
48.0%
0.6
0.018
0.066
 Prior AMI
6.9%
5.0%
0.01
0.082
0.103
 Liver Disease
3.6%
2.5%
0.07
0.059
0.126
 Heart Failure
45.0%
29.7%
< 0.001
0.320
0.274
 Hypertension
95.1%
93.0%
0.01
0.089
0.088
 Ischemic Cardiomyopathy
53.4%
46.4%
< 0.001
0.140
0.110
 Pulmonary Circulatory Disease
15.6%
11.0%
< 0.001
0.135
0.083
 COPD
35.7%
31.0%
0.003
0.1
0.115
 Blood Transfusion
3.2%
2.4%
0.18
0.044
0
 Revascularization
16.8%
14.5%
0.05
0.063
0.075
 Implantable cardiac device
15.5%
13.1%
0.04
0.068
0.011
 Valve Disease
44.1%
40.0%
0.015
0.083
0.024
Concurrent Drugs (+/− 90 days of initiating DOAC)
 SSRI/SNRI
34.0%
30.0%
0.01
0.085
0.126
 Strong and moderate p-gp inhibitors
7.3%
12.9%
< 0.001
0.188
0.135
 Warfarin
19.9%
24.0%
0.004
0.099
0.237
 Strong p-gp and cyp3a4 dual inhibitors
17.0%
18.2%
0.3
0.0325
0.032
 ACE inhibitors
68.4%
67.2%
0.45
0.0257
0.081
 Angiotensin receptor blockers
38.6%
38.1%
0.8
0.0094
0.010
 Beta blockers
90.7%
88.4%
0.03
0.0759
0.108
 Calcium channel blockers
65.5%
63.2%
0.15
0.0488
0.040
 Digoxin
32.1%
28.7%
0.03
0.0741
0.002
 Proton pump inhibitors
58.7%
55.2%
0.04
0.0707
0.008
 NSAIDs
48.3%
50.4%
0.2
0.0433
0.04
 Antiplatelets
30.5%
25.7%
0.0015
0.1064
0.093
 Insulin
14.5%
12.1%
0.03
0.072
0.137
 Statins
79.1%
78.7%
0.8
0.009
0.079
Renal Disease
  
< 0.001
  
 None or Mild
33.6%
57.2%
 
0.489
0.255
 Moderate (GFR 30–60 ml/min/1.73 m2)
66.4%
42.8%
   
Severe (GFR < 30 ml/min/1.73 m2)
0.0%
0.0%
   
Abbreviations: ACE Angiotensin converting enzyme, AMI Acute myocardial infarction, COPD Chronic obstructive pulmonary disease, ICD International classification of diseases, NSAIDS Non-steroidal antiinflammatory drugs, P-GP P-glycoprotein, CYP Cytochrome
Table 5
Bivariable associations between low dose rivaroxaban eligible patients’ characteristics on low or standard dose of rivaroxaban
 
Low dose eligible that received low dose Rivaroxaban
(n = 3916)
Low dose eligible that received standard dose Rivaroxaban (n = 3904)
P value
Before Matching
Standardized Difference
After Matching
(N = 2703 vs 2703)
Standardized Difference
Year
  
0.2
  
 2010–2012
7.2%
6.7%
 
0.021
0.028
 2013
18.3%
17.6%
 
0.017
0.0308
 2014
23.0%
23.0%
 
0.002
0.016
 2015
22.9%
21.8%
 
0.024
0.003
 2016–2017
28.6%
30.9%
 
0.050
0.058
Region
  
0.04
  
 Midwest
15.2%
13.0%
 
0.064
0.084
 Northeast
1.2%
1.1%
 
0.009
0.016
 South
73.0%
75.0%
 
0.044
0.028
 West
10.5%
10.9%
 
0.013
0.061
Age Category
  
< 0.001
  
 65–69
7.5%
14.5%
 
0.225
0
 70 to 74
14.0%
26.0%
 
0.302
0
 75 to 79
20.1%
25.8%
 
0.135
0
 80 to 84
25.5%
19.7%
 
0.139
0
 85 to 89
20.5%
10.1%
 
0.291
0
 90 or over
12.4%
3.9%
 
0.311
0
Sex
  
0.0008
  
 Female
56.0%
52.3%
 
0.076
0
 Male
44.0%
47.7%
   
Race Category
  
0.7
  
 White
84.5%
83.4%
 
0.031
0
 Black
10.2%
10.7%
 
0.017
0
 Hispanic
2.0%
2.2%
 
0.011
0
 Asian
0.7%
0.8%
 
0.015
0
 Other
2.7%
3.0%
 
0.019
0
 Smoker
19.6%
22.5%
0.001
0.073
0.033
Weight Category (ICD9/ICD-10 codes for BMI)
  
< 0.0001
  
 Under Weight
1.9%
1.2%
 
0.053
0.025
 Healthy or Overweight
18.4%
16.1%
 
0.062
0.008
 Obese or Severe Obese
28.9%
35.4%
 
0.139
0.079
 Others
50.8%
47.3%
 
0.070
0.062
Comorbid Conditions
 Prior Stroke
29.1%
25.1%
< 0.001
0.089
0.051
 Prior major bleeding
27.6%
28.0%
0.6
0.011
0.026
 Diabetes
55.9%
57.2%
0.2
0.027
0.021
 Prior AMI
10.4%
6.7%
< 0.001
0.132
0.119
 Liver Disease
3.2%
3.5%
0.4
0.019
0.017
 Heart Failure
41.2%
31.6%
< 0.001
0.201
0.151
 Hypertension
96.8%
95.3%
< 0.001
0.076
0.030
 Cardiomyopathy
50.4%
44.6%
< 0.001
0.115
0.100
 Pulmonary Circulatory Dis
17.2%
14.2%
< 0.001
0.082
0.021
 COPD
38.5%
37.0%
0.17
0.031
0.054
 Prior Blood Transfusion
5.8%
3.5%
< 0.001
0.107
0.083
 Revascularization
17.7%
15.4%
0.007
0.061
0.079
 Implantable Cardiac Device
18.3%
14.8%
< 0.001
0.094
0.078
 Valve Disease
42.2%
38.1%
< 0.001
0.084
0.064
Concurrent Drugs (+/− 90 days of initiating DOAC)
 SSRI/SNRI
34.0%
33.3%
0.5
0.014
0.034
 Strong and moderate p-gp inhibitors
19.9%
24.5%
< 0.001
0.110
0.012
 Warfarin
16.1%
19.6%
< 0.001
0.092
0.124
 Pgp inducers
17.9%
18.0%
0.9
0.003
0.018
 Strong p-gp and cyp3a4 dual inhibitors
26.1%
35.1%
< 0.001
0.194
0.009
 P-gp and cyp3a4 inducers
17.9%
18.1%
0.9
0.003
0.017
 Ace inhibitors
70.6%
69.6%
0.34
0.022
0.025
 Angiotensin receptor blockers
43.9%
43.8%
0.86
0.004
0.004
 Beta blockers
89.7%
89.0%
0.32
0.023
0.019
 Calcium channel blockers
67.6%
64.4%
=0.003
0.067
0.026
 Digoxin
23.0%
23.3%
0.7
0.008
0.003
 Proton pump inhibitors
59.6%
59.2%
0.7
0.008
0.026
 NSAIDS
52.6%
53.3%
0.5
0.015
0.022
 Antiplatelets
31.8%
28.4%
0.001
0.074
0.067
 Insulin
18.0%
17.1%
0.27
0.025
0.087
 Statins
81.8%
81.6%
0.79
0.006
0.024
Renal Disease
 None or mild
3.6%
17.1%
< 0.0001
0.452
0.058
 Moderate (GFR 30–60 ml/min/1.73 m2)
83.3%
77.4%
 
0.149
0.039
 Severe
(GFR < 30 ml/min/1.73 m2)
13.1%
5.5%
 
0.262
0.002
Abbreviations: ACE Angiotensin converting enzyme, AMI Acute myocardial infarction, COPD Chronic obstructive pulmonary disease, ICD International classification of diseases, NSAIDS Non-steroidal antiinflammatory drugs, P-GP P-glycoprotein, CYP Cytochrome
Table 6
Bivariable associations between standard dose rivaroxaban eligible patients’ characteristics on low or standard dose of rivaroxaban
 
Standard dose eligible that received low dose Rivaroxaban (n = 2551)
Standard dose eligible that received standard dose Rivaroxaban (n = 9341)
P value
Before Matching Standardized Difference
After Matching
(N = 2397 vs 2397)
Standardized Difference
Year
  
0.06
  
 2010–2012
8.2%
7.1%
 
0.0439
0.0031
 2013
17.9%
17.1%
 
0.0204
0.0098
 2014
23.8%
22.7%
 
0.0256
0.0248
 2015
21.4%
22.7%
 
0.0312
0.0051
 2016–2017
28.7%
30.4%
 
0.0379
0.0284
Region
  
0.12
  
 Midwest
15.1%
14.4%
 
0.0178
0.1713
 Northeast
1.1%
1.3%
 
0.0229
0.1515
 South
73.1%
72.0%
 
0.0254
0.2038
 West
10.8%
12.3%
 
0.0476
0.0186
Age Category
  
< 0.0001
  
 65–69
8.5%
19.7%
 
0.3266
0
 70 to 74
15.9%
30.3%
 
0.3453
0
 75 to 79
21.6%
25.5%
 
0.0916
0
 80 to 84
25.3%
15.9%
 
0.2337
0
 85 to 89
18.8%
6.9%
 
0.3622
0
 90 or over
9.8%
1.7%
 
0.3544
0
Sex
  
< 0.0001
  
 Female
58.7%
43.7%
 
0.3044
0
 Male
41.3%
56.3%
   
Race Category
  
< 0.0001
  
 White
86.2%
86.8%
 
0.0164
0
 Black
7.3%
6.9%
 
0.0144
0
 Hispanic
2.4%
1.3%
 
0.0813
0
 Asian
1.1%
0.8%
 
0.0304
0
 Other
3.0%
4.2%
 
0.065
0
 Smoker
21.1%
23.0%
0.05
0.044
0.003
Weight Category (based on ICD-9/ICD-10 codes for BMI)
  
< 0.0001
  
 Under
2.9%
1.4%
 
0.1038
0.0682
 Healthy or Overweight
20.1%
13.6%
 
0.1737
0.2328
 Obese or Severe Obese
22.1%
29.4%
 
0.1684
0.0561
 Others
55.0%
55.6%
 
0.0126
0.2406
Comorbid Conditions
 Prior Stroke
26.9%
21.3%
< 0.001
0.1295
0
 Prior major bleeding
25.2%
22.2%
0.001
0.071
0
 Diabetes
43.4%
43.8%
0.76
0.0069
0.1063
 Prior AMI
7.0%
5.3%
0.001
0.0701
0
 Liver Disease
2.4%
2.9%
0.14
0.0337
0.0139
 Heart Failure
27.0%
21.3%
< 0.001
0.1339
0
 Hypertension
91.2%
90.8%
0.57
0.0128
0.0474
 Cardiomyopathy
43.5%
38.3%
< 0.001
0.1063
0.1134
 Pulmonary Circulatory dis
13.9%
9.8%
< 0.001
0.1277
0.0414
 COPD
33.0%
29.1%
< 0.001
0.0846
0.0972
 Prior blood transfusion
3.7%
1.8%
< 0.001
0.1173
0.0855
 Revascularization
14.7%
13.0%
0.02
0.0495
0.08
 Implantable Devices
14.1%
11.6%
< 0.001
0.0766
0.0553
 Valve Disease
38.1%
35.7%
0.02
0.051
0.0347
Concurrent Drugs (+/− 90 days of initiating DOAC)
 SSRI/SNRI
33.3%
27.4%
< 0.001
0.1289
0.0682
 Strong and moderate p-gp inhibitors
15.7%
16.1%
0.67
0.0096
0
 Warfarin
13.2%
14.6%
0.08
0.0391
0.1199
 Pgp inducers
19.5%
15.8%
< 0.001
0.097
0
 Strong p-gp and cyp3a4 dual inhibitors
17.3%
14.8%
0.002
0.069
0.0232
 P-gp and cyp3a4 inducers
19.5%
15.8%
< 0.001
0.097
0
 Ace inhibitors
62.4%
62.1%
0.77
0.0066
0.006
 Angiotensin receptor blockers
33.8%
33.8%
0.98
0.0005
0.0222
 Beta blockers
85.7%
85.4%
0.74
0.0075
0.042
 Calcium channel blockers
59.6%
57.3%
0.03
0.0475
0.0179
 Digoxin
23.4%
19.8%
<.001
0.0892
0.0403
 Proton pump inhibitors
56.2%
49.5%
< 0.001
0.1356
0.0753
 NSAIDS
53.2%
48.7%
< 0.001
0.091
0.133
 Antiplatelets
27.9%
22.5%
< 0.001
0.1229
0.0785
 Insulin
8.5%
8.2%
0.54
0.0136
0.0247
 Statins
75.4%
75.9%
0.6
0.0114
0.0608
Renal Disease
 None or mild
100.0%
100.0%
  
Not available due to perfect match
Moderate (GFR 30–60 ml/min/1.73 m2)
     
Severe
(GFR < 30 ml/min/1.73 m2)
     
Abbreviations: ACE Angiotensin converting enzyme, AMI Acute myocardial infarction, COPD Chronic obstructive pulmonary disease, ICD International classification of diseases, NSAIDS Non-steroidal antiinflammatory drugs, P-GP P-glycoprotein, CYP Cytochrome
Mean follow-up for patients eligible for low dose dabigatran, standard dose dabigatran, low dose rivaroxaban, and standard dose rivaroxaban were 13.9, 15.1, 10.1, and 12.3 months, respectively.

Outcomes

Stroke

The absolute event rates and event rates/year for ischemic stroke in each dosing category are presented in Table 7. Before adjustment for patient characteristics or propensity-match analysis, use of low dose dabigatran among patients eligible for standard dose dabigatran did not affect ischemic stroke risk. Among those eligible for standard dose rivaroxaban but receiving low dose, no significantly different risk of ischemic stroke was found. Among patients eligible for low dose dabigatran who received standard dose, we did not identify any relationship to ischemic stroke risk (Table 7). Also, among patients eligible for low dose rivaroxaban, use of standard dose rivaroxaban was not associated with increased risk of ischemic stroke. After propensity matching, we found no difference in risk of ischemic stroke in 732 patients eligible for low dose dabigatran who received low dose compared to 732 matched patients eligible for low dose dabigatran who received standard dose, or among propensity-matched patients eligible for standard dose dabigatran who received either standard dose (n = 1960) or low dose (n = 1960). Similarly, analysis of propensity matched samples of patients eligible for low dose rivaroxaban (5328 on low dose and 5328 on standard dose) or patients eligible for standard dose rivaroxaban (4500 on standard dose and 4500 on low dose) found no significant association of dose to risk of ischemic stroke (Table 7).
Table 7
Hazard ratios (95% Confidence Intervals) of Outcomes in Matched Cohorts of Dabigatran and Rivaroxaban low and standard doses in Non-Valvular Atrial Fibrillation
 
Patients Eligible for low dose but taking standard dose vs. those taking low dose
Patients Eligible for Standard dose but taking low dose vs. those taking dose standard dose
Received Low Dose
Events (Events/year)
Received Standard Dose
Events (Events/year)
95% Confidence Interval, p-value
Received Low Dose
Events (Events/year)
Received Standard Dose
Events (Events/year)
95% Confidence Interval, p-value
Ischemic Stroke
 Dabigatran
Unadjusted
25 (0.059)
57 (0.043)
0.77 (0.47–1.26; p = 0.3)
59 (0.051)
319 (0.042)
1.2 (0.92–1.6, P = 0.17)
Propensity Matched
22 (0.059)
28 (0.061)
1.13 (0.63–2; p = 0.69)
55 (0.049)
88 (0.071)
0.74 (0.53–1.04; p = 0.08)
 Rivaroxaban
Unadjusted
176 (0.057)
161 (0.043)
0.87 (0.7–1.1; p = 0.2)
89 (0.040)
256 (0.026)
1.36 (1.07–1.7, P = 0.01)
Propensity Matched
103 (0.048)
128 (0.053)
1.24 (0.96–1.6; p = 0.1)
78 (0.040)
73 (0.030)
1.13 (0.83–1.56, P = 0.43)
Major Bleeding
 Dabigatran
Unadjusted
62 (0.1456)
124 (0.093)
0.69 (0.51–0.94; p = 0.02)
82 (0.071)
393 (0.051)
1.35 (1.06–1.7, p = 0.01)
Propensity Matched
46 (0.1228)
58 (0.1258)
1.07 (0.7–1.58 P = 0.75)
78 (0.069)
92 (0.075)
1.01 (0.74–1.37, P = 0.97)
 Rivaroxaban
Unadjusted
307 (0.099)
294 (0.079)
0.95 (0.8–1.1, P = 0.5)
147 (0.067)
426 (0.043)
1.35 (1.12–1.6, P = 0.001)
Propensity Matched
197 (0.091)
216 (0.089)
1.12 (0.92–1.35, P = 0.26)
131 (0.068)
116 (0.048)
1.2 (0.93–1.53, P = 0.21)
GI Hemorrhage
 Dabigatran
Unadjusted
46 (0.108)
100(0.075)
0.75 (0.5–1.07, P = 0.1)
67 (0.058)
292 (0.038)
1.47 (1.13–1.9, P = 0005)
Propensity Matched
33 (0.088)
45 (0.098)
1.16 (0.74–1.82, P = 0.53)
63 (0.056)
68 (0.055)
1.08 (0.77–1.53, P = 0.66)
 Rivaroxaban
Unadjusted
243 (0.078)
227 (0.061)
0.93 (0.78–1.1, P = 0.4)
111 (0.050)
341 (0.034)
1.28 (1.04–1.6, P = 0.02)
Propensity Matched
158 (0.073)
166 (0.069)
1.07 (0.86–1.32, P = 0.5)
97 (0.050)
96 (0.040)
1.08 (0.82–1.43, P = 0.58)
Intracranial Hemorrhage
 Dabigatran
Unadjusted
< 11
< 11
0.33 (0.12–0.9; p = 0.03)
< 11
52 (0.007)
0.77 (0.33–1.8; p = 0.54)
Propensity Matched
< 11
< 11
0.63 (0.2–1.99; p = 0.43)
< 11
12(0.009)
0.58 (0.22–1.54; p = 0.27)
 Rivaroxaban
Unadjusted
25 (0.008)
26 (0.007)
0.99 (0.57–1.7; p = 0.96)
13 (0.006)
38 (0.004)
1.33 (0.7–2.5; p = 0.37)
Propensity Matched
17 (0.008)
22 (0.009)
1.26 (0.67–2.38; p = 0.47)
12 (0.006)
< 11
1.57 (0.64–3.84; p = 0.32)

Major bleeding

The absolute event rates and event rates/year for bleeding complications in each dosing category are presented in Table 7. Among patients on dabigatran who met criteria for standard dose, use of low dose was associated with significantly higher risk of any major bleeding (HR = 1.44; 95% CI 1.14–1.8, P = 0.002, Table 7), and higher risk of GI bleeding (HR 1.48, 95% CI 1,08–2, P = 0.016) but not intracranial bleeding compared with patients on standard doses of dabigatran. A similar pattern of increased major bleeding risk (HR 1.34, 95% CI 1.11–1.6, P = 0.002) was identified among patients on rivaroxaban who met criteria for standard dose but received low dose, along with a trend towards increased risk of GI bleeding (HR 1.26, 95% CI 0.99–1.6, P = 0.06) but not intracranial bleeding.
In patients who met criteria for low dose dabigatran, there was lower risk of major bleeding (HR = 0.59; 95% CI 0.43–0.8, P < 0.001, Table 7) and intracranial bleeding (HR = 0.33; 95% CI 0.12–0.9, P = 0.03, Table 7) but not GI bleeding in patients who received standard compared to low dose dabigatran. Among patients who met criteria for low dose rivaroxaban, there was lower risk of GI bleeding (HR = 0.79; 95% CI 0.64–0.98, P = 0.03, Table 7) without differences in the risk of major, and intracranial bleeding. After controlling for patient characteristics in propensity-matched samples, we did not find any association of off-label use of low dose or standard dose and the risk of any bleeding events for either dabigatran or rivaroxaban.

Sensitivity analysis

We performed sensitivity analysis among patients with dose adjustments based on valid GFR measurements only and not based on pharmacologic interactions. The propensity matched analysis showed that standard dose dabigatran is associated with higher risk of stroke among patients eligible for low dose according to GFR (HR 2.6, 95% CI 1.03–6.7; p = 0.04). The analysis did not suggest any other significant differences in stroke and bleeding risks between off-label and standard dosing of dabigatran and rivaroxaban. The results are presented in Additional file 2: Table S2.

Discussion

The findings of this retrospective cohort analysis of Medicare beneficiaries with AF treated with dabigatran or rivaroxaban between 2010 and 2016 can be summarized as follows: i) among patients on dabigatran or rivaroxaban who met criteria for low dose, the majority received standard dose; ii) among patients on dabigatran or rivaroxaban who met criteria for standard dose, less than one fourth received the low dose; iii) older age, female sex, black race, bleeding history, and heart failure were associated with receipt of lower than recommended dose for patients receiving dabigatran or rivaroxaban; iv) unadjusted analysis suggested that in patients receiving lower dose than recommended, the risk of any major bleeding was increased, likely reflecting higher baseline bleeding risk, while in patients receiving higher dose than recommended, the risk of bleeding was decreased; v) after risk adjusting using multivariable models or propensity-matching, off-label dosing of dabigatran or rivaroxaban was not associated with increased risk of stroke or bleeding compared to recommended dosing. An increased risk of ischemic stroke with standard dose dabigatran was found among patients eligible for low dose based on eGFR only.
The results of our analysis are in accordance with findings of previous studies which demonstrated that a significant part of AF population on DOACs receive an off-label dose [14, 15, 19]. An updated analysis of the ORBITA-AF II registry from 2013 until 2016, including 7925 AF patients treated with DOACs, showed that 84% received DOACs at standard dose (mainly rivaroxaban and apixaban, only 451 patients on dabigatran), which was consistent with FDA labeling in 96% of cases [ 15]. Reduced DOAC dose was prescribed to 16% of patients, which was consistent with FDA labeling in 43%. In unadjusted analysis, under-dosing was associated with higher rates of all-cause mortality and major bleeding [15]. Nevertheless, after risk- adjustment, the use of lower-than-recommended dose resulted in similar thromboembolic and bleeding risk compared to appropriately dosed DOAC use [15]. Our cohort included a larger sample size than ORBITA-AF II and focused on dabigatran and rivaroxaban as opposed to apixaban and rivaroxaban in ORBITA-AF II. Another methodological difference is the use of calculated creatinine clearance with the Cockcroft-Gault formula instead of the MDRD or the CKD-EPI equations for eGFR calculation that we applied. Although in clinical practice, eGFR by MDRD or CKD-EPI is more commonly used than calculated creatinine clearance, discordances in dabigatran and rivaroxaban doses may occur in up to 30% of elderly patients with creatinine clearance < 60 ml/min [ 19]. Despite these methodological differences both our analysis and the previous report from the ORBITA-AF II registry suggest that among patients on dabigatran or rivaroxaban who met criteria for low dose, the majority received standard dose. Moreover, unadjusted analysis suggested that in patients receiving lower dose than recommended, the risk of any major bleeding was increased. In both analyses, propensity matching did not reveal any significant differences in stroke and bleeding.
Yao et al.19 previously evaluated potential over- and under- dosing of DOACs based only on renal indication for dose reduction using the data for privately insured and Medicare Advantage enrollees. Like our study, Yao et al. found no significant relationship between risk of stroke or bleeding and dose in dabigatran or rivaroxaban-treated patients with renal indication for dose reduction. However, in aggregate analyses of patients taking dabigatran, rivaroxaban or apixaban with renal indication for low dose, patients had significantly higher bleeding risk if they received standard dose. Consistent with our study, Yao et al. also found no statistically significant relationship between dose reduction and risk of stroke or bleeding in the dabigatran- or rivaroxaban-treated patients who did not have a renal indication for low dose. In contrast to our study, Yao et al. evaluated renal indications for dose reductions only, and did not consider use of p-gp inhibitors or dual p-gp and CYP3A4 inhibitors in assessing dosing criteria.
In aggregate our findings demonstrate that decisions by healthcare providers about DOAC dosing may be based on patient clinical conditions not reflected in FDA dosing recommendations. Our analysis suggests that patients deemed by providers to be at higher bleeding risk may have received low dose DOACs even though FDA criteria suggest that they qualified for standard dose. It is noteworthy that our unadjusted analyses found higher risk of bleeding in patients who met criteria for standard dose rivaroxaban or dabigatran but received low dose, suggesting that the perception of higher bleeding risk by physicians was warranted, and may point to important patient characteristics not reflected in FDA criteria. Similarly, patients qualifying for low dose may have been prescribed standard doses if providers deemed their bleeding risk to be low.
DOAC-specific factors should predominantly affect dosing decisions. Renal function is the main indicator for low dose dabigatran and rivaroxaban. Nearly ~ 80% of ingested dabigatran is metabolized by the kidney, while ~ 30% of rivaroxaban is metabolized by the kidney. With chronic kidney disease, the half-lives of these medications are extended, leading to potentially high plasma concentrations of the medications and increased bleeding risk [20, 21]. DOAC dose adjustments based on renal function therefore reflect the increased bleeding risk in patients with compromised renal function. Several drug interactions also affect dosing of DOACs. Dabigatran is a substrate for P-glycoprotein. Concomitant use of dabigatran with P-gp inducers such as rifampin reduces the anticoagulant effect of dabigatran and should be avoided whereas use of dabigatran with P-gp inhibitors (eg, ketoconazole, dronedarone) in patients with renal disease may increase the anticoagulant effect, hence dose adjustment is required. Combined P-gp and CYP3A4 inhibitors (ketoconazole, protease inhibitors) increase the anticoagulant effects of rivaroxaban and should not be used concomitantly with rivaroxaban. In our analysis, we found a significant percentage of patients on medications interfering with the metabolism of dabigatran and rivaroxaban. Notably, receipt of standard dose dabigatran among patients qualifying for low dose dabigatran due to eGFR 30–60 ml/min with concomitant use of a p-gp inhibitor was particularly common, suggesting that some providers may not recognize the potential interaction. We also note, however, that our pharmacy data only permit evaluation of prescription fills, thus it is possible that some patients receiving concomitant p-gp inhibitors were instructed not to take them or switched to an alternative drug, in which case they would not meet criteria for low dose dabigatran. Other combinations that increase the risk of bleeding with DOACs are antiplatelet agents and non-steroidal anti-inflammatory drugs (NSAIDs), although no specific dosing adjustments are recommended for patients on these drugs. In our study, we found that use of prescription anti-platelet use among patients who met criteria for standard dose dabigatran or rivaroxaban was associated with a modestly higher use of low dose dabigatran and rivaroxaban. Similarly, use of low dose rivaroxaban was also more frequent among rivaroxaban patients taking NSAIDS.

Limitations

An important limitation of this paper is our inability to measure GFR using the Cockcroft-Gault [CG] equation, which is reflected in FDA recommendations for DOAC dosage reductions. There are three commonly used equations for estimating GFR: the oldest is the CG equation, originally published in 1976, followed by the MDRD, updated MDRD, and CKD-EPI equations in 1999, 2005, and 2009, respectively [15, 16, 22]. The MDRD and CKD-EPI equations use serum creatinine in combination with age, sex, and race to estimate GFR, while CG also uses patient weight. The use of alternative equations for dosing decisions has been the topic of considerable debate. In clinical practice, physicians rarely use the CG-estimated GFR and instead rely on the MDRD or CKD-EPI equations, which are easy to calculate and often automatically reported with serum creatinine laboratory tests. Notably, the National Kidney Disease Education Program (NKDEP) previously indicated that either the MDRD or CG equation may be used for drug dosing decisions [23], while more recently, the National Institute of Diabetes and Digestive and Kidney Diseases suggested that either CKD-EPI or CG equations are appropriate for drug dosing purpose [24]. In contrast, a previous review of FDA-recommended drug dosing showed that the CG equation is historically the most common renal function equation cited in drug dosing recommendations [25]. As noted by Yao et al. [26], it is likely that FDA drug labels historically relied on CG-estimated GFR because this method was available before the MDRD or CKD-EPI equations were developed and widely adopted, and drug dosage recommendations have not caught up to standard clinical practice with respect to assessing renal function. Nevertheless, this poses inconsistencies with clinical practice. While for most patients, GFR estimated by the MDRD or CKD-EPI equations has reasonable concordance with CG-estimated GFR, for older patients and patients with significant comorbidity, malnutrition leading to decreased muscle mass, or morbid obesity, there may be less agreement and dosing of medications based on the former may not be consistent with FDA recommendations [ 2729]. Schwartz [30] found that use of the MDRD or CKD-EPI equations rather than the CG equation for estimating GFR may fail to identify 20 to 50% of patients for whom reduced dabigatran and rivaroxaban doses are recommended. Thus, our analysis likely underestimates the number of patients who qualify for low dose rivaroxaban and dabigatran if decisions are based on the CG equation, thereby underestimating the proportion of patients who are overdosed, or overestimating the proportion of patients who are underdosed, relative to FDA criteria. However, they do likely reflect dosing decisions based on GFR estimates typically used in clinical practice.
Other potential limitations of this study should also be considered. First, due to the observational nature of the study, it is possible that unmeasured confounders could have affected our results in spite of using propensity matched analysis. Second, our analysis included patients over the age of 65 and the findings require validation in younger patients. Finally, we lacked detailed evidence on AF burden and estimation of thromboembolic risk. Strengths our study are the large sample size, the availability of laboratory results (GFR) for most patients, incorporating concomitant medication use for assessing dosing criteria, and application of risk adjustment methods including propensity-matched analyses.

Conclusion

The purpose of our study was to improve understanding of safety and efficacy of DOACs in AF patients receiving low or standard dosing of these medications that was inconsistent with FDA criteria. The majority of patients qualifying for low dose DOACs received standard doses and a percentage of patients qualifying for standard dosing received low dose. After adjustment for comorbidities the risk of stroke and major bleeding was not affected by use of dose inconsistent with FDA criteria. Further validation of our results is warranted especially in patients at high thromboembolic or bleeding risk.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12872-020-01340-4.

Acknowledgements

None.

Author disclosures

The authors have no conflicts of interest to disclose.
All data used in this study were anonymized for use and accessed through a HIPAA-compliant platform maintained by PearlDiver, Inc.
Not applicable, this not a case report.

Competing interests

Alexandros Briasoulis is a member of the editorial board of this journal.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent risk factor for stroke: the Framingham study. Stroke. 1991;22:983–8.CrossRef Wolf PA, Abbott RD, Kannel WB. Atrial fibrillation as an independent risk factor for stroke: the Framingham study. Stroke. 1991;22:983–8.CrossRef
2.
Zurück zum Zitat Friberg L, Rosenqvist M, Lip GY. Net clinical benefit of warfarin in patients with atrial fibrillation: a report from the Swedish atrial fibrillation cohort study. Circulation. 2012;125:2298–307.CrossRef Friberg L, Rosenqvist M, Lip GY. Net clinical benefit of warfarin in patients with atrial fibrillation: a report from the Swedish atrial fibrillation cohort study. Circulation. 2012;125:2298–307.CrossRef
3.
Zurück zum Zitat Poli D, Antonucci E, Testa S, Tosetto A, Ageno W, Palareti G, Italian Federation of Anticoagulation C. Bleeding risk in very old patients on vitamin K antagonist treatment: results of a prospective collaborative study on elderly patients followed by Italian Centres for anticoagulation. Circulation 2011;124:824–829.CrossRef Poli D, Antonucci E, Testa S, Tosetto A, Ageno W, Palareti G, Italian Federation of Anticoagulation C. Bleeding risk in very old patients on vitamin K antagonist treatment: results of a prospective collaborative study on elderly patients followed by Italian Centres for anticoagulation. Circulation 2011;124:824–829.CrossRef
4.
Zurück zum Zitat Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A, Pogue J, Reilly PA, Themeles E, Varrone J, Wang S, Alings M, Xavier D, Zhu J, Diaz R, Lewis BS, Darius H, Diener HC, Joyner CD, Wallentin L, Committee R-LS, Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 2009;361:1139–1151.CrossRef Connolly SJ, Ezekowitz MD, Yusuf S, Eikelboom J, Oldgren J, Parekh A, Pogue J, Reilly PA, Themeles E, Varrone J, Wang S, Alings M, Xavier D, Zhu J, Diaz R, Lewis BS, Darius H, Diener HC, Joyner CD, Wallentin L, Committee R-LS, Investigators. Dabigatran versus warfarin in patients with atrial fibrillation. N Engl J Med 2009;361:1139–1151.CrossRef
5.
Zurück zum Zitat Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, Breithardt G, Halperin JL, Hankey GJ, Piccini JP, Becker RC, Nessel CC, Paolini JF, Berkowitz SD, Fox KA, Califf RM, Investigators RA. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883–91.CrossRef Patel MR, Mahaffey KW, Garg J, Pan G, Singer DE, Hacke W, Breithardt G, Halperin JL, Hankey GJ, Piccini JP, Becker RC, Nessel CC, Paolini JF, Berkowitz SD, Fox KA, Califf RM, Investigators RA. Rivaroxaban versus warfarin in nonvalvular atrial fibrillation. N Engl J Med. 2011;365:883–91.CrossRef
6.
Zurück zum Zitat Granger CB, Alexander JH, McMurray JJ, Lopes RD, Hylek EM, Hanna M, Al-Khalidi HR, Ansell J, Atar D, Avezum A, Bahit MC, Diaz R, Easton JD, Ezekowitz JA, Flaker G, Garcia D, Geraldes M, Gersh BJ, Golitsyn S, Goto S, Hermosillo AG, Hohnloser SH, Horowitz J, Mohan P, Jansky P, Lewis BS, Lopez-Sendon JL, Pais P, Parkhomenko A, Verheugt FW, Zhu J, Wallentin L, Committees A, Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2011;365:981–992.CrossRef Granger CB, Alexander JH, McMurray JJ, Lopes RD, Hylek EM, Hanna M, Al-Khalidi HR, Ansell J, Atar D, Avezum A, Bahit MC, Diaz R, Easton JD, Ezekowitz JA, Flaker G, Garcia D, Geraldes M, Gersh BJ, Golitsyn S, Goto S, Hermosillo AG, Hohnloser SH, Horowitz J, Mohan P, Jansky P, Lewis BS, Lopez-Sendon JL, Pais P, Parkhomenko A, Verheugt FW, Zhu J, Wallentin L, Committees A, Investigators. Apixaban versus warfarin in patients with atrial fibrillation. N Engl J Med 2011;365:981–992.CrossRef
7.
Zurück zum Zitat Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL, Waldo AL, Ezekowitz MD, Weitz JI, Spinar J, Ruzyllo W, Ruda M, Koretsune Y, Betcher J, Shi M, Grip LT, Patel SP, Patel I, Hanyok JJ, Mercuri M, Antman EM, Investigators EA-T. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093–104.CrossRef Giugliano RP, Ruff CT, Braunwald E, Murphy SA, Wiviott SD, Halperin JL, Waldo AL, Ezekowitz MD, Weitz JI, Spinar J, Ruzyllo W, Ruda M, Koretsune Y, Betcher J, Shi M, Grip LT, Patel SP, Patel I, Hanyok JJ, Mercuri M, Antman EM, Investigators EA-T. Edoxaban versus warfarin in patients with atrial fibrillation. N Engl J Med. 2013;369:2093–104.CrossRef
8.
Zurück zum Zitat Lehr T, Haertter S, Liesenfeld KH, Staab A, Clemens A, Reilly PA, Friedman J. Dabigatran etexilate in atrial fibrillation patients with severe renal impairment: dose identification using pharmacokinetic modeling and simulation. J Clin Pharmacol. 2012;52:1373–8.CrossRef Lehr T, Haertter S, Liesenfeld KH, Staab A, Clemens A, Reilly PA, Friedman J. Dabigatran etexilate in atrial fibrillation patients with severe renal impairment: dose identification using pharmacokinetic modeling and simulation. J Clin Pharmacol. 2012;52:1373–8.CrossRef
9.
Zurück zum Zitat Martin JL, Esmaeili H, Manuel RC, Petrini M, Wiebe S, Maas H. Pharmacokinetics/ pharmacodynamics of Dabigatran 75 mg twice daily in patients with Nonvalvular atrial fibrillation and severely impaired renal function. J Cardiovasc Pharmacol Ther. 2018;23:399–406.CrossRef Martin JL, Esmaeili H, Manuel RC, Petrini M, Wiebe S, Maas H. Pharmacokinetics/ pharmacodynamics of Dabigatran 75 mg twice daily in patients with Nonvalvular atrial fibrillation and severely impaired renal function. J Cardiovasc Pharmacol Ther. 2018;23:399–406.CrossRef
10.
Zurück zum Zitat Camm AJ, Lip GY, De Caterina R, Savelieva I, Atar D, Hohnloser SH, Hindricks G, Kirchhof P, Guidelines ESCCfP. 2012 focused update of the ESC Guidelines for the management of atrial fibrillation: an update of the 2010 ESC Guidelines for the management of atrial fibrillation. Developed with the special contribution of the European Heart Rhythm Association. Eur Heart J 2012;33:2719–2747. Camm AJ, Lip GY, De Caterina R, Savelieva I, Atar D, Hohnloser SH, Hindricks G, Kirchhof P, Guidelines ESCCfP. 2012 focused update of the ESC Guidelines for the management of atrial fibrillation: an update of the 2010 ESC Guidelines for the management of atrial fibrillation. Developed with the special contribution of the European Heart Rhythm Association. Eur Heart J 2012;33:2719–2747.
11.
Zurück zum Zitat January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC Jr, Conti JB, Ellinor PT, Ezekowitz MD, Field ME, Murray KT, Sacco RL, Stevenson WG, Tchou PJ, Tracy CM, Yancy CW, American College of Cardiology/American Heart Association Task Force on Practice G. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association task force on practice guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2014;64:e1–76.CrossRef January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC Jr, Conti JB, Ellinor PT, Ezekowitz MD, Field ME, Murray KT, Sacco RL, Stevenson WG, Tchou PJ, Tracy CM, Yancy CW, American College of Cardiology/American Heart Association Task Force on Practice G. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association task force on practice guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2014;64:e1–76.CrossRef
12.
Zurück zum Zitat Basaran O, Filiz Basaran N, Cekic EG, Altun I, Dogan V, Mert GO, Mert KU, Akin F, Soylu MO, Memic Sancar K, Biteker M. PRescriptiOn PattERns of Oral anticoagulants in Nonvalvular atrial fibrillation (PROPER study). Clin Appl Thromb Hemost. 2017;23:384–91.CrossRef Basaran O, Filiz Basaran N, Cekic EG, Altun I, Dogan V, Mert GO, Mert KU, Akin F, Soylu MO, Memic Sancar K, Biteker M. PRescriptiOn PattERns of Oral anticoagulants in Nonvalvular atrial fibrillation (PROPER study). Clin Appl Thromb Hemost. 2017;23:384–91.CrossRef
14.
Zurück zum Zitat Steinberg BA, Shrader P, Thomas L, Ansell J, Fonarow GC, Gersh BJ, Kowey PR, Mahaffey KW, Naccarelli G, Reiffel J, Singer DE, Peterson ED, Piccini JP, Investigators O-A, Patients. Off-label dosing of non-vitamin K antagonist Oral anticoagulants and adverse outcomes: the ORBIT-AF II registry. J Am Coll Cardiol 2016;68:2597–2604.CrossRef Steinberg BA, Shrader P, Thomas L, Ansell J, Fonarow GC, Gersh BJ, Kowey PR, Mahaffey KW, Naccarelli G, Reiffel J, Singer DE, Peterson ED, Piccini JP, Investigators O-A, Patients. Off-label dosing of non-vitamin K antagonist Oral anticoagulants and adverse outcomes: the ORBIT-AF II registry. J Am Coll Cardiol 2016;68:2597–2604.CrossRef
15.
Zurück zum Zitat Steinberg BA, Shrader P, Pieper K, Thomas L, Allen LA, Ansell J, Chan PS, Ezekowitz MD, Fonarow GC, Freeman JV, Gersh BJ, Kowey PR, Mahaffey KW, Naccarelli GV, Reiffel JA, Singer DE, Peterson ED, Piccini JP. Outcomes Registry for Better Informed Treatment of Atrial Fibrillation III. Frequency and Outcomes of Reduced Dose Non-Vitamin K Antagonist Anticoagulants: Results From ORBIT-AF II (The Outcomes Registry for Better Informed Treatment of Atrial Fibrillation II). J Am Heart Assoc. 2018;7(4). https://doi.org/10.1161/JAHA.117.007633. Steinberg BA, Shrader P, Pieper K, Thomas L, Allen LA, Ansell J, Chan PS, Ezekowitz MD, Fonarow GC, Freeman JV, Gersh BJ, Kowey PR, Mahaffey KW, Naccarelli GV, Reiffel JA, Singer DE, Peterson ED, Piccini JP. Outcomes Registry for Better Informed Treatment of Atrial Fibrillation III. Frequency and Outcomes of Reduced Dose Non-Vitamin K Antagonist Anticoagulants: Results From ORBIT-AF II (The Outcomes Registry for Better Informed Treatment of Atrial Fibrillation II). J Am Heart Assoc. 2018;7(4). https://​doi.​org/​10.​1161/​JAHA.​117.​007633.
16.
Zurück zum Zitat Levey AS, Coresh J, Greene T, et al. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247–54.CrossRef Levey AS, Coresh J, Greene T, et al. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247–54.CrossRef
17.
Zurück zum Zitat Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–12.CrossRef Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–12.CrossRef
18.
Zurück zum Zitat Charlson M, Szatrowski TP, Peterson J, Gold J. Validation of a combined comorbidity index. J Clin Epidemiol. 1994;47:1245–51.CrossRef Charlson M, Szatrowski TP, Peterson J, Gold J. Validation of a combined comorbidity index. J Clin Epidemiol. 1994;47:1245–51.CrossRef
19.
Zurück zum Zitat Yao X, Shah ND, Sangaralingham LR, Gersh BJ, Noseworthy PA. Non-vitamin K antagonist oral anticoagulant dosing in patients with atrial fibrillation and renal dysfunction. J Am Coll Cardiol. 2017;69:2779–90.CrossRef Yao X, Shah ND, Sangaralingham LR, Gersh BJ, Noseworthy PA. Non-vitamin K antagonist oral anticoagulant dosing in patients with atrial fibrillation and renal dysfunction. J Am Coll Cardiol. 2017;69:2779–90.CrossRef
20.
Zurück zum Zitat Stangier J, Rathgen K, Stahle H, Mazur D. Influence of renal impairment on the pharmacokinetics and pharmacodynamics of oral dabigatran etexilate: an open-label, parallel-group, single-Centre study. Clin Pharmacokinet. 2010;49:259–68.CrossRef Stangier J, Rathgen K, Stahle H, Mazur D. Influence of renal impairment on the pharmacokinetics and pharmacodynamics of oral dabigatran etexilate: an open-label, parallel-group, single-Centre study. Clin Pharmacokinet. 2010;49:259–68.CrossRef
22.
Zurück zum Zitat Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31–41.CrossRef Cockcroft DW, Gault MH. Prediction of creatinine clearance from serum creatinine. Nephron. 1976;16(1):31–41.CrossRef
25.
Zurück zum Zitat Dowling TC, Matzke GR, Murphy JE, Burckart GJ. Evaluation of renal drug dosing: prescribing information and clinical pharmacist approaches. Pharmacotherapy. 2010;30(8):776–86.CrossRef Dowling TC, Matzke GR, Murphy JE, Burckart GJ. Evaluation of renal drug dosing: prescribing information and clinical pharmacist approaches. Pharmacotherapy. 2010;30(8):776–86.CrossRef
26.
Zurück zum Zitat Yao X, Shah ND, Sangaralingham LR, Gersh BJ, Noseworthy PA. Reply: NOAC dosing in patients with atrial fibrillation and renal dysfunction. J Am Coll Cardiol. 2017;70(21):2734–5.CrossRef Yao X, Shah ND, Sangaralingham LR, Gersh BJ, Noseworthy PA. Reply: NOAC dosing in patients with atrial fibrillation and renal dysfunction. J Am Coll Cardiol. 2017;70(21):2734–5.CrossRef
27.
Zurück zum Zitat Dowling TC, Wang ES, Ferrucci L, Sorkin JD. Glomerular filtration rate equations overestimate creatinine clearance in older individuals enrolled in the Baltimore longitudinal study on aging: impact on renal drug dosing. Pharmacotherapy. 2013;33:912–21.CrossRef Dowling TC, Wang ES, Ferrucci L, Sorkin JD. Glomerular filtration rate equations overestimate creatinine clearance in older individuals enrolled in the Baltimore longitudinal study on aging: impact on renal drug dosing. Pharmacotherapy. 2013;33:912–21.CrossRef
28.
Zurück zum Zitat Park EJ, Wu K, Mi Z. A systematic comparison of the Cockcroft-Gault and modification of diet in renal disease equations for classification of kidney dysfunction and dosage adjustment. Ann Pharmacother. 2012;46:1174–87.CrossRef Park EJ, Wu K, Mi Z. A systematic comparison of the Cockcroft-Gault and modification of diet in renal disease equations for classification of kidney dysfunction and dosage adjustment. Ann Pharmacother. 2012;46:1174–87.CrossRef
30.
Zurück zum Zitat Schwartz J. Potential impact of substituting estimated glomerular filtration rate for estimated creatinine clearance for dosing of direct oral anticoagulants. J Am Geriatr Soc. 2016;64:1996–2002.CrossRef Schwartz J. Potential impact of substituting estimated glomerular filtration rate for estimated creatinine clearance for dosing of direct oral anticoagulants. J Am Geriatr Soc. 2016;64:1996–2002.CrossRef
Metadaten
Titel
Characteristics and outcomes in patients with atrial fibrillation receiving direct oral anticoagulants in off-label doses
verfasst von
Alexandros Briasoulis
Yubo Gao
Chakradhari Inampudi
Paulino Alvarez
Rabea Asleh
Elizabeth Chrischilles
Enrique C. Leira
Mary Vaughan-Sarrazin
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Cardiovascular Disorders / Ausgabe 1/2020
Elektronische ISSN: 1471-2261
DOI
https://doi.org/10.1186/s12872-020-01340-4

Weitere Artikel der Ausgabe 1/2020

BMC Cardiovascular Disorders 1/2020 Zur Ausgabe

Update Kardiologie

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