Xarelto

Xarelto

rivaroxaban

Manufacturer:

Bayer

Distributor:

Bayer
Full Prescribing Info
Contents
Rivaroxaban.
Description
2.5 mg: 1 film-coated tablet contains 2.5 mg rivaroxaban.
Each 2.5 mg film-coated tablet contains 35.70 mg lactose monohydrate (= 33.92 mg lactose) per tablet.
10 mg: 1 film-coated tablet contains 10 mg rivaroxaban.
Each 10 mg film-coated tablet contains 27.90 mg lactose monohydrate (=26.51 mg lactose) per tablet.
15 mg: Each film-coated tablet contains: Rivaroxaban 15 mg.
Each 15 mg film-coated tablet contains 25.4 mg lactose monohydrate (24.1 mg lactose) per tablet.
20 mg: Each film-coated tablet contains: Rivaroxaban 20 mg.
Each 20 mg film-coated tablet contains 22.9 mg lactose monohydrate (21.8 mg lactose) per tablet.
Excipients/Inactive Ingredients: Excipients: Cellulose microcrystalline, Croscarmellose sodium, Hypromellose 5 cP, Lactose monohydrate, Magnesium stearate, sodium lauryl sulfate.
Film-coating: Ferric oxide yellow (for 2.5 mg), Ferric oxide red (for 10 mg, 15 mg and 20 mg), Hypromellose 15 cP, Macrogol 3350, Titanium dioxide.
Action
Pharmacotherapeutic group: Direct factor Xa inhibitors. ATC Code: B01AF01.
Pharmacology: Pharmacodynamics: Mechanism of Action: Rivaroxaban is a highly selective direct factor Xa inhibitor with oral bioavailability.
Activation of factor X to factor Xa (FXa) via the intrinsic and extrinsic pathway plays a central role in the cascade of blood coagulation. FXa directly converts prothrombin to thrombin through the prothrombinase complex, and ultimately, this reaction leads to fibrin clot formation and activation of platelets by thrombin.
One molecule of FXa is able to generate more than 1000 molecules of thrombin due to the amplification nature of the coagulation cascade. In addition, the reaction rate of prothrombinase-bound FXa increases 300,000-fold compared to that of free FXa and causes an explosive burst of thrombin generation. Selective inhibitors of FXa can terminate the amplified burst of thrombin generation. Consequently, several specific and global clotting tests are affected by rivaroxaban. Dose dependent inhibition of factor Xa activity was observed in humans.
Pharmacodynamics effects: Dose-dependent inhibition of factor Xa activity was observed in humans. Prothrombin time (PT) is influenced by rivaroxaban in a dose dependent way with a close correlation to plasma concentrations (r value equals 0.98) if Neoplastin is used for the assay. Other reagents would provide different results. The readout for PT is to be done in seconds, because the INR (International Normalized Ratio) is only calibrated and validated for coumarins and cannot be used for any other anticoagulant.
2.5 and 10 mg: In patients undergoing major orthopedic surgery, the 5/95 percentiles for PT (Neoplastin) 2-4 hours after tablet intake (i.e. at the time of maximum effect) ranged from 13 to 25 sec.
In a clinical pharmacology study on the reversal of rivaroxaban pharmacodynamics in healthy adult subjects (n=22), the effects of single doses (50 IU/kg) of two different types of PCCs, a 3-factor PCC (Factors II, IX and X) and a 4-factor PCC (Factors II, VII, IX and X) were assessed. The 3-factor PCC reduced mean Neoplastin PT values by approximately 1.0 second within 30 minutes, compared to reductions of approximately 3.5 seconds observed with the 4-factor PCC. In contrast, the 3-factor PCC had a greater and more rapid overall effect on reversing changes in endogenous thrombin generation than the 4-factor PCC (see Overdosage).
15 and 20 mg: In patients receiving rivaroxaban for treatment of DVT and PE and prevention of recurrent DVT and PE, the 5/95 percentiles for PT (Neoplastin) 2-4 hours after tablet intake (i.e. at the time of maximum effect) ranged from 17 sec to 32 sec for 15 mg twice daily or 15 sec to 30 sec for 20 mg once daily, respectively.
In patients with non-valvular atrial fibrillation receiving rivaroxaban for the prevention of stroke and systemic embolism, the 5/95 percentiles for PT (Neoplastin) 1-4 hours after tablet intake (i.e. at the time of maximum effect) ranged from 14 sec to 40 sec in patients treated with 20 mg once daily and from 10 sec to 50 sec in patients with moderate renal impairment treated with 15 mg once daily.
The activated partial thromboplastin time (aPTT) and HepTest are also prolonged dose-dependently; however, they are not recommended to assess the pharmacodynamic effect of rivaroxaban. Anti-factor Xa activity is also influenced by rivaroxaban; however, no standard for calibration is available.
There is no need for monitoring of coagulation parameters during routine clinical treatment with Rivaroxaban (Xarelto).
Clinical efficacy and safety: 2.5 mg: The rivaroxaban clinical program was designed to demonstrate the efficacy of Rivaroxaban (Xarelto) for the prevention of cardiovascular (CV) death, MI, or stroke in subjects with a recent ACS (ST-elevation myocardial infarction [STEMI], non-ST-elevation myocardial infarction [NSTEMI] or unstable angina [UA]). Few patients with a prior stroke or TIA were included. The limited data in patients with a prior stroke or TIA do not support that 2.5 mg Rivaroxaban (Xarelto) BID in combination with ASA or ASA in addition to clopidogrel/ticlopidine provides adequate efficacy in these patients. In the pivotal double-blind ATLAS ACS 2 TIMI 51 trial, 15,526 patients were randomly assigned in a 1:1:1 fashion to one of 3 treatment groups: Rivaroxaban (Xarelto) 2.5 mg orally twice daily, 5 mg orally twice daily or to placebo twice daily. The median time on treatment was 13 months and overall treatment duration was up to almost 3 years.
93.2% of patients received concomitantly ASA plus thienopyridine treatment and 6.8% ASA only.
Among patients receiving dual anti-platelet therapy 98.8% received clopidogrel, 0.9 % received ticlopidine and 0.3 % received prasugrel.
Relative to placebo, Rivaroxaban (Xarelto) significantly reduced the primary composite endpoint of CV death, MI, or stroke. In addition the secondary endpoint 1 (all cause death, MI, or stroke; ) was also reduced significantly (see Table 1).
Subjects with a history of CHF derived substantial benefit from rivaroxaban treatment (see Table 1). An additional analysis showed differences in the incidence rates of stent thrombosis compared with placebo in the 2.5 mg bid (HR: 0.70, 95% CI: 0.51, 0.97) and 5 mg bid (HR: 0.70, 95% CI: 0.51, 0.98) groups (see Table 1). The incidence rates for the principal safety outcome (non-CABG TIMI major bleeding events) were higher in patients treated with Rivaroxaban (Xarelto) than in patients who received placebo (see Table 2), as were the incidence rates of life-threatening bleeding events, but were balanced between Rivaroxaban (Xarelto) and placebo for the components of fatal bleeding events, hypotension requiring treatment with intravenous inotropic agents and surgical intervention for ongoing bleeding.
Patients received the first dose of Rivaroxaban (Xarelto) at a minimum of 24 hours and up to 7 days (mean 4.7 days) after admission to hospital and as soon as possible after stabilization, of the index ACS event, including revascularization procedures, and when parenteral anticoagulation therapy would normally be discontinued.
Both the 2.5 mg twice daily and the 5 mg twice daily regimens of rivaroxaban were effective in further reducing the incidence of CV events on a background of standard antiplatelet care. The 2.5 mg twice daily regimen reduced mortality, and there is evidence that the lower dose had lower bleeding risks, therefore rivaroxaban 2.5 mg twice daily should be used for the prevention of CV events in patients after an ACS in combination with ASA alone or with ASA plus a thienopyridine. (See Tables 1 and 2.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

CAD or PAD (2.5 mg only): The Rivaroxaban (Xarelto) clinical program was designed to demonstrate the efficacy and safety of Rivaroxaban (Xarelto) for the prevention of stroke, myocardial infarction, or cardiovascular death in patients with CAD or PAD. In the pivotal phase III double-blind COMPASS trial, 27,395 patients were randomly assigned to one of three antithrombotic treatment groups: Rivaroxaban (Xarelto) 2.5 mg twice daily in combination with ASA 100 mg once daily, Rivaroxaban (Xarelto) 5 mg twice daily or to ASA 100 mg once daily in a 1:1:1 fashion. Patients with established CAD or PAD were eligible. Patients with CAD who were younger than 65 years of age were also required to have documentation of atherosclerosis involving at least two vascular beds or to have at least two additional cardiovascular risk factors (current smoking, diabetes mellitus, an estimated glomerular filtration rate [eGFR] <60 ml per minute, heart failure, or non-lacunar ischemic stroke ≥1 month earlier). Certain patients were excluded, such as those patients in need of dual antiplatelet, other non-ASA antiplatelet, or oral anticoagulant therapies, as well as patients with a history of ischemic, non-lacunar stroke within 1 month, any history of hemorrhagic or lacunar stroke, or patients with eGFR < 15 ml/min.
The mean duration of follow-up was 23 months and the maximum follow-up was 3.9 years. The mean age was 68 years and 21% of the subject population were ≥75 years. Of the patients included, 91% had CAD, 27% had PAD, and 18% had both CAD and PAD. Of the patients with CAD, 69% had prior myocardial infarction, 60% had prior percutaneous transluminal coronary angioplasty (PTCA)/atherectomy/percutaneous coronary intervention (PCI), and 26% had a history of coronary artery bypass grafting (CABG) prior to study. Of the patients with PAD, 49% had intermittent claudication, 27% had peripheral artery bypass surgery or peripheral percutaneous transluminal angioplasty (PTA), 26% had asymptomatic carotid artery stenosis >50%, and 5% had limb or foot amputation for arterial vascular disease.
Relative to ASA 100 mg, Rivaroxaban (Xarelto) 2.5 mg twice daily in combination with ASA 100 mg once daily was superior in the reduction of the primary composite outcome of stroke, myocardial infarction or cardiovascular death. The benefit was observed early with a constant treatment effect over the entire treatment period (see Table 3 and figure). The composite secondary outcomes (composites of coronary heart disease death, or cardiovascular death, with myocardial infarction, ischemic stroke, and acute limb ischemia) as well as all-cause mortality were reduced (see Table 3). Acute limb ischemic events were reduced (hazard ratio [HR] 0.55; 95% confidence interval [CI] 0.32-0.92). Amputations for cardiovascular reasons were also reduced (HR 0.48; 95% CI 0.26-0.89). The composite outcome of stroke, myocardial infarction and all-cause mortality was also reduced (HR 0.79; 95% CI 0.70-0.88; p=0.00005, post-hoc analysis). There was a significant 1.7-fold increase of the primary safety outcome (modified International Society on Thrombosis and Haemostasis [ISTH] major bleeding events) in patients treated with Rivaroxaban (Xarelto) 2.5 mg twice daily in combination with ASA 100 mg once daily compared to patients who received ASA 100 mg (see Table 5). However the incidence rates for fatal bleeding events, non-fatal symptomatic bleeding into a critical organ as well as intracranial bleeding events did not differ significantly. The prespecified composite outcome for net clinical benefit (cardiovascular death, myocardial infarction, stroke, fatal or symptomatic critical-organ bleeding events) was reduced (see Table 3). The results in patients with PAD, CAD, and both CAD and PAD were consistent with the overall efficacy and safety results (Table 4).
In the 3.8% of patients with a history of ischemic, non-lacunar stroke (median time since stroke: 5 years), the reduction of stroke, myocardial infarction, cardiovascular death, and the increase of major bleeding (net clinical benefit HR 0.64; 95%CI 0.4-1.0) were consistent with the overall population (see Precautions).
Relative to ASA 100 mg, Rivaroxaban (Xarelto) 5 mg twice daily alone did not significantly reduce the primary composite efficacy outcome of stroke, myocardial infarction or cardiovascular death (HR 0.90; 95% CI 0.79-1.03; p = 0.11490). The incidence rates for the primary safety outcome (modified ISTH major bleeding events) were significantly increased in patients treated with Rivaroxaban (Xarelto) 5 mg twice daily compared with patients who received ASA 100 mg once daily (HR 1.51; 95% CI 1.25-1.84; p = 0.00003). (See Tables 3, 4, 5 and figure.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image


Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

Treatment and prevention of recurrent DVT and PE (10, 15 and 20 mg only): The Rivaroxaban (Xarelto) clinical program was designed to demonstrate the efficacy of Rivaroxaban (Xarelto) in the initial and continued treatment of acute DVT and PE and prevention of recurrent DVT and PE.
Over 12,800 patients were studied in four randomized controlled phase III clinical studies (EINSTEIN DVT, EINSTEIN PE, EINSTEIN Extension and EINSTEIN CHOICE) and additionally a predefined analysis of the pooled Einstein DVT and Einstein PE studies was conducted (see Table 8).
The overall combined treatment duration in all studies was up to 21 months.
In EINSTEIN DVT, 3,449 patients with acute DVT were studied for the treatment of DVT and the prevention of recurrent DVT and PE. The treatment duration was up to 12 months depending on the clinical judgment of the investigator.
For the initial 3 week treatment of acute DVT 15 mg of Rivaroxaban (Xarelto) was administered twice daily. This was followed by 20 mg of Rivaroxaban (Xarelto) once daily.
In EINSTEIN PE, 4,832 patients with acute PE were studied for the treatment of PE and the prevention of recurrent DVT and PE. The treatment duration was up to 12 months depending on the clinical judgement of the investigator.
For the initial treatment of acute PE 15 mg rivaroxaban was administered twice daily for three weeks. This was followed by 20 mg rivaroxaban once daily.
In both the EINSTEIN DVT and the Einstein PE study, the comparator treatment regimen consisted of enoxaparin administered for at least 5 days in combination with vitamin K antagonist treatment until the PT/INR was in therapeutic range (≥2.0). Treatment was continued with a vitamin K antagonist dose-adjusted to maintain the PT/INR values within the therapeutic range of 2.0 to 3.0.
In EINSTEIN Extension, 1,197 patients with DVT or PE were studied for the prevention of recurrent DVT and PE. The treatment duration was up to 12 months depending on the clinical judgment of the investigator. Rivaroxaban (Xarelto) 20 mg once daily was compared with placebo.
EINSTEIN DVT, PE and Extension used the same pre-defined primary and secondary efficacy outcomes. The primary efficacy outcome was symptomatic recurrent VTE defined as the composite of recurrent DVT or fatal or non-fatal PE. The secondary efficacy outcome was defined as the composite of recurrent DVT, non-fatal PE and all-cause mortality.
In EINSTEIN CHOICE, 3,396 patients with confirmed symptomatic DVT and/or PE who completed 6-12 months of anticoagulant treatment were studied for the prevention of fatal PE or non-fatal symptomatic recurrent DVT or PE. Patients with an indication for continued therapeutic-dosed anticoagulation were excluded from the study. The treatment duration was up to 12 months depending on the individual randomization date (median: 351 days). Rivaroxaban (Xarelto) 20 mg once daily and Rivaroxaban (Xarelto) 10 mg once daily were compared with 100 mg acetylsalicylic acid once daily.
The primary efficacy outcome was symptomatic recurrent VTE defined as the composite of recurrent DVT or fatal or non-fatal PE. The secondary efficacy outcome was the composite of the primary efficacy outcome, MI, ischemic stroke, or non-CNS systemic embolism.
In the EINSTEIN DVT study (see Table 6) Rivaroxaban (Xarelto) was demonstrated to be non-inferior to enoxaparin/VKA for the primary outcome.
The prespecified NCB (primary efficacy outcome plus major bleeding events) was reported with a HR of 0.67 ((95% CI=0.47 0.95), nominal p value p=0.027) in favour of rivaroxaban.
The incidence rates for the principal safety outcome (major or clinically relevant non-major bleeding events) as well as the secondary safety outcome (major bleeding events), were similar for both treatment groups.
In the EINSTEIN PE study (see Table 7) Rivaroxaban (Xarelto) was demonstrated to be non-inferior to enoxaparin/VKA for the primary outcome (p=0.0026 (test for non-inferiority); hazard ratio: 1.12 (0.75 1.68)).
The prespecified NCB (primary efficacy outcome plus major bleeding events) was reported with a HR of 0.85 ((95% CI= 0.63 - 1.14), nominal p value p=0.275).
A prespecified pooled analysis of the outcome of the EINSTEIN DVT and PE studies was conducted (see Table 8).
In the EINSTEIN Extension study (see Table 9) Rivaroxaban (Xarelto) was superior to placebo for the primary and secondary efficacy outcomes. For the principal safety outcome (major bleeding events) there was a non-significant numerically higher incidence rate for patients treated with Rivaroxaban (Xarelto) 20 mg once daily compared to placebo. The secondary safety outcome (major or clinically relevant non-major bleeding events) showed higher rates for patients treated with Rivaroxaban (Xarelto) 20 mg once daily compared to placebo.
In the EINSTEIN CHOICE study Rivaroxaban (Xarelto) 20 mg and 10 mg were both superior to 100 mg acetylsalicylic acid for the primary efficacy outcome. The secondary efficacy outcome was significantly reduced when comparing Rivaroxaban (Xarelto) 20 mg or 10 mg vs. 100 mg acetylsalicylic acid. The principal safety outcome (major bleeding events) was similar for patients treated with Rivaroxaban (Xarelto) 20 mg and 10 mg once daily compared to 100 mg acetylsalicylic acid. The secondary safety outcome (non-major bleeding associated with treatment cessation of more than 14 days) was similar when comparing Rivaroxaban (Xarelto) 20 mg or 10 mg vs. 100 mg acetylsalicylic acid. Outcomes were consistent across the patients with provoked and unprovoked VTE (see Table 10).
In a prespecified net clinical benefit analysis (NCB) (primary efficacy outcome plus major bleeding events) of EINSTEIN CHOICE, a HR of 0.44 (95% CI 0.27 - 0.71, p=0.0009) for Rivaroxaban (Xarelto) 20 mg once daily vs 100 mg acetylsalicylic acid once daily and a HR of 0.32 (95% CI 0.18 - 0.55, p<0.0001) for Rivaroxaban (Xarelto) 10 mg once daily vs 100 mg acetylsalicylic acid once daily were reported. (See Tables 6, 7, 8, 9 and 10.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image


Click on icon to see table/diagram/image


Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

In addition to the phase III EINSTEIN program, a prospective, non-interventional, open-label cohort study (XALIA) with central outcome adjudication including recurrent VTE, major bleeding and death has been conducted. 5,142 patients with acute DVT were enrolled to investigate the long-term safety of rivaroxaban compared with standard-of-care anticoagulation therapy under real-world conditions. Rates of major bleeding, recurrent VTE and all-cause mortality for rivaroxaban were 0.7%, 1.4% and 0.5%, respectively. Hazard ratios comparing rivaroxaban and standard-of-care were adjusted to account for differences in patient baseline characteristics. Adjusted hazard ratios for major bleeding, recurrent VTE and all-cause mortality were 0.77 (95% CI 0.40-1.50), 0.91 (95% CI 0.54-1.54) and 0.51 (95% CI 0.24-1.07), respectively.
Rivaroxaban showed similar safety and efficacy compared to standard anticoagulation.
These results in patients who were observed in routine clinical practice are consistent with those observed in the EINSTEIN DVT study.
VTE prevention (10 mg only):
Prevention of venous thromboembolic events (VTE) in patients undergoing major orthopedic surgery of the lower limbs.
The rivaroxaban clinical program was designed to demonstrate the efficacy of Rivaroxaban (Xarelto) for the prevention of venous thromboembolic events (VTE), i.e. proximal and distal deep vein thrombosis (DVT) and pulmonary embolism (PE) in patients undergoing major orthopedic surgery of the lower limbs. Over 9,500 patients (7,050 in total hip replacement surgery - 2,531 in total knee replacement surgery) were studied in controlled randomized double-blind phase III clinical studies, the RECORD-program.
Rivaroxaban (Xarelto) 10 mg once daily started not earlier than 6 hours postoperatively was compared with enoxaparin 40 mg once daily started 12 hours preoperatively.
In three phase III studies (see Table 11) rivaroxaban significantly reduced the rate of total VTE (any venographically detected or symptomatic DVT, non-fatal PE or death) and major VTE (proximal DVT, non-fatal PE and VTE-related death), the pre-specified primary and major secondary efficacy endpoints. Furthermore in all three studies the rate of symptomatic VTE (symptomatic DVT, non-fatal PE, VTE-related death) was lower in Rivaroxaban (Xarelto) treated patients compared to patients treated with enoxaparin.
The main safety endpoint, major bleeding, showed comparable rates for patients treated with Rivaroxaban (Xarelto) 10 mg compared to enoxaparin 40 mg. (See Table 11.)

Click on icon to see table/diagram/image

The analysis of the pooled results of the phase III trials corroborated the data obtained in the individual studies regarding reduction of total VTE, major VTE and symptomatic VTE with Rivaroxaban (Xarelto) 10 mg once daily compared to enoxaparin 40 mg once daily.
In addition to the phase III RECORD program, a post-authorization, non-interventional, open-label cohort study (XAMOS) has been conducted in 17,413 patients undergoing major orthopedic surgery of the hip or knee, to compare rivaroxaban with other standard-of-care pharmacological thromboprophylaxis in real-life setting. Symptomatic VTE occurred in 57 (0.6%) patients in the rivaroxaban group (n=8,778) and 88 (1.0%) of patients in the standard-of-care group (n=8,635; HR 0.63; 95% CI 0.43-0.91); safety population). Major bleeding occurred in 35 (0.4%) and 29 (0.3%) of patients in the rivaroxaban and standard-of-care groups (HR 1.10; 95% CI 0.67-1.80). This non-interventional study confirmed the efficacy and safety results seen in the RECORD program.
SPAF (15 and 20 mg only): The Rivaroxaban (Xarelto) clinical program was designed to demonstrate the efficacy of Rivaroxaban (Xarelto) for the prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation (AF).
In the pivotal double-blind ROCKET AF study, 14,264 patients were assigned either to Rivaroxaban (Xarelto) 20 mg orally once daily (15 mg orally once daily in patients with moderate (CrC: <50-30 mL/min) renal impairment) or to warfarin titrated to a target INR of 2.5 (therapeutic range 2.0 to 3.0). The median time on treatment was 19 months and overall treatment duration was up to 41 months.
34.9% of patients were on ASA treatment and 11.4% were on class III antiarrythmics including amiodarone.
Relative to warfarin, Rivaroxaban (Xarelto) significantly reduced the primary composite endpoint of stroke and non-CNS systemic embolism. In addition major secondary endpoints (composite of stroke, non-CNS systemic embolism and vascular death and composite of stroke, non-CNS systemic embolism, MI and vascular death) were also reduced significantly (see Table 12). The incidence rates for the principal safety outcome (major and non-major clinically relevant bleeding events) were similar for both treatment groups (see Table 13). (See Tables 12 and 13.)

Click on icon to see table/diagram/image


Click on icon to see table/diagram/image

In addition to the phase III ROCKET AF study, a prospective, single-arm, post-authorization, non-interventional, open-label cohort study (XANTUS) with central outcome adjudication including thromoboembolic events and major bleeding has been conducted. 6,785 patients with non-valvular atrial fibrillation were enrolled for prevention of stroke and non-central nervous system (CNS) systemic embolism under real-world conditions. The mean CHADS2 score was 2.0 compared to a mean CHADS2 score of 3.5 in ROCKET AF. Major bleeding occurred in 2.1 per 100 patient years. Fatal haemorrhage was reported in 0.2 per 100 patient years and intracranial haemorrhage in 0.4 per 100 patient years. Stroke or non-CNS systemic embolism was recorded in 0.8 per 100 patient years. These observations from routine clinical practice are consistent with the results observed in the ROCKET AF study.
SPAF: Cardioversion: A prospective, randomized, open-label, multicenter, exploratory study with blinded endpoint evaluation (X-VERT) was conducted in 1504 patients (oral anticoagulant naïve and pre-treated) with non-valvular atrial fibrillation scheduled for cardioversion to compare rivaroxaban with dose-adjusted VKA (randomized 2:1), for the prevention of cardiovascular events. TEE- guided (1-5 days of pre-treatment) or conventional cardioversion (at least three weeks of pre-treatment) strategies were employed. The primary efficacy outcome (all stroke, transient ischemic attack, non-CNS systemic embolism, MI and cardiovascular death) occurred in 5 (0.5%) patients in the rivaroxaban group (n=978) and 5 (1.0%) patients in the VKA group (n=492; RR 0.50; 95% CI 0.15-1.73; modified ITT population). The principal safety outcome (major bleeding) occurred in 6 (0.6%) and 4 (0.8%) patients in the rivaroxaban (n=988) and VKA (n=499) groups, respectively (RR 0.76; 95% CI 0.21-2.67; safety population). This exploratory study showed comparable efficacy and safety between rivaroxaban and VKAs treatment groups in the setting of cardioversion.
SPAF: Patients who undergo PCI with stent placement: A randomized, open-label, multicenter study (PIONEER AF-PCI) was conducted in 2124 patients with non-valvular atrial fibrillation who underwent PCI with stent placement for primary atherosclerotic disease to compare safety of two rivaroxaban regimens and one VKA regimen. Patients were randomly assigned in a 1:1:1 fashion for an overall 12 month-therapy.
Group 1 received rivaroxaban 15 mg once daily (10 mg once daily in patients with CrCl: 30 to <50 mL/min) plus P2Y12 inhibitor. Group 2 received rivaroxaban 2.5 mg twice daily plus DAPT (dual antiplatelet therapy i.e. clopidogrel 75 mg [or alternate P2Y12 inhibitor] plus low-dose acetylsalicylic acid [ASA]) for 1, 6 or 12 months followed by rivaroxaban 15 mg (or 10 mg for subjects with CrCl: 30 to <50 mL/min) once daily plus low-dose ASA. Group 3 received dose-adjusted VKA plus DAPT for 1, 6 or 12 months followed by dose-adjusted VKA plus low-dose ASA.
The primary safety endpoint, clinically significant bleeding events, occurred in 109 (15.7%), 117 (16.6%), and 167 (24.0%) subjects in group 1, group 2, and group 3, respectively (HR 0.59; 95% CI 0.47-0.76; p<0.001, and HR 0.63; 95% CI 0.50-0.80; p<0.001, respectively). The secondary endpoint (composite of cardiovascular events CV death, MI, or stroke) occurred in 41 (5.9%), 36 (5.1%), and 36 (5.2%) subjects in the group 1, group 2, and group 3 respectively. Each of the rivaroxaban regimens showed a significant reduction in clinically significant bleeding events compared to the VKA regimen in patients with non-valvular atrial fibrillation who underwent a PCI with stent placement.
Special patient populations: Ethnic differences: (see Pharmacokinetics as follows).
Geriatric patients: see Pharmacokinetics as follows.
Gender: see Pharmacokinetics as follows.
Body weight: see Pharmacokinetics as follows.
Hepatic impairment: see Pharmacokinetics as follows.
Renal impairment: see Pharmacokinetics as follows.
Effect on QTc: No QTc prolonging effects were observed in healthy men and women older than 50 years.
Patients with prosthetic heart valves having recently undergone TAVR: In the randomised, open label, active-controlled, event-driven multicenter phase III GALILEO study 1644 patients were randomized to either a rivaroxaban-based strategy or an antiplatelet-based strategy 26 / Xarelto (SPAF, DVT, PE) / 10, 15, 20 mg film-coated tablets / 1-7 days after a successful transcatheter aortic valve replacement. Patients with previous atrial fibrillation or with an ongoing indication for oral anticoagulation were excluded.
The main objective was to assess the efficacy and safety of a rivaroxaban-based treatment strategy (10 mg rivaroxaban od plus 75-100 mg acetylsalicylic acid (ASA) od for 90 days followed by rivaroxaban 10 mg od) compared to standard of care (clopidogrel 75 mg od plus 75-100 mg ASA od for 90 days followed by ASA od). The study was terminated early due to an imbalance in death and thromboembolic events.
In the intention-to-treat (ITT) analysis the primary efficacy endpoint, i.e. death and thromboembolic events, occurred in 105 patients (9.8 per 100 patient years) in the rivaroxaban arm and in 78 patients (7.21 per 100 patient years) in the antiplatelet arm; the HR was 1.35 (95% CI: 1.01; 1.81). In the on-treatment analysis the primary efficacy outcome occurred in 68 patients (8.11 per 100 patient years) in the rivaroxaban arm compared to 63 (6.6 per 100 patient years) in the antiplatelet arm; the HR was 1.21 (95% CI: 0.86; 1.70).
In the ITT analysis the primary safety endpoint, i.e. composite of life-threatening, disabling or major bleeding, occurred in 46 patients (4.29 per 100 patient years) in the rivaroxaban arm compared to 31 (2.83 per 100 patient years) in the antiplatelet arm; the HR was 1.50 (95% CI 0.95; 2.37).
Patients with high risk triple positive antiphospholipid syndrome: In an investigator sponsored randomized open-label multicenter study with blinded endpoint adjudication, rivaroxaban was compared to warfarin in patients with a history of thrombosis, diagnosed with antiphospholipid syndrome and at high risk for thromboembolic events (positive for all 3 antiphospholipid tests: lupus anticoagulant, anticardiolipin antibodies, and anti-beta 2-glycoprotein I antibodies). The trial was terminated prematurely after the enrollment of 120 patients due to an excess of events among patients in the rivaroxaban arm. Mean follow-up was 569 days. Fifty-nine patients were randomized to rivaroxaban 20mg (15 mg for patients with creatinine clearance <50 mL/min) and 61 to warfarin (INR 2.0-3.0). Thromboembolic events occurred in 12% of patients randomized to rivaroxaban(4 ischaemic stroke and 3 myocardial infarction). No events were reported in patients randomized to warfarin. Major bleeding occurred in 4 patients (7%) of the rivaroxaban group and 2 patients (3%) of the warfarin group.
Pharmacokinetics: Absorption and Biovailability: Rivaroxaban is rapidly absorbed with maximum concentrations (Cmax) appearing 2-4 hours after tablet intake.
Oral absorption of rivaroxaban is almost complete and oral bioavailability is high (80-100%) for the 2.5 mg and 10 mg tablet dose, irrespective of fasting/fed conditions. Intake with food does not affect rivaroxaban AUC or Cmax at the 10 mg dose. Rivaroxaban (Xarelto) 2.5 mg tablets and 10 mg tablets can be taken with or without food (see Dosage & Administration).
Due to reduced extent of absorption an oral bioavailability of 66% was determined for the 20 mg tablet under fasting conditions. When Rivaroxaban (Xarelto) 20 mg tablets are taken together with food increases in mean AUC by 39% were observed when compared to tablet intake under fasting conditions, indicating almost complete absorption and high oral bioavailability. Rivaroxaban (Xarelto) 15 mg and 20 mg should be taken with food (see Dosage & Administration).
Under fed condition Rivaroxaban (Xarelto) 10 mg, 15 mg and 20 mg demonstrated dose-proportionality.
Variability in rivaroxaban pharmacokinetics is moderate with inter-individual variability (CV%) ranging from 30% to 40%.
Absorption of rivaroxaban is dependent on the site of drug release in the GI tract. A 29% and 56% decrease in AUC and Cmax compared to tablet was reported when rivaroxaban granulate is released in the proximal small intestine. Exposure is further reduced when drug is released in the distal small intestine, or ascending colon. Avoid administration of rivaroxaban distal to the stomach which can result in reduced absorption and related drug exposure.
Bioavailability (AUC and Cmax) was comparable for 20 mg rivaroxaban administered orally as a crushed tablet mixed in applesauce, or suspended in water and administered via a gastric tube followed by a liquid meal, compared to a whole tablet. Given the predictable dose-proportional pharmacokinetic profile of rivaroxaban, the bioavailability results from this study are likely applicable to lower rivaroxaban doses.
Distribution: Plasma protein binding in humans is high at approximately 92% to 95%, with serum albumin being the main binding component. The volume of distribution is moderate with Vss being approximately 50 L.
Metabolism and Elimination: Of the administered rivaroxaban dose, approximately 2/3 undergoes metabolic degradation, with half then eliminated renally and the other half eliminated by the fecal route. The other 1/3 of the administered dose undergoes direct renal excretion as unchanged active substance in the urine, mainly via active renal secretion.
Rivaroxaban is metabolized via CYP 3A4, CYP 2J2 and CYP-independent mechanisms. Oxidative degradation of the morpholinone moiety and hydrolysis of the amide bonds are the major sites of biotransformation. Based on in vitro investigations rivaroxaban is a substrate of the transporter proteins P-gp (P-glycoprotein) and Bcrp (breast cancer resistance protein).
Unchanged rivaroxaban is the most important compound in human plasma with no major or active circulating metabolites being present. With a systemic clearance of about 10 L/h rivaroxaban can be classified as low-clearance drug. Elimination of rivaroxaban from plasma occurred with terminal half-lives of 5 to 9 hours in young individuals, and with terminal half-lives of 11 to 13 hours in the elderly.
Geriatric patients: Elderly patients exhibited higher plasma concentrations than younger patients with mean AUC values being approximately 1.5-fold higher, mainly due to reduced (apparent) total and renal clearance (see Dosage & Administration).
Gender: There were no clinically relevant differences in pharmacokinetics between male and female patients (see Dosage & Administration).
Body weight: Extremes in body weight (<50 kg vs >120 kg) had only a small influence on rivaroxaban plasma concentrations (less than 25%) (see Dosage & Administration).
Children and adolescents: Safety and efficacy have not been established for children and adolescents below 18 years. (see Dosage & Administration).
Ethnic differences: No clinically relevant interethnic differences among Caucasia, African-American, Hispanic, Japanese or Chinese patients were observed regarding pharmacokinetics and pharmacodynamics (see Dosage & Administration).
Hepatic impairment: The effect of hepatic impairment on rivaroxaban pharmacokinetics has been studied in subjects categorized according to the Child Pugh classification, a standard procedure in clinical development. The Child Pugh classification's original purpose is to assess the prognosis of chronic liver disease, mainly cirrhosis. In patients for whom anticoagulation is intended, the critical aspect of liver impairment is the reduced synthesis of normal coagulation factors in the liver. Since this aspect is captured by only one of the five clinical/biochemical measurements composing the Child Pugh classification system, the bleeding risk in patients may not clearly correlate with this classification scheme. The decision to treat patients with an anticoagulant should therefore be made independently of the Child Pugh classification.
Rivaroxaban (Xarelto) is contraindicated in patients with hepatic disease which is associated with coagulopathy leading to a clinically relevant bleeding risk.
Cirrhotic patients with mild hepatic impairment (classified as Child Pugh A) exhibited only minor changes in rivaroxaban pharmacokinetics (1.2-fold increase in rivaroxaban AUC on average), nearly comparable to their matched healthy control group. No relevant difference in pharmacodynamic properties was observed between these groups.
In cirrhotic patients with moderate hepatic impairment (classified as Child Pugh B), rivaroxaban mean AUC was significantly increased by 2.3-fold compared to healthy volunteers, due to significantly impaired drug clearance which indicates significant liver disease. Unbound AUC was increased 2.6-fold. There are no data in patients with severe hepatic impairment.
The inhibition of factor Xa activity was increased by a factor of 2.6 as compared to healthy volunteers; prolongation of PT was similarly increased by a factor of 2.1. The global clotting test PT assesses the extrinsic pathway that comprises of the coagulation factors VII, X, V, II and I which are synthesized in the liver. Patients with moderate hepatic impairment were more sensitive to rivaroxaban resulting in a steeper PK/PD relationship between concentration and PT.
No data are available for Child Pugh C patients (see Dosage & Administration and Contraindications).
Renal impairment: There was an increase in rivaroxaban exposure being inversely correlated to the decrease in renal function, as assessed via creatinine clearance measurements.
In individuals with mild (CrC: ≤80-50 mL/min), moderate (CrC: <50-30 mL/min) or severe (CrC: <30-15 mL/min) renal impairment, rivaroxaban plasma concentrations (AUC) were 1.4, 1.5 and 1.6-fold increased respectively as compared to healthy volunteers (see Dosage & Administration and Precautions).
Corresponding increases in pharmacodynamic effects were more pronounced (see Dosage & Administration and Precautions).
In individuals with mild, moderate or severe renal impairment the overall inhibition of factor Xa activity was increased by a factor of 1.5, 1.9 and 2.0 respectively as compared to healthy volunteers; prolongation of PT was similarly increased by a factor of 1.3, 2.2 and 2.4 respectively.
There are no data in patients with CrC <15 mL/min.
Use is not recommended in patients with creatinine clearance <15 mL/min. Rivaroxaban (Xarelto) is to be used with caution in patients with severe renal impairment creatinine clearance 15-30 mL/min (see Dosage & Administration and Precautions).
Due to the underlying disease patients with severe renal impairment are at an increased risk of both bleeding and thrombosis.
Concomitant administration of strong CYP 3A4 inducers: In a phase I trial, co-administration of Rivaroxaban (Xarelto) with the strong CYP 3A4 and P-gp inducer rifampicin led to an approximate 50% decrease in mean rivaroxaban AUC, with parallel decreases in its pharmacodynamic effects (see Interactions). In a phase IIa trial, the PK/PD of an adapted rivaroxaban dosing regimen (30 mg twice daily in the first 3 weeks of treatment, followed by 20 mg twice daily) has been studied in 19 patients treated for DVT or PE and who concomitantly were medicated with a strong CYP 3A4 and P-gp inducer (rifampicin or phenytoin). The adapted dosing regimen in these patients led to a similar exposure and pharmacodynamics when compared to patients treated for DVT (15 mg twice daily in the first 3 weeks of treatment, followed by 20 mg once daily) without the concomitant administration of a strong CYP 3A4 inducer.
Toxicology: Preclinical safety data: The non-clinical safety evaluation in the data from conventional and appropriate studies of safety pharmacology, single and repeat-dose toxicity, genotoxicity, phototoxicity, and carcinogenicity and toxicity to reproduction reveal no special hazard for humans.
No organ-specific toxicity of rivaroxaban was observed up to the highest doses tested.
Safety Pharmacology: Cardiovascular, respiratory, and CNS functions were not affected. No pro-arrhythmogenic potential was observed.
No clinically relevant effects on gastro-intestinal motility, liver function, renal function, and blood glucose levels were observed.
Acute and repeat-dose toxicity: Rivaroxaban showed low acute toxicity in rats and mice.
Rivaroxaban was tested in repeat dose studies up to 6 months in rats and up to 12 months in dogs. Based on the pharmacological mode of action a NOEL could not be established due to effects on clotting time. All adverse findings except for a slight body weight gain reduction in rats and dogs could be related to an exaggerated pharmacological effect of the compound. In dogs at very high exposures severe spontaneous bleedings were observed. The NOAELs after chronic exposure are 12.5 mg/kg in rats and 5 mg/kg in dogs.
Carcinogenicity: Rivaroxaban was tested up to 60 mg/kg/day reaching exposure levels similar to humans (mice) or up to 3.6-fold higher (rats) than in humans.
Rivaroxaban showed no carcinogenic potential in rats and mice.
Reproductive toxicology: Rivaroxaban was tested in developmental toxicity studies at exposure levels of up to 14-fold (rat) and up to 33-fold (rabbit) above the therapeutic exposure in humans. The toxicological profile is mainly characterized by maternal toxicity due to exaggerated pharmacodynamic effects.
Up to the highest dose tested no primary teratogenic potential was identified (see Use in Pregnancy & Lactation).
[14C] rivaroxaban-related radioactivity penetrated the placental barrier in rats. In none of the fetal organs and tissues the exposure in terms of maximum concentrations or AUC exceeded the maternal blood exposure. The average exposure in the fetuses based on AUC (0-24) reached about 20% of the exposure in maternal blood. The mammary glands had an approximately blood-equivalent AUC which indicates secretion of radioactivity into milk (see Use in Pregnancy & Lactation).
Rivaroxaban did not show an effect on male or female fertility up to 200 mg/kg (see Use in Pregnancy & Lactation).
Lactation: [14C] rivaroxaban was administered orally to lactating Wistar rats (day 8 to 10 post partum) as a single oral dose of 3 mg/kg body weight.
[14C] rivaroxaban-related radioactivity was secreted into the milk of lactating rats only to a low extent in relation to the administered dose: The estimated amount of radioactivity excreted with milk was 2.12% of the maternal dose within 32 hours after administration (see see Use in Pregnancy & Lactation).
Genotoxicity: No genotoxicity was observed in a test for gene mutation in bacteria (Ames-Test), an in vitro test for chromosomal aberrations or in the in vivo micronucleus test.
Indications/Uses
2.5 mg: Rivaroxaban (Xarelto) is indicated for the prevention of cardiovascular death, myocardial infarction and stent thrombosis in patients after an acute coronary syndrome (ACS) (non-ST elevation or ST elevation myocardial infarction or unstable angina) in combination with acetylsalicylic acid (ASA) alone or with ASA plus thienopyridines clopidogrel or ticlopidine (see Dosage & Administration).
Rivaroxaban (Xarelto) is indicated for the prevention of stroke, myocardial infarction and cardiovascular death, and for the prevention of acute limb ischemia and mortality in patients with coronary artery disease (CAD) or peripheral artery disease (PAD) in combination with ASA (see Dosage & Administration).
10 mg: Rivaroxaban (Xarelto) is indicated for the prevention of venous thromboembolism (VTE) in adult patients undergoing elective hip or knee replacement surgery.
Rivaroxaban (Xarelto) is indicated for the treatment of Deep Vein Thrombosis (DVT) and pulmonary embolism (PE), and prevention of recurrent DVT and PE.
15 and 20 mg: Rivaroxaban (Xarelto) is indicated for prevention of stroke and systemic embolism in patients with non-valvular atrial fibrillation (SPAF).
Rivaroxaban (Xarelto) is indicated for the treatment of Deep Vein Thrombosis (DVT) and for the prevention of recurrent DVT and Pulmonary Embolism (PE).
Rivaroxaban (Xarelto) is indicated for the treatment of Pulmonary Embolism (PE) and for the prevention of recurrent PE and DVT.
Dosage/Direction for Use
2.5 mg: ACS: Method of administration: Oral use.
ACS: Recommended usual dose: After an acute coronary syndrome, the recommended vascular protection regimen is one tablet of 2.5 mg Rivaroxaban (Xarelto) twice daily. Patients should also take a daily dose of 75-100 mg ASA or a daily dose of 75-100 mg ASA in addition to a daily dose of 75 mg clopidogrel or a standard daily dose of ticlopidine.
ACS: Duration of treatment: Treatment is recommended for at least 24 months.
Patients after ACS continue to be at risk for cardiovascular events and therefore may benefit from extended treatment.
ACS: Method and frequency of administration: Treatment with Rivaroxaban (Xarelto) 2.5 mg should be started as soon as possible after stabilization of the index ACS event (including revascularization procedures). Rivaroxaban (Xarelto) should be started earliest 24 hours after admission to hospital. Rivaroxaban (Xarelto) 2.5 mg should be started at the time when parenteral anticoagulation therapy would normally be discontinued.
One 2.5 mg tablet of Rivaroxaban (Xarelto) should be taken twice daily.
Rivaroxaban (Xarelto) 2.5 mg tablets may be taken with or without food.
For patients who are unable to swallow whole tablets, Rivaroxaban (Xarelto) tablet may be crushed and mixed with water or soft foods such as applesauce immediately prior to use and administered orally.
The crushed Rivaroxaban (Xarelto) tablet may be given through gastric tubes. Gastric placement of the tube should be confirmed before administering Rivaroxaban (Xarelto). The crushed tablet should be administered in a small amount of water via a gastric tube after which it should be flushed with water. (see Pharmacology: Pharmacokinetics under Actions).
ACS: Missed Dose: If a dose is missed the patient should continue with the regular 2.5 mg Rivaroxaban (Xarelto) dose as recommended at the next scheduled time.
CAD or PAD: Method of administration: Oral use.
CAD or PAD: Recommended usual dose: The recommended vascular protection regimen for patients with CAD or PAD is one tablet of 2.5 mg Rivaroxaban (Xarelto) twice daily in combination with a daily dose of 75-100 mg ASA.
CAD or PAD: Duration of treatment: Therapy with Rivaroxaban (Xarelto) should be continued long term provided the benefit outweighs the risk.
In patients with an acute thrombotic event or vascular procedure and a need for dual antiplatelet therapy, the continuation of Rivaroxaban (Xarelto) 2.5 mg twice daily should be evaluated depending on type of event or procedure and antiplatelet regimen. Safety and efficacy of Rivaroxaban (Xarelto) 2.5 mg twice daily in combination with ASA plus clopidogrel/ticlopidine has only been studied in patients with recent ACS. Dual antiplatelet therapy has not been studied in combination with Rivaroxaban (Xarelto) 2.5 mg twice daily in patients with CAD or PAD (see Pharmacology: Pharmacodynamics under Actions).
CAD or PAD: Method and frequency of administration: In patients diagnosed with CAD or PAD, treatment with Rivaroxaban (Xarelto) 2.5 mg twice daily in combination with ASA 75-100 mg once daily can be started at any time.
One 2.5 mg tablet of Rivaroxaban (Xarelto) should be taken twice daily.
Rivaroxaban (Xarelto) 2.5 mg tablets may be taken with or without food.
For patients who are unable to swallow whole tablets, Rivaroxaban (Xarelto) tablet may be crushed and mixed with water or soft foods such as applesauce immediately prior to use and administered orally.
The crushed Rivaroxaban (Xarelto) tablet may be given through gastric tubes. Gastric placement of the tube should be confirmed before administering Rivaroxaban (Xarelto). The crushed tablet should be administered in a small amount of water via a gastric tube after which it should be flushed with water (see Pharmacology: Pharmacokinetics under Actions).
CAD or PAD: Missed Dose: If a dose is missed, the patient should continue with the regular Rivaroxaban (Xarelto) dose as recommended at the next scheduled time.
Additional information on special populations: Patients with hepatic impairment: Rivaroxaban (Xarelto) is contraindicated in patients with hepatic disease which is associated with coagulopathy leading to a clinically relevant bleeding risk (see Contraindications).
No dose adjustment is necessary in patients with other hepatic diseases (see Pharmacology: Pharmacokinetics under Actions).
Limited clinical data in patients with moderate hepatic impairment (Child Pugh B) indicate a significant increase in the pharmacological activity. No clinical data are available for patients with severe hepatic impairment (Child Pugh C) (see Contraindications and Pharmacology: Pharmacokinetics under Actions).
Patients with renal impairment: No dose adjustment is required if Rivaroxaban (Xarelto) is administered in patients with mild (Creatinine clearance (CrC): ≤80-50 mL/min) or moderate (CrC: <50-30 mL/min) renal impairment (see Pharmacology: Pharmacokinetics under Actions).
Limited clinical data for patients with severe renal impairment (CrC: <30-15 mL/min) indicate that rivaroxaban plasma levels are significantly increased in this patient population. Therefore Rivaroxaban (Xarelto) must be used with caution in these patients (see Precautions and Pharmacology: Pharmacokinetics under Actions).
Use of Rivaroxaban (Xarelto) is not recommended in patients with CrC <15 mL/min (see Precautions and Pharmacology: Pharmacokinetics under Actions).
Converting from Vitamin K Antagonists (VKA) to Rivaroxaban (Xareltomisse): When converting patients from VKAs to Rivaroxaban (Xarelto), INR values will be falsely elevated after the intake of Rivaroxaban (Xarelto). The INR is not valid to measure the anticoagulant activity of Rivaroxaban (Xarelto), and therefore should not be used (see Interactions).
Converting from Rivaroxaban (Xarelto) to Vitamin K antagonists (VKA): There is a potential for inadequate anticoagulation during the transition from Rivaroxaban (Xarelto) to VKA. Continuous adequate anticoagulation should be ensured during any transition to an alternate anticoagulant. It should be noted that Rivaroxaban (Xarelto) can contribute to an elevated INR.
In patients converting from Rivaroxaban (Xarelto) to VKA, VKA should be given concurrently until the INR is ≥2.0. For the first two days of the conversion period, standard VKA dosing should be used followed by VKA dosing guided by INR testing. While patients are on both Rivaroxaban (Xarelto) and VKA, the INR should not be tested earlier than 24hours (after the previous dose but prior to the next dose of Rivaroxaban (Xarelto). Once Rivaroxaban (Xarelto) is discontinued INR testing may be done reliably 24 hours after the last dose (see Interactions).
Converting from parenteral anti-coagulants to Rivaroxaban (Xarelto): For patients currently receiving a parenteral anticoagulant, start Rivaroxaban (Xarelto) 0 to 2 hours before the time of the next scheduled administration of the parenteral drug (e.g. LMWH) or at the time of discontinuation of a continuously administered parenteral drug (e.g. intravenous unfractionated heparin).
Converting from Rivaroxaban (Xarelto) to parenteral anti-coagulants: Discontinue Rivaroxaban (Xarelto) and give the first dose of parenteral anticoagulant at the time that the next Rivaroxaban (Xarelto) dose would be taken.
Children and adolescents (from birth to 16 or 18 years depending on local law): Safety and efficacy have not been established in children and adolescents below 18 years.
Geriatric patients: No dose adjustment is required based on age (see Pharmacology: Pharmacokinetics under Actions).
Gender: No dose adjustment is required based on gender (see Pharmacology: Pharmacokinetics under Actions).
Body weight: No dose adjustment is required based on body weight (see Pharmacology: Pharmacokinetics under Action).
Ethnic differences: No dose adjustment is required based on ethnic differences (see Pharmacology: Pharmacokinetics under Action).
10, 15 and 20 mg: Treatment and prevention of recurrent DVT and PE: Method of administration: Oral use.
Treatment and prevention of recurrent DVT and PE: Recommended usual dose: The recommended dose for the initial treatment of acute DVT and PE is Rivaroxaban (Xarelto) 15 mg twice daily for the first three weeks followed by Rivaroxaban (Xarelto) 20 mg once daily for the continued treatment and the prevention of recurrent DVT and PE. Following completion of at least 6 months treatment for DVT or PE, Rivaroxaban (Xarelto) 10 mg once daily or Rivaroxaban (Xarelto) 20 mg once daily is recommended based on an individual assessment of the risk of recurrent DVT or PE against the risk for bleeding. See Table 14.

Click on icon to see table/diagram/image

Treatment and prevention of recurrent DVT and PE: Duration of treatment: Therapy should be continued as long as the VTE risk persist (see Pharmacology: Pharmacodynamics: SPAF: Clinical efficacy and safety under Actions).
Treatment and prevention of recurrent DVT and PE: Method and frequency of administration: During the initial 3 weeks of acute treatment 15 mg of Rivaroxaban (Xarelto) should be taken twice daily.
After the initial 3 weeks treatment Rivaroxaban (Xarelto) should be continued at 20 mg once daily.
After at least 6 months treatment Rivaroxaban (Xarelto) should be taken at 10 mg once daily or 20 mg once daily (see Pharmacology: Pharmacodynamics under Actions).
Rivaroxaban (Xarelto) 15 mg tablets and Rivaroxaban (Xarelto) 20 mg tablets should be taken with food.
Rivaroxaban (Xarelto) 10 mg tablets may be taken with or without food (see Pharmacology: Pharmacokinetics under Actions).
For patients who are unable to swallow whole tablets, Rivaroxaban (Xarelto) tablet may be crushed and mixed with water or soft foods such as applesauce immediately prior to use and administered orally. After the administration of crushed Rivaroxaban (Xarelto) 15 mg or Rivaroxaban (Xarelto) 20 mg tablets, the dose should be immediately followed by food.
The crushed Rivaroxaban (Xarelto) tablet may be given through gastric tubes. Gastric placement of the tube should be confirmed before administering Rivaroxaban (Xarelto). The crushed tablet should be administered in a small amount of water via a gastric tube after which it should be flushed with water. After the administration of crushed Rivaroxaban (Xarelto)15 mg or 20 mg tablets, the dose should then be immediately followed by enteral feeding (see Pharmacology: Pharmacokinetics under Actions).
Treatment and prevention of recurrent DVT and PE: Missed Dose: It is essential to adhere to the dosage schedule provided.
If a dose is missed during the 15 mg twice daily treatment phase the patient should take Rivaroxaban (Xarelto) immediately to ensure intake of 30 mg Rivaroxaban (Xarelto) per day. In this case two 15 mg tablets may be taken at once. The patient should continue with the regular 15 mg twice daily intake as recommended on the following day.
If a dose is missed during the once daily treatment phase the patient should take Rivaroxaban (Xarelto) immediately to ensure intake of the recommended daily dose. The patient should continue with the regular once daily dose as recommended on the following day.
Treatment and prevention of recurrent DVT and PE: Maximum daily dose: The recommended maximum daily dose is 30 mg during the first 3 weeks of treatment.
In the following treatment phase the recommended maximum daily dose is 20 mg.
Treatment and prevention of recurrent DVT and PE: Additional information on special populations: Treatment and prevention of recurrent DVT and PE: Patients with hepatic impairment: Rivaroxaban (Xarelto) is contraindicated in patients with hepatic disease which is associated with coagulopathy leading to a clinically relevant bleeding risk (see Contraindications).
No dose adjustment is necessary in patients with other hepatic diseases (see Pharmacology: Pharmacokinetics under Actions).
Limited clinical data in patients with moderate hepatic impairment (Child Pugh B) indicate a significant increase in the pharmacological activity (see Pharmacology: Pharmacokinetics under Actions).
No clinical data are available for patients with severe hepatic impairment (Child Pugh C) (see Contraindications and Pharmacology: Pharmacokinetics under Actions).
Treatment and prevention of recurrent DVT and PE: Patients with renal impairment: No dose adjustment is required if Rivaroxaban (Xarelto) is administered in patients with mild (Creatinine clearance (CrC): ≤80-50 mL/min) or moderate (CrC: <50-30 mL/min) renal impairment (see Pharmacology: Pharmacokinetics under Actions).
Limited clinical data for patients with severe renal impairment (CrC: <30-15 mL/min) indicate that rivaroxaban plasma levels are significantly increased in this patient population. Therefore Rivaroxaban (Xarelto) must be used with caution in these patients (see Precautions and Pharmacology: Pharmacokinetics under Actions).
Use of Rivaroxaban (Xarelto) is not recommended in patients with CrC: <15 mL/min (see Precautions and Pharmacology: Pharmacokinetics under Actions).
Treatment and prevention of recurrent DVT and PE: Converting from Vitamin K Antagonists (VKA) to Rivaroxaban (Xarelto): VKA treatment should be stopped and Rivaroxaban (Xarelto) therapy should be initiated once the INR is ≤ 2.5.
When converting patients from VKAs to Rivaroxaban (Xarelto), INR values will be falsely elevated after the intake of Rivaroxaban (Xarelto). The INR is not valid to measure the anticoagulant activity of Rivaroxaban (Xarelto), and therefore should not be used (see Interactions).
Treatment and prevention of recurrent DVT and PE: Converting from Rivaroxaban (Xarelto) to Vitamin K antagonists (VKA): There is a potential for inadequate anticoagulation during the transition from Rivaroxaban (Xarelto) to VKA. Continuous adequate anticoagulation should be ensured during any transition to an alternate anticoagulant. It should be noted that Rivaroxaban (Xarelto) can contribute to an elevated INR.
In patients converting from Rivaroxaban (Xarelto) to VKA, VKA should be given concurrently until the INR is ≥2.0. For the first two days of the conversion period, standard VKA dosing should be used followed by VKA dosing guided by INR testing. While patients are on both Rivaroxaban (Xarelto) and VKA, the INR should not be tested earlier than 24 hours (after the previous dose but prior to the next dose of Rivaroxaban (Xarelto). Once Rivaroxaban (Xarelto) is discontinued INR testing may be done reliably 24 hours after the last dose (see Interactions).
Treatment and prevention of recurrent DVT and PE: Converting from parenteral anti-coagulants to Rivaroxaban (Xarelto): For patients currently receiving a parenteral anticoagulant, start Rivaroxaban (Xarelto) 0 to 2 hours before the time of the next scheduled administration of the parenteral drug (e.g. LMWH) or at the time of discontinuation of a continuously administered parenteral drug (e.g. intravenous unfractionated heparin).
Treatment and prevention of recurrent DVT and PE: Converting from Rivaroxaban (Xarelto) to parenteral anti-coagulants: Discontinue Rivaroxaban (Xarelto) and give the first dose of parenteral anticoagulant at the time that the next Rivaroxaban (Xarelto) dose would be taken.
Treatment and prevention of recurrent DVT and PE: Children and adolescents (from birth to 16 or 18 years depending on local law): Safety and efficacy have not been established in children and adolescents below 18 years.
Treatment and prevention of recurrent DVT and PE: Geriatric patients: No dose adjustment is required based on age (see Pharmacology: Pharmacokinetics under Actions).
Treatment and prevention of recurrent DVT and PE: Gender: No dose adjustment is required based on gender (see Pharmacology: Pharmacokinetics under Actions).
Treatment and prevention of recurrent DVT and PE: Body weight: No dose adjustment is required based on body weight (see Pharmacology: Pharmacokinetics under Actions).
Treatment and prevention of recurrent DVT and PE: Ethnic differences: No dose adjustment is required based on ethnic differences (see Pharmacology: Pharmacokinetics under Actions).
15 and 20 mg: SPAF: Method of administration: Oral use.
SPAF: Recommended usual dose: The recommended dose is 20 mg once daily.
For patients with moderate renal impairment (creatinine clearance (CrC): <50-30 mL/min) the recommended dose is 15 mg once daily.
SPAF: Duration of treatment: Therapy should be continued as long as risk factors for stroke and systemic embolism persist.
SPAF: Method and frequency of administration: One 20 mg tablet of Rivaroxaban (Xarelto) should be taken once daily.
For patients with moderate renal impairment (CrC: <50-30 mL/min) one 15 mg tablet of Rivaroxaban (Xarelto) should be taken once daily.
Rivaroxaban (Xarelto) 15 mg tablets and Rivaroxaban (Xarelto) 20 mg tablets should be taken with food (see Pharmacology: Pharmacokinetics under Actions).
For patients who are unable to swallow whole tablets, Rivaroxaban (Xarelto) tablet may be crushed and mixed with water or soft foods such as applesauce immediately prior to use and administered orally. After the administration of crushed Rivaroxaban (Xarelto) 15 mg or Rivaroxaban (Xarelto) 20 mg tablets, the dose should be immediately followed by food.
The crushed Rivaroxaban (Xarelto) tablet may be given through gastric tubes. Gastric placement of the tube should be confirmed before administering Rivaroxaban (Xarelto). The crushed tablet should be administered in a small amount of water via a gastric tube after which it should be flushed with water. After the administration of crushed Rivaroxaban (Xarelto) 15 mg or 20 mg tablets, the dose should then be immediately followed by enteral feeding (see Pharmacology: Pharmacokinetics under Actions).
SPAF: Missed Dose: If a dose is missed the patient should take Rivaroxaban (Xarelto) immediately and continue with the once daily intake as recommended on the following day.
The dose should not be doubled to make up for a missed dose within the same day.
SPAF: Maximum daily dose: The recommended maximum daily dose is 20 mg.
SPAF: Additional information on special populations: SPAF: Patients with hepatic impairment: Rivaroxaban (Xarelto) is contraindicated in patients with hepatic disease which is associated with coagulopathy leading to a clinically relevant bleeding risk (see Contraindications).
No dose adjustment is necessary in patients with other hepatic diseases (see Pharmacology: Pharmacokinetics under Actions).
Limited clinical data in patients with moderate hepatic impairment (Child Pugh B) indicate a significant increase in the pharmacological activity. No clinical data are available for patients with severe hepatic impairment (Child Pugh C) (see Pharmacology: Pharmacokinetics under Actions and Contraindications).
SPAF: Patients with renal impairment: No dose adjustment is required if Rivaroxaban (Xarelto) is administered in patients with mild (creatinine clearance (CrC): ≤80-50 mL/min) renal impairment (see Pharmacology: Pharmacokinetics under Actions).
For patients with moderate (CrC: <50-30 mL/min) renal impairment the recommended dose is 15 mg once daily (see Pharmacology: Pharmacokinetics under Actions).
Limited clinical data for patients with severe renal impairment (CrC: <30-15 mL/min) indicate that rivaroxaban plasma levels are significantly increased in this patient population. Therefore, Rivaroxaban (Xarelto) 15 mg must be used with caution in these patients (see Pharmacology: Pharmacokinetics under Actions and Precautions).
Use of Rivaroxaban (Xarelto) is not recommended in patients with CrC <15 mL/min (see Pharmacology: Pharmacokinetics under Actions and Precautions).
SPAF: Converting from Vitamin K Antagonists (VKA) to Rivaroxaban (Xarelto): VKA treatment should be stopped and Rivaroxaban (Xarelto) therapy should be initiated when the INR is ≤ 3.0.
When converting patients from VKAs to Rivaroxaban (Xarelto), INR values will be falsely elevated after the intake of Rivaroxaban (Xarelto). The INR is not valid to measure the anticoagulant activity of Rivaroxaban (Xarelto), and therefore should not be used (see Interactions).
SPAF: Converting from Rivaroxaban (Xarelto) to Vitamin K antagonists (VKA): There is a potential for inadequate anticoagulation during the transition from Rivaroxaban (Xarelto) to VKA. Continuous adequate anticoagulation should be ensured during any transition to an alternate anticoagulant. It should be noted that Rivaroxaban (Xarelto) can contribute to an elevated INR.
In patients converting from Rivaroxaban (Xarelto) to VKA, VKA should be given concurrently until the INR is ≥2.0. For the first two days of the conversion period, standard VKA dosing should be used followed by VKA dosing guided by INR testing. While patients are on both Rivaroxaban (Xarelto) and VKA, the INR should not be tested earlier than 24 hours (after the previous dose but prior to the next dose of Rivaroxaban (Xarelto). Once Rivaroxaban (Xarelto) is discontinued INR testing may be done reliably 24 hours after the last dose (see Interactions).
SPAF: Converting from parenteral anti-coagulants to Rivaroxaban (Xarelto): For patients currently receiving a parenteral anticoagulant, start Rivaroxaban (Xarelto) 0 to 2 hours before the time of the next scheduled administration of the parenteral drug (e.g. LMWH) or at the time of discontinuation of a continuously administered parenteral drug (e.g. intravenous unfractionated heparin).
SPAF: Converting from Rivaroxaban (Xarelto) to parenteral anticoagulants: Discontinue Rivaroxaban (Xarelto) and give the first dose of parenteral anticoagulant at the time that the next Rivaroxaban (Xarelto) dose would be taken.
SPAF: Cardioversion: Rivaroxaban (Xarelto) can be initiated or continued in patients who may require cardioversion.
For transesophageal echocardiogram (TEE) guided cardioversion in patients not previously treated with anticoagulants, Rivaroxaban (Xarelto) treatment should be started at least 4 hours before cardioversion to ensure adequate anticoagulation (see Pharmacology: Pharmacodynamics and Pharmacokinetics under Actions).
SPAF: Patients who undergo PCI (percutaneous coronary intervention) with stent placement: Patients with non-valvular atrial fibrillation who undergo PCI with stent placement should receive reduced dose of 15 mg Rivaroxaban (Xarelto) once daily (or 10 mg Rivaroxaban (Xarelto) once daily for patients with moderate renal impairment [CrCl: <50-30 mL/min]) in addition to a P2Y12 inhibitor. This treatment regimen is recommended for a maximum of 12 months after PCI with stent placement (see Pharmacology: Pharmacodynamics under Actions and Precautions). After completion of the antiplatelet therapy, rivaroxaban dosage should be increased to the standard dose for patients with non-valvular atrial fibrillation.
SPAF: Children and adolescents (from birth to 16 or 18 years depending on local law): Safety and efficacy have not been established in children and adolescents below 18 years.
SPAF: Geriatric patients: No dose adjustment is required based on age (see Pharmacology: Pharmacokinetics under Actions).
SPAF: Gender: No dose adjustment is required based on gender (see Pharmacology: Pharmacokinetics under Actions).
SPAF: Body weight: No dose adjustment is required based on body weight (see Pharmacology: Pharmacokinetics under Actions).
SPAF: Ethnic differences: No dose adjustment is required based on ethnic differences (see Pharmacology: Pharmacokinetics under Actions).
Overdosage
Rare cases of overdose up to 600 mg have been reported without bleeding complications or other adverse reactions. Due to limited absorption a ceiling effect with no further increase in average plasma exposure is expected at supratherapeutic doses of 50 mg or above.
A specific antidote antagonizing the pharmacodynamic effect of rivaroxaban is not available. The use of activated charcoal to reduce absorption in case of Rivaroxaban (Xarelto) overdose may be considered. Due to the high plasma protein binding rivaroxaban is not expected to be dialyzable.
Management of Bleeding: Should a bleeding complication arise in a patient receiving rivaroxaban, the next administration should be delayed or treatment should be discontinued as appropriate. Rivaroxaban has a half-life of approximately 5 to 13 hours. Management should be individualized according to the severity and location of the hemorrhage. Appropriate symptomatic treatment could be used as needed, such as mechanical compression (e.g., for severe epistaxis), surgical hemostasis with bleeding control procedures, fluid replacement and hemodynamic support, blood products (packed red cells or fresh frozen plasma, depending on associated anemia or coagulopathy) or platelets.
If bleeding cannot be controlled by the previous measures, administration of a specific procoagulant reversal agent should be considered, such as prothrombin complex concentrate (PCC), activated prothrombin complex concentrate (APCC), or recombinant factor VIIa (r-FVIIa). However, there is currently very limited clinical experience with the use of these products in individuals receiving Rivaroxaban (Xarelto) (see Pharmacology: Pharmacodynamics under Actions).
Protamine sulfate and vitamin K are not expected to affect the anticoagulant activity of rivaroxaban.
There is limited experience with tranexamic acid and no experience with aminocaproic acid and aprotinin in individuals receiving Rivaroxaban (Xarelto). There is neither scientific rationale for benefit nor experience with the systemic hemostatic desmopressin in individuals receiving Rivaroxaban (Xarelto).
Contraindications
Rivaroxaban (Xarelto) is contraindicated in patients with hypersensitivity to rivaroxaban or any excipient of the tablet (see Excipients/Inactive Ingredients under Description).
Rivaroxaban (Xarelto) is contraindicated in patients with clinically significant active bleeding (e.g., intracranial bleeding, gastrointestinal bleeding).
Rivaroxaban (Xarelto) is contraindicated in patients with hepatic disease which is associated with coagulopathy leading to a clinically relevant bleeding risk (see Pharmacology: Pharmacokinetics under Actions).
Safety and efficacy of Rivaroxaban (Xarelto) have not been established in pregnant women. Animal data show that rivaroxaban crosses the placental barrier. Therefore use of Rivaroxaban (Xarelto) is contraindicated throughout pregnancy (see Pharmacology: Toxicology: Preclinical Safety Data under Actions and Use in Pregnancy & Lactation).
Safety and efficacy of Rivaroxaban (Xarelto) have not been established in nursing mothers. Animal data indicate that rivaroxaban is secreted into breast milk. Therefore Rivaroxaban (Xarelto) may only be administered after breastfeeding is discontinued (see Pharmacology: Toxicology: Preclinical Safety Data under Actions and Use in Pregnancy & Lactation).
Special Precautions
Patients with prosthetic heart valves: Rivaroxaban (Xarelto) is not recommended for thromboprophylaxis in patients having recently undergone transcatheter aortic valve replacement (TAVR) based on data from a randomized controlled clinical study comparing a Xarelto-regimen to an antiplatelet regimen (see Pharmacology: Pharmacodynamics under Actions).
The safety and efficacy of Rivaroxaban (Xarelto) have not been studied in patients with other prosthetic heart valves or other valve procedures; therefore, there are no data to support that Xarelto provides adequate anticoagulation in those patient populations.
Concomitant medication: Rivaroxaban (Xarelto) is not recommended in patients receiving concomitant systemic treatment with azole-antimycotics (e.g. ketoconazole) or HIV protease inhibitors (e.g. ritonavir). These drugs are strong inhibitors of both CYP 3A4 and P-gp. Therefore, these drugs may increase rivaroxaban plasma concentrations to a clinically relevant degree (2.6-fold on average) which may lead to an increased bleeding risk (see Interactions).
The Azole-antimycotic fluconazole, a moderate CYP 3A4 inhibitor, has however less effect on rivaroxaban exposure and can be co-administered (see Interactions).
Renal Impairment: Rivaroxaban (Xarelto) is to be used with caution in patients with moderate renal impairment (CrC: <50-30 mL/min) receiving co-medications leading to increased rivaroxaban plasma concentrations (see Interactions).
In patients with severe renal impairment (CrC: <30 mL/min), rivaroxaban plasma levels may be significantly elevated (1.6-fold on average) which may lead to an increased bleeding risk. Due to the underlying disease these patients are at an increased risk of both bleeding and thrombosis.
Due to limited clinical data Rivaroxaban (Xarelto) should be used with caution in patients with CrCl <30-15 mL/min (see Pharmacology: Pharmacokinetics under Actions).
No clinical data are available for patients with severe renal impairment (CrCl <15 mL/min). Therefore, use of Rivaroxaban (Xarelto) is not recommended in these patients (see Pharmacology and Pharmacokinetics under Actions and Dosage & Administration).
Patients with severe renal impairment or increased bleeding risk and patients receiving concomitant systemic treatment with azole-antimycotics or HIV protease inhibitors are to be carefully monitored for signs of bleeding complications after initiation of treatment (see Interactions).
Effects on ability to drive or use machines: Syncope and dizziness have been reported and may affect the ability to drive and use machines (see Adverse Reactions). Patients experiencing these adverse reactions should not drive or use machines.
2.5 mg: This may be done by regular physical examination of the patients, close observation of the surgical wound drainage and periodic measurements of hemoglobin.
10 mg: In patients receiving Rivaroxaban (Xarelto) for the prevention of venous thromboembolism (VTE) after elective hip or knee replacement surgery, this may be done by regular physical examination of the patients, close observation of the surgical wound drainage and periodic measurements of hemoglobin.
In ACS patients, efficacy and safety of Rivaroxaban (Xarelto) 2.5 mg twice daily have been investigated in combination with the antiplatelet agents ASA alone or ASA plus clopidogrel/ticlopidine. Treatment in combination with other antiplatelet agents, e.g. prasugrel or ticagrelor, has not been studied and therefore, is not recommended.
Neuraxial (epidural/spinal) anesthesia: When neuraxial (epidural/spinal) anesthesia or spinal puncture is performed, patients treated with antithrombotics for prevention of thromboembolic complications are at risk for development of an epidural or spinal hematoma which may result in long-term paralysis.
The risk of these events is even further increased by use of indwelling epidural catheters or the concomitant use of drugs affecting hemostasis. The risk may also be increased by traumatic or repeated epidural or spinal puncture.
Patients should be frequently monitored for signs and symptoms of neurological impairment (e.g., numbness or weakness of the legs, bowel or bladder dysfunction). If neurological deficits are noted, urgent diagnosis and treatment is necessary.
The physician should consider the potential benefit versus the risk before neuraxial intervention in patients anticoagulated or to be anticoagulated for thromboprophylaxis.
There is no clinical experience with the use of 2.5 mg twice daily with ASA alone or with ASA plus clopidogrel or ticlopidine in these situations.
To reduce the potential risk of bleeding associated with the concurrent use of rivaroxaban and neuraxial (epidural/spinal) anesthesia or spinal puncture, consider the pharmacokinetic profile of rivaroxaban. Placement or removal of an epidural catheter or lumbar puncture is best performed when the anticoagulant effect of rivaroxaban is estimated to be low. However, the exact timing to reach a sufficiently low anticoagulant effect in each patient is not known.
The concomitant use of platelet aggregation inhibitors should be considered and such medication discontinued as appropriate.
10 mg: An epidural catheter should not be withdrawn earlier than 18 hours after the last administration of Rivaroxaban (Xarelto).
15 and 20 mg: There is no clinical experience with the use of 15 mg and 20 mg rivaroxaban in these situations.
For the removal of an epidural catheter and based on the general PK characteristics at least 2x half-life should elapse, i.e. at least 18 hour in young patients and 26 hours in elderly patients, after the last administration of Rivaroxaban (Xarelto) (see Pharmacology: Pharmacokinetics under Actions).
Rivaroxaban (Xarelto) should be administered at earliest 6 hours after the removal of the catheter.
If traumatic puncture occurs, the administration of Rivaroxaban (Xarelto) should be delayed for 24 hours.
Patients with high risk triple positive antiphospholipid syndrome: Rivaroxaban (Xarelto) is not recommended for patients with a history of thrombosis who are diagnosed with antiphospholipid syndrome and are persistently triple positive (for lupus anticoagulant, anticardiolipin antibodies, and anti-beta 2-glycoprotein I antibodies) as treatment with rivaroxaban is associated with an increased rate of recurrent thrombotic events compared with vitamin K antagonists (VKA) (see Pharmacology: Pharmacodynamics under Actions).
Bleeding risk: Rivaroxaban (Xarelto) like other antithrombotics should be used with caution in patients with an increased bleeding risk such as: congenital or acquired bleeding disorders; uncontrolled severe arterial hypertension; active ulcerative gastrointestinal disease; recent gastrointestinal ulcerations; vascular retinopathy; recent intracranial or intracerebral hemorrhage; intraspinal or intracerebral vascular abnormalities; recent brain, spinal or ophthalmological surgery; bronchiectasis or history of pulmonary bleeding.
Care should be taken if patients are treated concomitantly with drugs affecting hemostasis such as non-steroidal anti-inflammatory drugs (NSAIDs), platelet aggregation inhibitors, or other antithrombotics, or selective serotonin reuptake inhibitors (SSRI), and serotonin norepinephrine reuptake inhibitors (SNRIs), (see Interactions).
Patients on treatment with Rivaroxaban (Xarelto) and ASA or with Rivaroxaban (Xarelto) and ASA plus clopidogrel/ticlopidine should only receive chronic concomitant treatment with NSAIDS if the benefit outweighs the bleeding risk.
For patients at risk of ulcerative gastrointestinal disease an appropriate prophylactic treatment may be considered (see Interactions).
Any unexplained fall in hemoglobin or blood pressure should lead to a search for a bleeding site.
Surgery and interventions: If an invasive procedure or surgical intervention is required, Rivaroxaban (Xarelto) should be stopped at least 24 hours before the intervention, if possible and based on clinical judgment of the physician.
If the procedure cannot be delayed the increased risk of bleeding should be assessed against the urgency of the intervention.
If a patient concomitantly receiving platelet aggregation inhibitors is to undergo elective surgery and anti-platelet effect is not desired, platelet aggregation inhibitors should be discontinued as directed by the manufacturer's prescribing information.
Rivaroxaban (Xarelto) should be restarted as soon as possible after the invasive procedure or surgical intervention provided the clinical situation allows and adequate hemostasis has been established (see Pharmacology: Pharmacokinetics under Actions).
Women of childbearing potential: Rivaroxaban (Xarelto) should be used in women of childbearing potential only with effective contraception.
QTc prolongation: No QTc prolonging effect was observed with Rivaroxaban (Xarelto) (see Pharmacology: Pharmacokinetics under Actions).
Information about excipients: Since this medicinal product contains lactose, patients with rare hereditary problems of lactose or galactose intolerance (e.g., the Lapp lactase deficiency or glucose-galactose malabsorption) should not take Rivaroxaban (Xarelto) (see Description).
2.5 mg: ACS, CAD or PAD: Patients with prior stroke and/or TIA: ACS: Rivaroxaban (Xarelto) 2.5 mg twice daily should be avoided for the treatment of ACS in patients with a prior stroke or TIA. Few ACS patients with a prior stroke or TIA have been studied, but the limited efficacy data available indicates that these patients may not benefit from treatment.
CAD or PAD: Patients with haemorrhagic or lacunar stroke: CAD or PAD patients with previous haemorrhagic or lacunar stroke were not studied. Treatment with Rivaroxaban (Xarelto) 2.5 mg twice daily in combination with ASA should be avoided in these patients.
Patients with ischemic, non-lacunar stroke: CAD or PAD patients who have experienced an ischemic, non-lacunar stroke within the previous month were not studied. Treatment with Rivaroxaban (Xarelto) 2.5 mg twice daily in combination with ASA should be avoided in the first month after stroke (see Pharmacology: Pharmacokinetics under Actions).
10, 15 and 20 mg: Treatment and prevention of recurrent DVT and PE: Haemodynamically unstable PE patients or patients who require thrombolysis or pulmonary embolectomy: Rivaroxaban (Xarelto) is not recommended as an alternative to unfractionated heparin in patients with pulmonary embolism who are haemodynamically unstable or may receive thrombolysis or pulmonary embolectomy since the safety and efficacy of Rivaroxaban (Xarelto) have not been established in these clinical situations.
10 mg: Hip fracture surgery: Rivaroxaban (Xarelto) has not been studied in interventional clinical trials in patients undergoing hip fracture surgery. Limited clinical data from a non-interventional study are available for patients undergoing fracture related surgery of the lower limbs such as hip fracture surgery (see Pharmacology: Pharmacodynamics under Actions).
15 and 20 mg: SPAF: Patients who undergo PCI with stent placement: Clinical data are available from an interventional study with the primary objective to assess safety in patients with non-valvular atrial fibrillation who undergo PCI with stent placement. Data on efficacy in this population are limited (see Pharmacology: Pharmacodynamics under Actions and Dosage & Administration).
Use In Pregnancy & Lactation
Women of childbearing potential / Contraception: Rivaroxaban (Xarelto) should be used in women of childbearing potential only with effective contraception.
Pregnancy: Safety and efficacy of Rivaroxaban (Xarelto) have not been established in pregnant women.
In rats and rabbits rivaroxaban showed pronounced maternal toxicity with placental changes related to its pharmacological mode of action (e.g., hemorrhagic complications) leading to reproductive toxicity (see Pharmacology: Toxicology: Preclinical safety data under Actions). No primary teratogenic potential was identified. Due to the intrinsic risk of bleeding and the evidence that rivaroxaban passes the placenta, Rivaroxaban (Xarelto) is contraindicated in pregnancy (see Pharmacology: Toxicology: Preclinical safety data under Actions and Contraindications).
Lactation: Safety and efficacy of Rivaroxaban (Xarelto) have not been established in nursing mothers. In rats rivaroxaban is secreted into breast milk.
Therefore Rivaroxaban (Xarelto) may only be administered after breastfeeding is discontinued (see Pharmacology: Toxicology: Preclinical safety data under Actions and Contraindications).
Adverse Reactions
Summary of the safety profile: 2.5 mg: The safety of rivaroxaban has been evaluated in thirteen phase III studies including 53,103 patients exposed to rivaroxaban, as listed in the following table: See Table 15.

Click on icon to see table/diagram/image

In patients treated with 10 mg Rivaroxaban (Xarelto) undergoing hip or knee replacement surgery and in hospitalized medically ill patients, bleeding events occurred in approximately 6.8% and 12.6% of patients, respectively, and anemia occurred in approximately 5.9% and 2.1% of patients, respectively. In patients treated with either 15 mg twice daily Rivaroxaban (Xarelto) followed by 20 mg once daily for treatment of DVT or PE, or with 20 mg once daily for prevention of recurrent DVT and PE, bleeding events occurred in approximately 23% of patients and anemia occurred in approximately 1.6% of patients. In patients treated for prevention of stroke and systemic embolism, bleeding of any type or severity was reported with an event rate of 28 per 100 patient years, and anemia with an event rate of 2.5 per 100 patient years. In patients treated for prevention of cardiovascular death and myocardial infarction after an acute coronary syndrome (ACS), bleeding of any type or severity was reported with an event rate of 22 per 100 patient years. Anemia was reported with an event rate of 1.4 per 100 patient years. In patients treated for prevention of stroke, myocardial infarction and cardiovascular death, and prevention of acute limb ischemia and mortality in patients with CAD or PAD, bleeding of any type or severity was reported with an event rate of 6.7 per 100 patient years. Anemia was reported with an event rate of 0.15 per 100 patient years*.
*A pre-specified selective approach to adverse event collection was applied.
Due to the pharmacological mode of action, Rivaroxaban (Xarelto) may be associated with an increased risk of occult or overt bleeding from any tissue and organ which may result in post hemorrhagic anemia. The risk of bleedings may be increased in certain patient groups e.g. patients with uncontrolled severe arterial hypertension and/or on concomitant medication affecting hemostasis (see Precautions).
The signs, symptoms, and severity (including fatal outcome) will vary according to the location and degree or extent of the bleeding and/or anemia (see Overdosage).
Hemorrhagic complications may present as weakness, paleness, dizziness, headache or unexplained swelling, dyspnea, and unexplained shock. In some cases as a consequence of anemia, symptoms of cardiac ischemia like chest pain or angina pectoris have been observed.
Known complications secondary to severe bleeding such as compartment syndrome and renal failure due to hypoperfusion have been reported for Rivaroxaban (Xarelto). Therefore, the possibility of a hemorrhage should be considered in evaluating the condition in any anticoagulated patient.
10, 15 and 20 mg: The safety of rivaroxaban has been evaluated in twelve phase III studies including 34,859 patients exposed to rivaroxaban, as listed in the following table: See Table 16.

Click on icon to see table/diagram/image

In total about 65% of patients exposed to at least one dose of rivaroxaban were reported with treatment emergent adverse events. About 21% of patients experienced adverse events considered related to treatment as assessed by investigators. In patients treated with 10 mg Rivaroxaban (Xarelto) undergoing hip or knee replacement surgery and in hospitalized medically ill patients, bleeding events occurred in approximately 6.8% and 12.6% of patients, respectively, and anemia occurred in approximately 5.9% and 2.1% of patients, respectively. In patients treated with either 15 mg twice daily Rivaroxaban (Xarelto) followed by 20 mg once daily for treatment of DVT or PE, or with 20 mg once daily for prevention of recurrent DVT and PE, bleeding events occurred in approximately 23% of patients and anemia occurred in approximately 1.6% of patients. In patients treated for prevention of stroke and systemic embolism, bleeding of any type or severity was reported with an event rate of 28 per 100 patient years, and anemia with an event rate of 2.5 per 100 patient years. In patients treated for prevention of cardiovascular death and myocardial infarction after an acute coronary syndrome (ACS), bleeding of any type or severity was reported with an event rate of 22 per 100 patient years. Anemia was reported with an event rate of 1.4 per 100 patient years.
Due to the pharmacological mode of action, Rivaroxaban (Xarelto) may be associated with an increased risk of occult or overt bleeding from any tissue and organ which may result in post hemorrhagic anemia. The risk of bleedings may be increased in certain patient groups e.g. patients with uncontrolled severe arterial hypertension and/or on concomitant medication affecting hemostasis (see Precautions).
The signs, symptoms, and severity (including fatal outcome) will vary according to the location and degree or extent of the bleeding and/or anemia (see Overdosage).
Hemorrhagic complications may present as weakness, paleness, dizziness, headache or unexplained swelling, dyspnea, and unexplained shock. In some cases as a consequence of anemia, symptoms of cardiac ischemia like chest pain or angina pectoris have been observed.
Known complications secondary to severe bleeding such as compartment syndrome and renal failure due to hypoperfusion have been reported for Rivaroxaban (Xarelto). Therefore, the possibility of a hemorrhage should be considered in evaluating the condition in any anticoagulated patient.
Tabulated list of adverse reactions: The frequencies of ADRs reported with Rivaroxaban (Xarelto) are summarized in the table as follows. Within each frequency grouping, undesirable effects are presented in order of decreasing seriousness. Frequencies are defined as: very common (≥1/10), common (≥1/100 to <1/10), uncommon (≥1/1,000 to <1/100), rare (≥1/10,000 to <1/1,000).
2.5 mg: See Table 17.

Click on icon to see table/diagram/image

10, 15 and 20 mg: See Table 18.

Click on icon to see table/diagram/image

Post marketing observations: The following adverse reactions have been reported post-marketing in temporal association with the use of Rivaroxaban (Xarelto). The frequency of these adverse reactions reported from post-marketing experience cannot be estimated.
Immune system disorders: Angioedema and allergic oedema (In the pooled phase III trials, these events were uncommon (≥1/1,000 to <1/100)).
Hepatobiliary disorders: Cholestasis, Hepatitis (incl. hepatocellular injury) (In the pooled phase III trials, these events were rare (≥1/10,000 to <1/1,000)).
Blood and lymphatic system disorders: Thrombocytopenia (In the pooled phase III trials, these events were uncommon (≥1/1,000 to <1/100)).
Drug Interactions
Pharmacokinetic interactions: Rivaroxaban is cleared mainly via cytochrome P450-mediated (CYP 3A4, CYP 2J2) hepatic metabolism and renal excretion of the unchanged drug, involving the P-glycoprotein (P-gp) / breast cancer resistance protein (Bcrp) transporter systems.
CYP Inhibition: Rivaroxaban does not inhibit CYP 3A4 or any other major CYP isoforms.
CYP Induction: Rivaroxaban does not induce CYP 3A4 or any other major CYP isoforms.
Effects on rivaroxaban: The concomitant use of Rivaroxaban (Xarelto) with strong CYP 3A4 and P-gp inhibitors, may lead to both reduced hepatic and renal clearance and thus significantly increased systemic exposure.
Co-administration of Rivaroxaban (Xarelto) with the azole-antimycotic ketoconazole (400 mg once daily) a strong CYP 3A4 and P-gp inhibitor, led to a 2.6-fold increase in mean rivaroxaban steady state AUC and a 1.7-fold increase in mean rivaroxaban Cmax, with significant increases in its pharmacodynamic effects.
Co-administration of Rivaroxaban (Xarelto) with the HIV protease inhibitor ritonavir (600 mg twice daily), a strong CYP 3A4 and P-gp inhibitor, led to a 2.5-fold increase in mean rivaroxaban AUC and a 1.6-fold increase in mean rivaroxaban Cmax, with significant increases in its pharmacodynamic effects.
Therefore Rivaroxaban (Xarelto) is not recommended in patients receiving concomitant systemic treatment with azole-antimycotics or HIV-protease inhibitors (see Precautions).
Other active substances strongly inhibiting only one of the rivaroxaban elimination pathways, either CYP 3A4 or P-gp, are expected to increase rivaroxaban plasma concentrations to a lesser extent.
Clarithromycin (500 mg twice daily), considered as strong CYP 3A4 inhibitor and moderate P-gp inhibitor, led to a 1.5-fold increase in mean rivaroxaban AUC and a 1.4-fold increase in Cmax. This increase, which is close to the magnitude of the normal variability of AUC and Cmax, is considered as clinically not relevant.
Erythromycin (500 mg three times daily), which inhibits CYP 3A4 and P-gp moderately, led to a 1.3-fold increase in mean rivaroxaban AUC and Cmax. This increase is within the magnitude of the normal variability of AUC and Cmax and is considered as clinically not relevant.
In subjects with mild renal impairment, erythromycin (500 mg three times a day) led to a 1.8-fold increase in mean rivaroxaban AUC and 1.6-fold increase in Cmax when compared to subjects with normal renal function without co-medication. In subjects with moderate renal impairment, erythromycin led to a 2.0-fold increase in mean rivaroxaban AUC and 1.6-fold increase in Cmax when compared to subjects with normal renal function without co-medication (see Precautions).
Fluconazole (400 mg once daily), considered as moderate CYP 3A4 inhibitor, led to a 1.4-fold increase in mean rivaroxaban AUC and a 1.3-fold increase in mean Cmax. This increase is within the magnitude of the normal variability of AUC and Cmax and is considered as clinically not relevant (see Precautions).
Co-administration of Rivaroxaban (Xarelto) with the strong CYP 3A4 and P-gp inducer rifampicin led to an approximate 50% decrease in mean rivaroxaban AUC, with parallel decreases in its pharmacodynamic effects (see Pharmacology: Pharmacokinetics under Actions).
The concomitant use of Rivaroxaban (Xarelto) with other strong CYP 3A4 inducers (e.g., phenytoin, carbamazepine, phenobarbitone or St. John's Wort) may also lead to a decreased rivaroxaban plasma concentration.
2.5 mg: Strong CYP 3A4 inducers must be used with caution in ACS, or CAD or PAD patients treated with 2.5 mg Rivaroxaban (Xarelto) twice daily.
10 mg: The decrease in rivaroxaban plasma concentrations is considered as clinically not relevant for patients treated with 10 mg Rivaroxaban (Xarelto) once daily for prevention of VTE after major orthopedic surgery of the lower limbs.
15 mg and 20 mg: Strong CYP 3A4 inducers should be co-administered with caution (see Pharmacology: Pharmacokinetics under Actions).
Pharmacodynamic interactions: After combined administration of enoxaparin (40 mg single dose) with Rivaroxaban (Xarelto) (10 mg single dose), an additive effect on anti-factor Xa activity was observed without any additional effects on clotting tests (PT, aPTT). Enoxaparin did not affect the pharmacokinetics of rivaroxaban (see Precautions).
Clopidogrel (300 mg loading dose followed by 75 mg maintenance dose) did not show a pharmacokinetic interaction (with Rivaroxaban (Xarelto) 15 mg) but a relevant increase in bleeding times was observed in a subset of patients which was not correlated to platelet aggregation, P-selectin or GPIIb/IIIa receptor levels (see Precautions).
No clinically relevant prolongation of bleeding time was observed after concomitant administration of Rivaroxaban (Xarelto) (15 mg) and 500 mg naproxen. Nevertheless there may be individuals with more pronounced pharmacodynamic response (see Precautions).
Converting patients from warfarin (INR 2.0 to 3.0) to Rivaroxaban (Xarelto) (20 mg) or from Rivaroxaban (Xarelto) (20 mg) to warfarin (INR 2.0 to 3.0) increased prothrombin time/INR (Neoplastin) more than additively (individual INR values up to 12 may be observed), whereas effects on aPTT, inhibition of factor Xa activity and endogenous thrombin potential were additive.
If it is desired to test the pharmacodynamic effects of Rivaroxaban (Xarelto) during the conversion period, anti-factor Xa activity, PiCT, and HepTest can be used as these tests were not affected by warfarin. From day 4 after stopping warfarin onwards, all tests (including PT, aPTT, inhibition of factor Xa activity and ETP) reflected only the effect of Rivaroxaban (Xarelto) (see Dosage & Administration).
If it is desired to test the pharmacodynamic effects of warfarin during the conversion period, INR measurement can be used at the Ctrough of rivaroxaban (24 hours after the previous intake of rivaroxaban) as this test is minimally affected by rivaroxaban at this time point.
No pharmacokinetic interaction was observed between warfarin and Rivaroxaban (Xarelto).
As with other anticoagulants the possibility may exist that patients are at increased risk of bleeding in case of concomitant use with SSRIs or SNRIs due to their reported effect on platelets. When concomitantly used in the rivaroxaban clinical program, numerically higher rates of major or non-major clinically relevant bleeding were observed in all treatment groups.
Food and dairy products: 2.5 and 10 mg: 2.5 mg and 10 mg Rivaroxaban (Xarelto) can be taken with or without food (see Pharmacology: Pharmacokinetics under Actions).
15 and 20 mg: Rivaroxaban (Xarelto) 15 mg tablets and Rivaroxaban (Xarelto) 20 mg tablets should be taken with food (see Pharmacology: Pharmacokinetics under Actions).
Interactions shown not to exist: There were no mutual pharmacokinetic interactions between rivaroxaban and midazolam (substrate of CYP 3A4), digoxin (substrate of P-glycoprotein) or atorvastatin (substrate of CYP 3A4 and P-gp).
Co-administration of the proton pump inhibitor omeprazole, the H2 receptor antagonist ranitidine, the antacid aluminum hydroxide/magnesium hydroxide, naproxen, clopidogrel or enoxaparin did not affect rivaroxaban bioavailability and pharmacokinetics.
No clinically significant pharmacokinetic or pharmacodynamic interactions were observed when Rivaroxaban (Xarelto) was co-administered with 500 mg acetylsalicylic acid.
Interactions with laboratory parameters: Clotting parameter tests (PT, aPTT, HepTest) are affected as expected by the mode of action of Rivaroxaban (Xarelto) (see Pharmacology: Pharmacodynamics under Actions).
Caution For Usage
Instructions for use / handling: None.
Incompatibilities: None Known.
Storage
Store at temperatures not exceeding 30°C.
2.5 and 10 mg:
For tablets, no storage restrictions (temperature, humidity, light) apply.
MIMS Class
Anticoagulants, Antiplatelets & Fibrinolytics (Thrombolytics)
ATC Classification
B01AF01 - rivaroxaban ; Belongs to the class of direct factor Xa inhibitors. Used in the treatment of thrombosis.
Presentation/Packing
Form
Xarelto FC tab 10 mg
Packing/Price
10's
Form
Xarelto FC tab 15 mg
Packing/Price
10's
Form
Xarelto FC tab 2.5 mg
Packing/Price
30's
Form
Xarelto FC tab 20 mg
Packing/Price
10's
Register or sign in to continue
Asia's one-stop resource for medical news, clinical reference and education
Already a member? Sign in
Register or sign in to continue
Asia's one-stop resource for medical news, clinical reference and education
Already a member? Sign in