scholarly journals Pharmacokinetic and Pharmacodynamic (PKPD) Modeling to Determine the Andexanet Alfa Dose to Reverse the Anticoagulant Activity of Direct Fxa Inhibitors

Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 1135-1135
Author(s):  
Leeds Janet ◽  
Genmin Lu ◽  
John T. Curnutte ◽  
Pamela B. Conley ◽  
Jaap W. Mandema

Abstract Introduction Andexanet alfa (AnXa) is a specific antidote under development for direct and indirect factor Xa (fXa) inhibitors. AnXa is a recombinant, engineered version of human fXa that is catalytically inactive but retains high affinity for direct fXa inhibitors. AnXa binds fXa inhibitors, sequestering them in plasma, thereby reducing the free inhibitor concentration, reversing fXa inhibition, and normalizing thrombin generation. The decreased unbound concentrations of the direct fXa inhibitors (apixaban, rivaroxaban and edoxaban) cause a portion of the extravascular fXa inhibitors to move into the vasculature. Reversal of fXa inhibitor-induced anticoagulation requires a molar excess of AnXa relative to the total amount of fXa inhibitor in the blood - i.e., the initial fXa inhibitor plasma concentration plus the redistributed amount. We report a PKPD model that accounts for this extravascular-intravascular redistribution and allows determination of the appropriate AnXa dose to reverse the anti-fXa activity for each approved direct fXa inhibitor. Methods A model for rivaroxaban was constructed first by jointly analyzing AnXa concentrations, total and unbound rivaroxaban concentrations, and anti-fXa activity for 5 different doses of AnXa (each administered at the peak plasma level of 20 mg QD rivaroxaban at steady state). Model parameters were estimated by maximum likelihood using nonlinear regression (NONMEM, v. 7.2.0). The model was used to simulate the potential level of reversal of anti-fXa activity after 2.5-40 mg/day rivaroxaban (QD and BID) for different bolus doses of AnXa and to predict maintenance of reversal by various follow-on infusion rates. A similar model was developed to characterize the interaction between AnXa and apixaban. Learnings from the rivaroxaban and apixaban models were used to develop a universal model to predict the level of reversal of anticoagulation for 60 mg edoxaban after a particular dose of AnXa. The model accurately predicted the degree of reversal of edoxaban anticoagulation. The final model was used to construct a nomogram of AnXa doses that could be used for each fXa inhibitor at each approved dose based on timing of the last dose of the inhibitor. Results The final PKPD model was a three-compartment PK model for the fXa inhibitors, including one central and two tissue compartments. The two tissue compartments included one that rapidly equilibrated with the central compartment and a second that equilibrated slowly. The combined volume of the central and rapidly equilibrating compartments was similar to the volume of central compartment of the fXa inhibitor PK models in the absence of AnXa. The AnXa PK was best described by a two-compartment PK model with an additional saturable binding component. The AnXa-fXa inhibitor complex moved to a "sequestration" compartment. Upon release from AnXa, the fXa inhibitor was able to return to the central compartment. The fXa inhibitor did not clear with AnXa. The binding between AnXa and free fXa inhibitor was characterized by a reversible binding equilibrium. There was a direct PKPD linear relationship between free fXa inhibitor concentrations and anti-fXa activity. Simulations were used to estimate the best dosing regimen to reverse anticoagulation for each direct fXa inhibitor. A bolus dose of 800 mg of AnXa followed by an 8 mg/min infusion was sufficient to fully reverse the peak level of anti-fXa activity after the 20 mg QD dose of rivaroxaban. This represents a >90% decrease in anti-fXa activity compared to pre-AnXa administration. Sustained reversal of peak anti-fXa levels for the 5 mg BID dose of apixaban required a 400 mg bolus AnXa dose with a follow-on infusion of 4 mg/min. The peak anticoagulant activity of the 60 mg dose of edoxaban could be reversed by an 800 mg bolus dose of AnXa followed by an 8 mg/min infusion. Conclusions A PKPD model was constructed that accurately predicted the AnXa dose necessary to reverse coagulation inhibition of each direct fXa inhibitor. The model incorporated the PK of AnXa, the anti-fXa activity, unbound and total levels of the fXa inhibitor, and the redistribution of each fXa inhibitor between the extravascular and intravascular compartments. Simulations were used to predict the AnXa dose necessary to reverse each approved dose of each direct fXa inhibitor and for different times after the last dose taken of the fXa inhibitor. Disclosures Janet: Portola Pharmaceuticals, Inc.: Employment. Lu:Portola Pharmaceuticals, Inc.: Employment. Curnutte:3-V Biosciences: Equity Ownership; Sea Lane Biotechnologies: Consultancy; Portola Pharmaceuticals, Inc.: Employment. Conley:Portola Pharmaceuticals, Inc.: Employment. Mandema:Portola Pharmaceuticals, Inc.: Consultancy.

1966 ◽  
Vol 44 (4) ◽  
pp. 635-640 ◽  
Author(s):  
F. C. Monkhouse ◽  
E. C. Abbott

The clearing factor lipase (CFL) response in rats to the intramuscular and intravenous injection of two synthetic sulfonated polysaccharides (RO1-8307, N-formyl chitosan polysulfuric acid, from Hoffmann – La Roche; and Heparinoid G-31150, a breakdown product of polygalacturonic acid, from Geigy (Canada) Ltd.) has been studied in comparison with that of heparin. The intravenous studies showed that both synthetic drugs released adequate levels of CFL but, on a weight basis, were less effective than heparin. Daily intramuscular doses of 20 mg of RO1-8307, with its very low anticoagulant activity, were well tolerated by rats for periods of 5 to 6 weeks. With this dose continuous release of CFL was maintained. After 2 weeks of continuous treatment, there was a measurable decrease in the peak plasma level of CFL following injection but the duration of response was not decreased.


2005 ◽  
Vol 102 (5) ◽  
pp. 985-994 ◽  
Author(s):  
Åke Norberg ◽  
Kirk I. Brauer ◽  
Donald S. Prough ◽  
Johan Gabrielsson ◽  
Robert G. Hahn ◽  
...  

Background Hemorrhage is commonly treated with intravenous infusion of crystalloids. However, the dynamics of fluid shifts between body fluid spaces are not completely known, causing contradictory recommendations regarding timing and volume of fluid infusions. The authors have developed a turnover model that characterizes these fluid shifts. Methods Conscious, chronically instrumented sheep (n = 12) were randomly assigned to three protocol groups: infusion of 25 ml/kg of 0.9% saline over 20 min (infusion only), hemorrhage of 300 ml (7.8 +/- 1.1 ml/kg) over 5 min (hemorrhage only), and hemorrhage of 300 ml over 5 min followed by infusion as noted above (hemorrhage plus infusion). A two-compartment volume turnover kinetic model containing seven model parameters was fitted to data obtained by repeated sampling of hemoglobin concentration and urinary excretion. Results The volume turnover model successfully predicted fluid shifts. Mean baseline volumes of the central and tissue compartments were 1799 +/- 1276 ml and 7653 +/- 5478 ml, respectively. Immediate fluid infusion failed to prevent hemorrhage-induced depression of cardiac output and diuresis. The model suggested that volume recruitment to the central compartment after hemorrhage was primarily achieved by mechanisms other than volume equilibration between the two model compartments. Conclusion Volume turnover kinetics is a promising tool for explaining fluid shifts between body compartments after perturbations such as hemorrhage and intravenous fluid infusions. The pronounced inhibition of renal output after hemorrhage prevailed regardless of fluid infusion and caused fluid retention, which expanded the tissue compartment.


Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Genmin Lu ◽  
Jayaprakash Kotha ◽  
Juan M Cardenas ◽  
Michael J Herr ◽  
Anjali Pandey ◽  
...  

New oral fXa inhibitors have been increasingly adopted for VTE or AF treatment in the outpatient setting instead of warfarin. Andexanet alfa (AnXa) is a modified, recombinant human fXa molecule developed as a specific antidote to reverse anticoagulant activity of fXa inhibitors during episodes of serious bleeding or before urgent surgery. PER977, a small molecule under development by Perosphere, Inc., is reported to reverse the effect of a broad range of anticoagulants (fXa and thrombin inhibitors, LMWH). In order to compare AnXa and PER977 mechanisms of action, we studied the in vitro activity of both agents in the presence or absence of fXa inhibitors rivaroxaban, apixaban, edoxaban or enoxaparin. In a buffer system containing purified human fXa with physiologic Ca 2+ (5 mM), AnXa dose-dependently reversed oral fXa inhibitor activity. Reversal activity was not observed for any fXa inhibitor with PER977 over a wide concentration range (≤2 mM). In human plasma, AnXa reversed both direct and ATIII-dependent fXa inhibitors, whereas no reversal effect by PER977 was detected. In the absence of a fXa inhibitor, PER977 potentiated fX activation by fIXa in a buffer system (purified human fX, fIXa and 5mM Ca 2+ ). Similar cofactor activity was observed with polylysine, indicating that PER977 may have properties similar to poly-cationic molecules. Procoagulant activity was observed in human plasma with PER977 at low concentrations (≤100 μM) as measured by clotting (aPTT) or thrombin generation, but inhibition in these assays was seen at higher concentrations ( ~1 mM). PER977 also potentiated human platelet activation as determined by P-selectin expression induced by 10 μM ADP. Platelet aggregation, whole blood hemolysis, or complement activation testing showed no effect with either AnXa or PER977. These data indicate that PER977 does not reverse the anticoagulant activity of a direct or indirect fXa inhibitor in vitro. Its observed in vivo effect on blood loss in animals may not be mediated by direct interaction of PER977 with the inhibitor, but may be in part attributed to an off-target effect, suggested by its potential procoagulant activity. In contrast, AnXa acts as a specific antidote to fXa inhibitors by sequestering them with a defined stoichiometry (1:1 molar ratio).


2013 ◽  
Vol 109 (04) ◽  
pp. 596-605 ◽  
Author(s):  
Stephan Formella ◽  
Erol Wiegert ◽  
Viktoria Moschetti ◽  
Torsten Slowinski ◽  
Hans-H. Neumayer ◽  
...  

SummaryDabigatran, a specific, reversible direct thrombin inhibitor, is used to prevent ischaemic and haemorrhagic strokes in patients with atrial fibrillation. As with every anticoagulant, there is a need to rapidly reverse its effects in emergency situations. In an open-label, single-centre phase I study with two fixed multiple dosing periods, we investigated the pharmacokinetics, pharmacodynamics and safety of dabigatran before, during and after 4 hour haemodialysis sessions with either 200 or 400 ml/min targeted blood flow in seven end-stage renal disease patients without atrial fibrillation. Dabigatran was administered over three days in a regimen designed to achieve peak plasma concentrations comparable to those observed in atrial fibrillation patients receiving 150 mg b.i.d. and to attain adequate distribution of dabigatran in the central and peripheral compartments. Plasma concentration-time profiles were similar in both periods on Day 3 (Cmax: 176 and 159 ng/ml). Four hours of haemodialysis removed 48.8% and 59.3% of total dabigatran from the central compartment with 200 and 400 ml/minute targeted blood flow, respectively. The anticoagulant activity of dabigatran was linearly related to its plasma levels. There was a minor redistribution of dabigatran (<16%) after the end of the haemodialysis session. In conclusion, a 4 hour haemodialysis session can rapidly eliminate a substantial amount of dabigatran from the central compartment with a concomitant marked reduction in its anticoagulant activity. There was a clinically negligible redistribution of dabigatran after haemodialysis. These results demonstrate that haemodialysis can be a suitable approach to eliminate dabigatran in emergency situations.


Neurosurgery ◽  
2017 ◽  
Vol 64 (CN_suppl_1) ◽  
pp. 267-267 ◽  
Author(s):  
Mark Crowther ◽  
Genmin Lu ◽  
Janet Leeds ◽  
Joyce Lin ◽  
Alex Gold ◽  
...  

Abstract INTRODUCTION Andexanet alfa (andexanet) is a modified, recombinant human factor Xa (FXa) that acts as a decoy to bind and sequester FXa inhibitors, thus reversing their anticoagulation effects. Here, we report the efficacy of andexanet in reversing the anticoagulant activity of betrixaban, a direct FXa inhibitor which has recently completed a large Phase 3 clinical trial in acute medically ill patients (APEX). METHODS In this Phase 2, randomized, double-blind study, healthy subjects were dosed with 80 mg qd po betrixaban to steady state (7 days). In Cohort 1, subjects (n = 6) received 800-mg andexanet bolus 3 hours after the last dose of betrixaban, or matching placebo (n = 3). In Cohort 2, subjects (n = 6) received 800-mg andexanet bolus 4 hours after the last betrixaban dose, followed immediately by a 2-hour andexanet infusion (8 mg/min), or matching placebo (n = 3). Study endpoints included assessments of safety and pharmacodynamic markers of anticoagulation reversal. RESULTS >Following dosing with betrixaban, andexanet rapidly (2 minutes after the bolus) decreased anti-FXa activity by ∼80% in both cohorts (P < 0.001 vs. placebo) and decreased unbound betrixaban plasma concentration by 73% and 83% in Cohorts 1 and 2, respectively (P < 0.001 vs. placebo). The effects were maintained during the 2-hour infusion of andexanet. Thrombin generation was restored in 11/12 (91.7%) subjects administered andexanet vs. 2/6 (33.3%) placebo subjects. Andexanet was well-tolerated; there were no thrombotic events or other serious/severe adverse events. CONCLUSION Andexanet was well-tolerated and rapidly reversed anticoagulation effects of betrixaban in healthy subjects. The results of this and previous studies in healthy subjects indicate the potential of andexanet as a universal antidote for FXa inhibitors. An ongoing Phase 3b/4 study (ANNEXA-4) in patients receiving a FXa inhibitor who present with acute major bleeding and require urgent reversal of anticoagulation will provide efficacy and safety information on andexanet in this target patient population.


2021 ◽  
pp. 039139882110322
Author(s):  
Frédéric J Baud ◽  
Vanessa Seif ◽  
Pascal Houzé ◽  
Jean-Herlé Raphalen ◽  
Benoît Pilmis ◽  
...  

Introduction: Adsorption of gentamicin in a polyacrylonitrile filter was previously evidenced in a session lasting 6 h using the NeckEpur model. We extended the study over three consecutive days to mimic the 72-h life span of a filter. Methods: Prismaflex® monitor and ST150® filter were used in the continuous diafiltration (CDF) mode at a 2.5 L/h flowrate. The daily session started with a 6-h session of CDF. Thereafter, the 5-L central compartment was changed using a bag free of gentamicin to assess gentamicin release over the following 18 h. Experiments were repeated on Day 2 and stopped at the end of the 6-h session of CDF on Day 3. The experiment was performed in duplicate. Results: At a 2.5 L/h diafiltration flowrate, the mean daily clearances of gentamicin were 5.5, 4.0, and 3.3 L/h, respectively. The mean diafiltration and adsorption ratios in the daily elimination of gentamicin were 32/68%, 58/42%, and 88/12%, respectively. During days 1 and 2, the mean amount of gentamicin released from the ST150® filter were 14 and 34 mg, respectively. Conclusion: The pharmacokinetics of gentamicin over 3 days is strongly altered by adsorption in the same filter with a progressive decrease of elimination by adsorption, suggesting saturation of the filter. One limitation of our study results from the mode of administration using a bolus dose instead of an infusion over 30 min. Adsorption adds a clearance to those of diafiltration. The time-dependency of gentamicin clearance precludes using a constant dosage regimen over the filter’s life span.


2019 ◽  
Vol 25 ◽  
pp. 107602961986349 ◽  
Author(s):  
Fakiha Siddiqui ◽  
Alfonso Tafur ◽  
Lorenzo Storino Ramacciotti ◽  
Walter Jeske ◽  
Debra Hoppensteadt ◽  
...  

Recombinant coagulation factor Xa (FXa), inactivated Zh-zo, also known as andexanet alfa (AA), is a modified version of human FXa that has been developed to neutralize FXa inhibitors. We studied the reversal effect of AA for these inhibitors in various anticoagulant and thrombin generation (TG) assays. Individual aliquots of normal human plasma containing 1 µg/mL of apixaban, betrixaban, edoxaban, and rivaroxaban, were supplemented with saline or AA at a concentration of 100 µg/mL. Clotting profiles include prothrombinase-induced clotting time, activated partial thromboplastin time, and prothrombin time. Factor Xa activity was measured using an amidolytic method. Thrombin generation was measured using a calibrated automated thrombogram. Differential neutralization of all 4 anticoagulants was noted in the activated clotting time and other clotting tests. The FXa activity reversal profile varied with an observed decrease in apixaban (22%), betrixaban (56%), edoxaban (28%), and rivaroxaban (49%). Andexanet alfa also led to an increased TG in comparison to saline. The peak thrombin was higher (40%), area under the curve (AUC) increased (15%), whereas the lag time (LT) decreased (17%). Andexanet alfa added at 100 µg/mL to various FXa supplemented systems resulted in reversal of the inhibitory effects, restoring the TG profile; AUC, LT, and peak thrombin levels were comparable to those of unsupplemented samples. Andexanet alfa is capable of reversing anti-Xa activity of different oral FXa inhibitors but overshoots thrombogenesis in both the saline and FXa inhibitor supplemented systems. The degree of neutralization of Xa inhibitor is specific to each agent.


1990 ◽  
Vol 47 (4) ◽  
pp. 766-771 ◽  
Author(s):  
S. M. Plakas ◽  
R. W. Dickey ◽  
M. G. Barron ◽  
A. M. Guarino

Ormetoprim is used to potentiate sulfadimethoxine in treating certain bacterial diseases of aquatic species. The tissue disposition and renal excretion of ormetoprim and metabolites were examined after intravascular and oral administration (4 mg∙kg−1) in channel catfish (Ictalurus punctatus). Peak plasma level (0.66 μg∙mL−1) of 14C-ormetoprim occurred at 6 h after oral dosing. The oral bioavailability was estimated at 52%. Ormetoprim and metabolites were widely distributed in the tissues. The tissue concentrations were highest in the liver, trunk kidney, head kidney, and spleen. Clearance of the radiolabel from tissues was rapid. The muscle contained 49.3% of the intravascularly administered dose at 2 h; however, at 72 h, less than 1% of the dose remained in this tissue. 14C-Ormetoprim was more persistent in the skin than in the muscle. Ormetoprim was extensively metabolized in catfish. After intravascular administration, 21.1% of the dose of 14C-ormetoprim was eliminated in the urine in 48 h, predominantly as polar metabolites; less than 4% of the dose was eliminated as the parent compound. Biliary excretion was a minor route of elimination (5–6% of the dose). The data suggest branchial excretion of ormetoprim and/or metabolites.


2021 ◽  
Vol 27 ◽  
pp. 107602962110390
Author(s):  
Victoria M. Stevens ◽  
Toby C. Trujillo ◽  
Tyree H. Kiser ◽  
Robert MacLaren ◽  
Paul M. Reynolds ◽  
...  

The aim of this retrospective study was to compare andexanet alfa and 4-factor prothrombin complex (4F-PCC) for reversal of factor Xa (FXa)-inhibitor bleeding. Patients that received andexanet alfa for reversal were included. An equivalent number of patients administered 4F-PCC for FXa-inhibitor bleeding were randomly selected as historical controls. The primary outcome was effective hemostasis achievement within 12 h, defined using ANNEXA-4 criteria. Thromboembolic events and mortality within 30 days were also evaluated. A total of 32 patients were included. Baseline characteristics were not statistically different between andexanet alfa (n = 16) and 4F-PCC (n = 16). Intracranial bleeding was the primary reversal indication in 43.8% versus 62.5% of patients, respectively. Effective hemostasis was reached in 75.0% of andexanet alfa patients compared to 62.5% of 4F-PCC patients ( P = .70). Thromboembolic events occurred in 4 (25.0%) patients and 3 (18.8%) patients, respectively ( P = .99). Mortality incidence was 12.5% and 31.3%, respectively ( P = .39). Andexanet alfa and 4F-PCC attained hemostasis in a majority of patients. A high, but a similar rate of thromboembolic events was seen with both treatments. Prospective studies are needed to elucidate comparative risks and benefits of the 2 agents.


1985 ◽  
Vol 53 (03) ◽  
pp. 411-414 ◽  
Author(s):  
I R MacGregor ◽  
J Dawes ◽  
D S Pepper ◽  
C V Prowse ◽  
J Stocks

SummaryThree human volunteers were injected with a range of doses of pentosan polysulphate, SP54, i.v. or s.c. A competitive binding assay (CBA) for sulphated polysaccharides was used to detect circulating SP54 after doses as low as 1 mg i.v. and a linear relationship was observed between the peak plasma concentration of SP54 measured by CBA and the administered dose. A comparison was made between the clearance of SP54 measured by CBA and its anticoagulant and lipolytic activities. SP54 was detectable by CBA after doses which caused no alteration in activated partial thromboplastin time (APTT) or anti-factor Xa activity but after which a small increase of lipase activity was measurable. After SP54 at 10 mg i.v. or 100 mg s.c. anti-factor Xa activity was 4-6 times greater than would be expected from the in vitro activity of the concentrations of SP54 measured by CBA. Like heparin and other heparin analogues, SP54 caused an increase in plasma concentrations of platelet factor 4 (PF4) without a concomitant rise in p-thromboglobulin (β-TG).It is concluded that the newly developed CBA will provide a more sensitive means than conventional bioassays for the determination of plasma concentrations of SP54.


Sign in / Sign up

Export Citation Format

Share Document