Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Contemp Clin Trials Commun ; 14: 100318, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30656241

ABSTRACT

OBJECTIVES: The advent of large databases, wearable technology, and novel communications methods has the potential to expand the pool of candidate research participants and offer them the flexibility and convenience of participating in remote research. However, reports of their effectiveness are sparse. We assessed the use of various forms of outreach within a nationwide randomized clinical trial being conducted entirely by remote means. METHODS: Candidate participants at possibly higher risk for atrial fibrillation were identified by means of a large insurance claims database and invited to participate in the study by their insurance provider. Enrolled participants were randomly assigned to one of two groups testing a wearable sensor device for detection of the arrhythmia. RESULTS: Over 10 months, the various outreach methods used resulted in enrollment of 2659 participants meeting eligibility criteria. Starting with a baseline enrollment rate of 0.8% in response to an email invitation, the recruitment campaign was iteratively optimized to ultimately include website changes and the use of a five-step outreach process (three short, personalized emails and two direct mailers) that highlighted the appeal of new technology used in the study, resulting in an enrollment rate of 9.4%. Messaging that highlighted access to new technology outperformed both appeals to altruism and appeals that highlighted accessing personal health information. CONCLUSIONS: Targeted outreach, enrollment, and management of large remote clinical trials is feasible and can be improved with an iterative approach, although more work is needed to learn how to best recruit and retain potential research participants. TRIAL REGISTRATION: Clinicaltrials.govNCT02506244. Registered 23 July 2015.

2.
Am Heart J ; 175: 77-85, 2016 May.
Article in English | MEDLINE | ID: mdl-27179726

ABSTRACT

Efficient methods for screening populations for undiagnosed atrial fibrillation (AF) are needed to reduce its associated mortality, morbidity, and costs. The use of digital technologies, including wearable sensors and large health record data sets allowing for targeted outreach toward individuals at increased risk for AF, might allow for unprecedented opportunities for effective, economical screening. The trial's primary objective is to determine, in a real-world setting, whether using wearable sensors in a risk-targeted screening population can diagnose asymptomatic AF more effectively than routine care. Additional key objectives include (1) exploring 2 rhythm-monitoring strategies-electrocardiogram-based and exploratory pulse wave-based-for detection of new AF, and (2) comparing long-term clinical and resource outcomes among groups. In all, 2,100 Aetna members will be randomized 1:1 to either immediate or delayed monitoring, in which a wearable patch will capture a single-lead electrocardiogram during the first and last 2 weeks of a 4-month period beginning immediately or 4 months after enrollment, respectively. An observational, risk factor-matched control group (n = 4,000) will be developed from members who did not receive an invitation to participate. The primary end point is the incidence of new AF in the immediate- vs delayed-monitoring arms at the end of the 4-month monitoring period. Additional efficacy and safety end points will be captured at 1 and 3 years. The results of this digital medicine trial might benefit a substantial proportion of the population by helping identify and refine screening methods for undiagnosed AF.


Subject(s)
Asymptomatic Diseases/epidemiology , Atrial Fibrillation , Electrocardiography, Ambulatory/methods , Mass Screening , Stroke/prevention & control , Aged , Atrial Fibrillation/diagnosis , Atrial Fibrillation/epidemiology , Atrial Fibrillation/physiopathology , Cost Savings , Female , Humans , Incidence , Male , Mass Screening/economics , Mass Screening/instrumentation , Mass Screening/methods , Middle Aged , Outcome and Process Assessment, Health Care , Risk Factors , Stroke/etiology , Telemedicine/methods , United States/epidemiology
3.
Am Heart J ; 169(6): 751-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26027611

ABSTRACT

This white paper provides a summary of presentations and discussions that were held at an Anticoagulant-Induced Bleeding and Reversal Agents Think Tank co-sponsored by the Cardiac Safety Research Consortium and the US Food and Drug Administration (FDA) at the FDA's White Oak Headquarters on April 22, 2014. Attention focused on a development pathway for reversal agents for the novel oral anticoagulants (NOACs). This is important because anticoagulation is still widely underused for stroke prevention in patients with atrial fibrillation. Undertreatment persists, although NOACs, in general, overcome some of the difficulties associated with anticoagulation provided by vitamin K antagonists. One reason for the lack of a wider uptake is the absence of NOAC reversal agents. As there are neither widely accepted academic and industry standards nor a definitive regulatory policy on the development of such reversal agents, this meeting provided a forum for leaders in the fields of cardiovascular clinical trials and cardiovascular safety to discuss the issues and develop recommendations. Attendees included representatives from pharmaceutical companies; regulatory agencies; end point adjudication specialist groups; contract research organizations; and active, academically based physicians. There was wide and solid consensus that NOACs overall offer improvements in convenience, efficacy, and safety compared with warfarin, even without reversal agents. Still, it was broadly accepted that it would be helpful to have reversal agents available for clinicians to use. Because it is not feasible to do definitive outcomes studies demonstrating a reversal agent's clinical benefits, it was felt that these agents could be approved for use in life-threatening bleeding situations if the molecules were well characterized preclinically, their pharmacodynamic and pharmacokinetic profiles were well understood, and showed no harmful adverse events in early human testing. There was also consensus that after such approval, efforts should be made to augment the available clinical information until such time as there is a body of evidence to demonstrate real-world clinical outcomes with the reversal agents. No recommendations were made for more generalized use of these agents in the setting of non-life-threatening situations. This article reflects the views of the authors and should not be construed to represent FDA's views or policies.


Subject(s)
Anticoagulants/adverse effects , Anticoagulants/therapeutic use , Hemorrhage/prevention & control , Administration, Oral , Antibodies, Monoclonal, Humanized/therapeutic use , Arginine/analogs & derivatives , Arginine/therapeutic use , Cardiovascular Diseases/drug therapy , Factor Xa/therapeutic use , Factor Xa Inhibitors/therapeutic use , Hemorrhage/chemically induced , Humans , Piperazines/therapeutic use , Recombinant Proteins/therapeutic use , Stroke/prevention & control
4.
Clin Pharmacol Drug Dev ; 3(4): 321-7, 2014 07.
Article in English | MEDLINE | ID: mdl-27128839

ABSTRACT

PURPOSE: Because some patients have difficulty swallowing a whole tablet, we investigated the relative bioavailability of a crushed 20 mg rivaroxaban tablet and of 2 alternative crushed tablet dosing strategies. METHODS: Stability and nasogastric (NG) tube adsorption characteristics of a crushed rivaroxaban tablet were assessed. Then, in 55 healthy adults, relative bioavailability of rivaroxaban administered orally as a whole tablet (Reference [Whole-Oral]), crushed tablet in applesauce suspension (Crushed-Oral), or crushed tablet in water suspension via NG tube (Crushed-NG) were determined. RESULTS: There were no significant changes in mean percent of non-degraded rivaroxaban recovered over 4 hours from crushed tablet suspensions (>98.4% recovery across all suspensions and time points) or after NG tube exposure (recovery: 99.1% for silicone and 98.9% for polyvinyl chloride NG tubes). Relative bioavailability was similar between Crushed-Oral and Reference dosing (Cmax and AUC∞ were within the 80-125% bioequivalence limits). Relative bioavailability was also similar between the Crushed-NG and Reference dosing (AUC∞ was within bioequivalence limits; Cmax [90% CI range: 78.5-85.8%] was only slightly below the 80% lower bioequivalence limit). CONCLUSIONS: A crushed rivaroxaban tablet was stable and when administered orally or via NG tube, displayed similar relative bioavailability compared to a whole tablet administered orally.


Subject(s)
Factor Xa Inhibitors/pharmacokinetics , Rivaroxaban/pharmacokinetics , Administration, Oral , Adolescent , Adult , Area Under Curve , Biological Availability , Drug Compounding , Drug Monitoring , Drug Stability , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/chemistry , Female , Half-Life , Healthy Volunteers , Humans , Intubation, Gastrointestinal , Male , Metabolic Clearance Rate , Middle Aged , Rivaroxaban/administration & dosage , Rivaroxaban/adverse effects , Rivaroxaban/chemistry , Tablets , Young Adult
5.
Ann N Y Acad Sci ; 1291: 42-55, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23701516

ABSTRACT

The development of rivaroxaban (XARELTO®) is an important new medical advance in the field of oral anticoagulation. Thrombosis-mediated conditions constitute a major burden for patients, healthcare systems, and society. For more than 60 years, the prevention and treatment of these conditions have been dominated by oral vitamin K antagonists (such as warfarin) and the injectable heparins. Thrombosis can lead to several conditions, including deep vein thrombosis, pulmonary embolism, myocardial infarction, stroke, and/or death. Prevention and treatment of thrombosis with an effective, convenient-to-use oral anticoagulant with a favorable safety profile is critical, especially in an aging society in which the risk of thrombosis, and the potential for bleeding complications, is increasing. Rivaroxaban acts to prevent and treat thrombosis by potently inhibiting coagulation Factor Xa in the blood. Factor Xa converts prothrombin to thrombin, which initiates the formation of blood clots by converting fibrinogen to clot-forming fibrin and leads to platelet activation. After a large and novel clinical development program in over 75,000 patients to date, rivaroxaban has received approval for multiple indications in the United States, European Union, and other countries worldwide to prevent and treat several thrombosis-mediated conditions. This review will highlight some of the unique aspects of the rivaroxaban development program.


Subject(s)
Anticoagulants/administration & dosage , Factor Xa Inhibitors , Morpholines/administration & dosage , Thiophenes/administration & dosage , Thrombosis/drug therapy , Administration, Oral , Animals , Anticoagulants/pharmacokinetics , Atrial Fibrillation/drug therapy , Atrial Fibrillation/epidemiology , Atrial Fibrillation/metabolism , Factor Xa/metabolism , Humans , Morpholines/pharmacokinetics , Rivaroxaban , Thiophenes/pharmacokinetics , Thrombosis/epidemiology , Thrombosis/metabolism , Treatment Outcome
6.
Int J Clin Pharmacol Ther ; 50(8): 584-94, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22578199

ABSTRACT

OBJECTIVE: To assess and validate the application of a non-radioactive assay for cholesteryl ester transfer protein (CETP) activity in clinical samples. DESIGN AND METHODS: In this Phase 0 study, CETP activity was measured following addition of the CETP inhibitor JNJ-28545595 to plasma samples from normolipidemic and three subgroups of dyslipidemic subjects with differing lipid profiles. RESULTS: CETP activity was elevated in plasma samples from dyslipidemic subjects compared to normolipidemic subjects. Increased triglyceride levels correlated with decreased CETP inhibition. The assay was found to have good analytical precision and high throughput potential as required for clinical trial sample analysis. CONCLUSIONS: The results demonstrate that pharmacological inhibition of CETP is affected by the dyslipidemic nature of plasma samples. In addition, since the optimal degree of CETP inhibition for maximal cardiovascular benefit in patients is not known, this assay may be used to help define optimal dosing of CETP inhibitors.


Subject(s)
Biological Assay/methods , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Dyslipidemias/blood , Lipids/blood , Adult , Aged , Cholesterol Ester Transfer Proteins/blood , Dose-Response Relationship, Drug , Female , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Male , Middle Aged , Triglycerides/blood
7.
J Cardiovasc Pharmacol ; 55(5): 459-68, 2010 May.
Article in English | MEDLINE | ID: mdl-20051879

ABSTRACT

OBJECTIVE: Torcetrapib, a prototype cholesteryl ester transfer protein (CETP) inhibitor with potential for decreasing atherosclerotic disease, increased cardiovascular events in clinical trials. The identified hypertensive and aldosterone-elevating actions of torcetrapib may not fully account for this elevated cardiovascular risk. Therefore, we evaluated the effects of torcetrapib on endothelial mediated vasodilation in vivo. METHODS AND RESULTS: In vivo endothelial mediated vasodilation was assessed using ultrasound imaging of acetylcholine-induced changes in rabbit central ear artery diameter. Torcetrapib, in addition to producing hypertension and baseline vasoconstriction, markedly inhibited acetylcholine-induced vasodilation. A structurally distinct CETP inhibitor, JNJ-28545595, did not affect endothelial function despite producing similar degrees of CETP inhibition and high-density lipoprotein elevation. Nitroprusside normalized torcetrapib's basal vasoconstriction and elicited dose-dependent vasodilation of norepinephrine preconstricted arteries in torcetrapib-treated animals, indicating torcetrapib did not impair smooth muscle function. CONCLUSIONS: Torcetrapib significantly impairs endothelial function in vivo, independent of CETP inhibition and high-density lipoprotein elevation. Given the well-documented association of endothelial dysfunction with cardiovascular disease and risk, this activity of torcetrapib may have contributed to increased cardiovascular risk in clinical trials.


Subject(s)
Anticholesteremic Agents/adverse effects , Cardiovascular Diseases/chemically induced , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Endothelium, Vascular/drug effects , Quinolines/adverse effects , Vasodilation/drug effects , Administration, Oral , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/pharmacokinetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Dose-Response Relationship, Drug , Injections, Intravenous , Male , Molecular Structure , Quinolines/administration & dosage , Quinolines/pharmacokinetics , Rabbits
8.
Eur J Clin Pharmacol ; 63(6): 571-81, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17387462

ABSTRACT

OBJECTIVE: To study the effects of amoxicillin, doxycycline, ciprofloxacin, azithromycin, and cefuroxime on the pharmacokinetics and pharmacodynamics of melagatran, the active form of the oral direct thrombin inhibitor ximelagatran, which is a substrate for the P-glycoprotein pump (P-gp) transporter but is not metabolized by the cytochrome P450 (CYP450) enzyme system. METHODS: Five parallel groups of 16 healthy volunteers received two sequential treatments. The first treatment was a single 36-mg dose of ximelagatran. During the second treatment period, one of the above antibiotics was given on days 1-5 after a washout of at least 2 days. A single 36-mg oral dose of ximelagatran was given on the mornings of days 1 and 5 of the second treatment period. RESULTS: No pharmacokinetic interactions were detected between ximelagatran and amoxicillin, doxycycline, or ciprofloxacin as the least-squares geometric mean treatment ratio of ximelagatran with-to-without antibiotic fell within the intervals of 0.80-1.25 for the area under the curve (AUC) and 0.7-1.43 for C(max). After co-administration with azithromycin, the least square mean ratio with-to-without antibiotic for AUC of melagatran was 1.60 (90% CI, 1.40-1.82) on day 1 and 1.41 (90% CI, 1.24-1.61) on day 5. For melagatran C(max), the corresponding ratios were 1.63 (90% CI, 1.38-1.92) and 1.40 (90% CI, 1.18-1.66). After co-administration with cefuroxime, the ratios were 1.23 (90% CI, 1.07-1.42) and 1.16 (90% CI, 0.972-1.38) for AUC and 1.33 (90% CI, 1.07-1.66) and 1.19 (90%CI, 0.888-1.58) for C(max) of melagatran. Co-administration with the antibiotics did not change mean time to C(max), half-life, or renal clearance of melagatran. The melagatran plasma concentration-response relationship for activated partial thromboplastin time (APTT) prolongation was not altered by any of the studied antibiotics, but the increased plasma concentrations of melagatran after co-administration of ximelagatran with azithromycin resulted in a minor increase in the mean maximum APTT of about 15%. CONCLUSION: The pharmacokinetics of ximelagatran were not affected by amoxicillin, doxycycline, or ciprofloxacin. Melagatran exposure was increased when ximelagatran was co-administered with azithromycin and, to a lesser extent, with cefuroxime. APTT was not significantly altered by any of the antibiotics.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anticoagulants/pharmacology , Azetidines/pharmacology , Benzylamines/pharmacology , Thrombin/antagonists & inhibitors , Adult , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Anticoagulants/administration & dosage , Anticoagulants/pharmacokinetics , Azetidines/administration & dosage , Azetidines/pharmacokinetics , Benzylamines/administration & dosage , Benzylamines/pharmacokinetics , Drug Interactions , Female , Humans , Male
9.
Thromb Res ; 119(1): 121-7, 2007.
Article in English | MEDLINE | ID: mdl-16448687

ABSTRACT

INTRODUCTION: Pharmacological enhancement of coagulation using activated prothrombin complex concentrate (APCC) or activated factor VII (FVIIa) might be useful hemostatic approaches to bleeding emergencies during anticoagulant therapy. However, any such intervention should not increase thrombotic risk. We therefore investigated their hemostatic and prothrombotic potential during propagation of large arterial-type thrombin in anticoagulated baboons. MATERIALS AND METHODS: High dose melagatran, a competitive inhibitor of thrombin (0.6 mg/kg/h), or inactivated FVIIa (FVIIai), a competitive inhibitor of FVIIa (2 mg/kg) were used for anticoagulation. APCC or FVIIa were administered to melagatran-anticoagulated animals only. Primary hemostasis was assessed as template bleeding time (BT). Thrombus formation was quantified as fibrin deposition (FD) and platelet deposition (PLD) in synthetic vascular grafts that were deployed for 40 min into arteriovenous shunts. RESULTS: Melagatran (n=11) prolonged BT to 279% (95% CI +/-140%; P<0.019), reduced FD to 33% [+/-8%; P<0.001]; and PLD to 39% [+/-11%; P<0.001] of untreated controls. FVIIai (n=3) prolonged BT (222% [+/-51%; P<0.010]) without inhibiting thrombus propagation. APCC (n=10) reduced the antithrombotic effect of melagatran (FD 52% [+/-9%; P<0.002], PLD 61% [+/-17%; P=0.028] versus melagatran alone) at a dose (250 U/kg) that had no effect on the BT (327% [+/-150%; P=0.607]. Meanwhile, FVIIa (n=12) normalized the BT to 115% (+/-32%; P<0.05) at a dose (270 microg/kg) that was not yet prothrombotic (FD 26% [+/-4%; P<0.001], PLD 39% [+/-9%; P=0.970]). CONCLUSION: Administration of FVIIa during antithrombotic treatment with direct thrombin inhibitors might support hemostasis before promoting the intraluminal expansion of thrombi.


Subject(s)
Anticoagulants/pharmacology , Azetidines/pharmacology , Benzylamines/pharmacology , Factor VIIa/metabolism , Hemostasis , Thrombosis/metabolism , Animals , Case-Control Studies , Humans , Male , Papio , Protein C/metabolism , Recombinant Proteins/chemistry , Thrombosis/drug therapy , Thrombosis/pathology , Time Factors
10.
Thromb Haemost ; 95(3): 447-53, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16525572

ABSTRACT

It was the objective of this study to compare the antithrombotic effects and bleeding profiles of the oral direct thrombin inhibitor ximelagatran, an anticoagulant, and the antiplatelet agent clopidogrel on top of steady-state acetylsalicylic acid (ASA) in a human arterial thrombosis model. Healthy male volunteers (n=62) received ASA (160 mg once daily), plus either clopidogrel for 6 days (loading dose 300 mg, then 75 mg once daily), or a single dose of ximelagatran (36 or 72 mg) on Day 6. Changes in total thrombus area (TTA) under low shear rate (LSR; 212 s(-1)) and high shear rate (HSR; 1690 s(-1)) conditions were measured, using the ex vivo Badimon perfusion chamber model pre-dose and 2 and 5 hours after dosing on Day 6, and capillary bleeding times (CBT) were determined. Ximelagatran plus ASA significantly reduced TTA under LSR and HSR, compared with ASA alone. Ximelagatran plus ASA reduced TTA more than clopidogrel plus ASA under LSR after 2 hours (36 mg, P=0.0011; 72 mg, P<0.0001) and 5 hours (72 mg, P=0.0057), and under HSR after 2 and 5 hours (72 mg, P<0.05). Compared with ASA alone, CBT was markedly prolonged by clopidogrel plus ASA (ratio 6.4; P<0.0001) but only slightly by ximelagatran plus ASA (72 mg ximelagatran, ratio 1.4; P=0.0010). Both drug combinations were well tolerated. Oral ximelagatran plus ASA has a greater antithrombotic effect in this human ex vivo thrombosis model and a less pronounced prolongation of bleeding time than clopidogrel plus ASA.


Subject(s)
Anticoagulants/pharmacology , Aspirin/pharmacology , Azetidines/pharmacology , Benzylamines/pharmacology , Fibrinolytic Agents/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Thrombosis/prevention & control , Ticlopidine/analogs & derivatives , Administration, Oral , Adult , Anticoagulants/administration & dosage , Arteries/drug effects , Arteries/pathology , Aspirin/administration & dosage , Azetidines/administration & dosage , Azetidines/pharmacokinetics , Benzylamines/administration & dosage , Benzylamines/pharmacokinetics , Bleeding Time , Blood Coagulation/drug effects , Clopidogrel , Dose-Response Relationship, Drug , Fibrinolytic Agents/administration & dosage , Humans , Male , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/administration & dosage , Thrombin/antagonists & inhibitors , Thrombosis/pathology , Ticlopidine/administration & dosage , Ticlopidine/pharmacology
11.
Drug Metab Dispos ; 34(5): 775-82, 2006 May.
Article in English | MEDLINE | ID: mdl-16455803

ABSTRACT

A pharmacokinetic interaction between erythromycin and ximelagatran, an oral direct thrombin inhibitor, was demonstrated in this study in healthy volunteers. To investigate possible interaction mechanisms, the effects of erythromycin on active transport mediated by P-glycoprotein (P-gp) in vitro in Caco-2 and P-gp-over-expressing Madin-Darby canine kidney-human multidrug resistance-1 cell preparations and on biliary excretion of melagatran in rats were studied. In healthy volunteers (seven males and nine females; mean age 24 years) receiving a single dose of ximelagatran 36 mg on day 1, erythromycin 500 mg t.i.d. on days 2 to 5, and a single dose of ximelagatran 36 mg plus erythromycin 500 mg on day 6, the least-squares mean estimates (90% confidence intervals) for the ratio of ximelagatran with erythromycin to ximelagatran given alone were 1.82 (1.64-2.01) for the area under the concentration-time curve and 1.74 (1.52-2.00) for the maximum plasma concentration of melagatran, the active form of ximelagatran. Neither the slope nor the intercept of the melagatran plasma concentration-effect relationship for activated partial thromboplastin time statistically significantly differed as a function of whether or not erythromycin was administered with ximelagatran. Ximelagatran was well tolerated regardless of whether it was administered with erythromycin. Erythromycin inhibited P-gp-mediated transport of both ximelagatran and melagatran in vitro and decreased the biliary excretion of melagatran in the rat. These results indicate that the mechanism of the pharmacokinetic interaction between oral ximelagatran and erythromycin may involve inhibition of transport proteins, possibly P-gp, resulting in decreased melagatran biliary excretion and increased bioavailability of melagatran.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Anti-Bacterial Agents/pharmacology , Azetidines/pharmacokinetics , Benzylamines/pharmacokinetics , Erythromycin/pharmacology , Adult , Animals , Anti-Bacterial Agents/adverse effects , Antimalarials/pharmacology , Azetidines/adverse effects , Benzylamines/adverse effects , Bile/metabolism , Biological Transport, Active , Caco-2 Cells , Calcium Channel Blockers/pharmacology , Cell Line , Dogs , Dose-Response Relationship, Drug , Drug Interactions , Erythromycin/adverse effects , Female , Humans , In Vitro Techniques , Male , Partial Thromboplastin Time , Quinidine/pharmacology , Rats , Rats, Sprague-Dawley , Verapamil/pharmacology
12.
Semin Vasc Med ; 5(3): 235-44, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16123910

ABSTRACT

Thrombin plays a central role in thrombus formation through its conversion of fibrinogen to fibrin and activation of platelets as well as amplifying its own generation by feedback activation via factors V, VIII, and XI. Consequently, thrombin represents a logical and promising target for therapeutic interventions against arterial and venous thromboembolic disorders. Ximelagatran is the first oral agent in the new class of direct thrombin inhibitors and is rapidly absorbed and bioconverted to the active moiety, melagatran, which inhibits fluid-phase and clot-bound thrombin with similar high potency. Binding to the active site of thrombin is direct and competitive and does not require the presence of co-factors. Inhibition of thrombin generation and platelet activation has been demonstrated in vitro with melagatran as well as ex vivo after oral administration of ximelagatran to healthy human volunteers. Oral ximelagatran dose dependently reduced the total thrombus area in an ex vivo flow chamber model of arterial thrombosis, reflecting the cumulative effect of inhibition of thrombin activity, thrombin generation, and platelet activation. Melagatran has also been shown to reduce thrombin-mediated inflammation in vitro. The combination of antithrombotic and anti-inflammatory activity with the practicality of oral dosing provided by ximelagatran represents an important new option for the treatment of arterial and venous thromboembolic disorders.


Subject(s)
Anticoagulants/pharmacology , Azetidines/pharmacology , Thrombin/antagonists & inhibitors , Thromboembolism/drug therapy , Administration, Oral , Anti-Inflammatory Agents/administration & dosage , Anti-Inflammatory Agents/pharmacology , Anticoagulants/administration & dosage , Azetidines/administration & dosage , Benzylamines , Blood Coagulation/drug effects , Blood Coagulation/physiology , Humans , Inflammation/prevention & control , Platelet Activation/drug effects , Thromboembolism/prevention & control , Thrombosis/physiopathology , Thrombosis/prevention & control
14.
J Clin Pharmacol ; 44(9): 1063-71, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15317834

ABSTRACT

The oral direct thrombin inhibitor ximelagatran is being developed for the prevention and treatment of thromboembolism. This single-blind, randomized, placebo-controlled, parallel-group study investigated the potential for the interaction of ximelagatran (36 mg every 12 hours for 8 days, measured as its active form melagatran in blood) and amiodarone (single 600-mg oral dose on day 4) in healthy male subjects (n = 26). For amiodarone + ximelagatran versus amiodarone + placebo, geometric mean ratios (90% confidence intervals for amiodarone AUC(0-120) and C(max) were 0.87 (0.69-1.08) and 0.86 (0.66-1.11), respectively. For desethylamiodarone, the principal metabolite of amiodarone, the corresponding ratios were 1.00 (0.89-1.12) for AUC(0-120) and 0.92 (0.77-1.09) for C(max). The geometric mean ratios (90% confidence intervals) for ximelagatran + amiodarone versus ximelagatran were 1.21 (1.17-1.25) for melagatran AUC(0-12) and 1.23 (1.18-1.28) for melagatran C(max). These confidence intervals were within or only slightly outside the interval, suggesting no interaction (0.8-1.25 for the effect of amiodarone on melagatran and 0.7-1.43 for the effect of melagatran on amiodarone or desethylamiodarone). Amiodarone did not affect the concentration-effect relationship of melagatran on activated partial thromboplastin time. Ximelagatran was well tolerated when coadministered with a single dose of amiodarone. Evaluation of the safety of the combination is needed to confirm that the relatively small pharmacokinetic changes in this study are of no clinical significance.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/pharmacokinetics , Anti-Arrhythmia Agents/pharmacokinetics , Anticoagulants/pharmacokinetics , Azetidines/pharmacokinetics , Thrombin/antagonists & inhibitors , Adolescent , Adult , Amiodarone/administration & dosage , Amiodarone/blood , Anti-Arrhythmia Agents/administration & dosage , Anticoagulants/administration & dosage , Area Under Curve , Azetidines/administration & dosage , Azetidines/pharmacology , Benzylamines , Drug Interactions , Half-Life , Humans , Male , Middle Aged , Partial Thromboplastin Time , Regression Analysis , Single-Blind Method , Thrombin/administration & dosage , Thrombin/pharmacology
15.
J Clin Pharmacol ; 44(8): 928-34, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15286097

ABSTRACT

In this randomized, 2-way crossover study, the potential for interaction was investigated between atorvastatin and ximelagatran, an oral direct thrombin inhibitor. Healthy female and male volunteers (n = 16) received atorvastatin 40 mg as a single oral dose and, in a separate study period, ximelagatran 36 mg twice daily for 5 days plus a 40-mg oral dose of atorvastatin on the morning of day 4. In the 15 subjects completing the study, no pharmacokinetic interaction was detected between atorvastatin and ximelagatran for all parameters investigated, including melagatran (the active form of ximelagatran) area under the plasma concentration versus time curve (AUC) and maximum plasma concentration, atorvastatin acid AUC, and AUC of active 3-hydroxy-3-methyl-glutaryl-coenzyme-A (HMG-CoA) reductase inhibitors. Atorvastatin did not alter the melagatran-induced prolongation of the activated partial thromboplastin time, and both drugs were well tolerated when administered in combination. In conclusion, no pharmacokinetic or pharmacodynamic interaction between atorvastatin and ximelagatran was observed in this study.


Subject(s)
Anticoagulants/pharmacology , Anticoagulants/pharmacokinetics , Azetidines/pharmacology , Azetidines/pharmacokinetics , Glycine/analogs & derivatives , Heptanoic Acids/pharmacology , Heptanoic Acids/pharmacokinetics , Prodrugs/pharmacology , Prodrugs/pharmacokinetics , Pyrroles/pharmacology , Pyrroles/pharmacokinetics , Administration, Oral , Adult , Anticholesteremic Agents/pharmacokinetics , Anticholesteremic Agents/pharmacology , Area Under Curve , Atorvastatin , Benzylamines , Cross-Over Studies , Drug Combinations , Drug Interactions , Female , Glycine/blood , Half-Life , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/blood , Male , Partial Thromboplastin Time
16.
J Clin Pharmacol ; 44(8): 935-41, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15286098

ABSTRACT

The interaction potential of digoxin and ximelagatran, an oral direct thrombin inhibitor being developed for the prevention and treatment of thromboembolic disease, was investigated in this randomized, double-blind, 2-way crossover study. On 2 separate occasions, healthy female and male volunteers (n = 16) received ximelagatran 36 mg or placebo twice daily for 8 days separated by a 4- to 14-day washout period. All volunteers received a single oral dose of digoxin 0.5 mg on day 4 of both study periods. No interaction between ximelagatran and digoxin was detected in the pharmaco-kinetic parameters (using a 90% confidence interval [CI] of least squares mean estimate ratios), including melagatran (the active form of ximelagatran) AUC(tau) and C(max) and digoxin AUC(t) and C(max). Digoxin did not alter the melagatran-induced prolongation of the activated partial thromboplastin time, and both drugs were well tolerated when administered in combination. In conclusion, no pharmacokinetic or pharmacodynamic interaction between digoxin and ximelagatran was observed in this study.


Subject(s)
Anticoagulants/pharmacology , Anticoagulants/pharmacokinetics , Azetidines/pharmacology , Azetidines/pharmacokinetics , Digoxin/pharmacology , Digoxin/pharmacokinetics , Glycine/analogs & derivatives , Administration, Oral , Adult , Anti-Arrhythmia Agents/pharmacokinetics , Anti-Arrhythmia Agents/pharmacology , Area Under Curve , Benzylamines , Cross-Over Studies , Double-Blind Method , Drug Interactions , Female , Glycine/blood , Glycine/urine , Half-Life , Humans , Male , Partial Thromboplastin Time , Prodrugs/pharmacokinetics , Prodrugs/pharmacology
17.
Thromb Haemost ; 91(6): 1090-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15175794

ABSTRACT

The objectives were to investigate whether activation of the extrinsic coagulation cascade by recombinant factor VIIa (rFVIIa) reverses the inhibition of thrombin generation and platelet activation by melagatran, the active form of the oral direct thrombin inhibitor ximelagatran. In a single-blind, randomized, parallel-group study, volunteers (20 per group) received a 5-hour intravenous (i.v.) infusion to achieve steady-state melagatran plasma concentrations of approximately 0.5 micromol/L, with a single i.v. bolus of rFVIIa (90 microg/kg) or placebo at 60 minutes. Prothrombin fragment 1+2, thrombin-anti-thrombin complex, fibrinopeptide A, beta-thromboglobulin, and thrombin-activatable fibrinolysis inhibitor were quantified for venous and shed blood. Activated partial thromboplastin time (APTT), prothrombin time (PT), endogenous thrombin potential, thrombus precursor protein (TpP), and plasmin-alpha(2)-antiplasmin complex concentrations were determined in venous blood. Shed blood volume was measured. Melagatran reduced markers of thrombin generation and platelet activation in shed blood and prolonged APTT. rFVIIa increased FVIIa activity, PT, and TpP in venous blood. All other parameters were unaffected. In conclusion, rFVIIa did not reverse the anticoagulant effects of high constant concentrations of melagatran. However, the potential value of higher, continuous or repeated doses of rFVIIa or its use with lower melagatran concentrations has not been excluded.


Subject(s)
Factor VII/administration & dosage , Glycine/analogs & derivatives , Glycine/administration & dosage , Platelet Activation/drug effects , Recombinant Proteins/administration & dosage , Thrombosis/drug therapy , Adult , Azetidines , Benzylamines , Biomarkers/blood , Blood Coagulation Tests , Dose-Response Relationship, Drug , Drug Interactions , Factor VII/pharmacology , Factor VIIa , Glycine/pharmacology , Humans , Male , Recombinant Proteins/pharmacology , Single-Blind Method , Thrombosis/prevention & control , Time Factors
18.
Thromb Res ; 113(1): 85-91, 2004.
Article in English | MEDLINE | ID: mdl-15081569

ABSTRACT

INTRODUCTION: The effect of the oral direct thrombin inhibitor (DTI) ximelagatran (Exanta, AstraZeneca) on the endogenous thrombin potential (ETP) of activated plasma was investigated ex vivo using a thrombin generation assay and compared with recombinant (r)-hirudin and enoxaparin. MATERIALS AND METHODS: 120 healthy male volunteers were randomized to one of six treatment groups (n=20 in each): oral ximelagatran (15, 30, or 60 mg), intravenous r-hirudin (0.4 mg/kg bolus, 0.15 mg/kg/h infusion for 2 h, followed by 0.075 mg/kg/h infusion for 2 h), subcutaneous enoxaparin (100 IU/kg), or control (tap water administered orally). Venous blood was collected predose and at 2, 4, and 10 h postdosing. Thrombin generation was triggered by the addition of tissue factor to platelet-poor plasma, and the ETP and time to peak thrombin generation were measured. RESULTS AND CONCLUSIONS: A significant and dose-dependent reduction in ETP was observed 2 and 4 h after the administration of ximelagatran 30 mg (70.3% of predose, 95% confidence intervals 63.0-78.5, P<0.0001 at 2 h) and 60 mg (49.8%, 43.2-57.4, P<0.0001 at 2 h), r-hirudin (19.5%, 10.1-37.6, P<0.0001 at 2 h), and enoxaparin (34.2%, 21.4-54.7, P<0.0001 at 2 h). Ximelagatran (30 mg, 3.79 min, 3.52-4.08 at 2 h), r-hirudin (6.23 min, 4.93-7.86 at 2 h), and enoxaparin (4.68 min, 3.30-6.64 at 2 h) also delayed the lag phase before the thrombin generation burst compared to placebo (2.92 min, 2.71-3.25 at 2 h). The oral DTI ximelagatran, in its active form melagatran, is a potent thrombin inhibitor that efficiently decreases ETP and delays the generation of thrombin in plasma in this ex vivo model.


Subject(s)
Anticoagulants/pharmacology , Thrombin/antagonists & inhibitors , Administration, Oral , Adult , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Area Under Curve , Azetidines/administration & dosage , Azetidines/adverse effects , Azetidines/pharmacology , Benzylamines , Blood Coagulation Tests , Dose-Response Relationship, Drug , Enoxaparin/administration & dosage , Enoxaparin/adverse effects , Enoxaparin/pharmacology , Hirudins/administration & dosage , Hirudins/adverse effects , Hirudins/pharmacology , Humans , Infusions, Intravenous , Injections, Subcutaneous , Male , Prodrugs/administration & dosage , Prodrugs/adverse effects , Prodrugs/pharmacology , Safety , Thrombin/chemistry , Time Factors
19.
J Clin Pharmacol ; 44(4): 388-93, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15051746

ABSTRACT

Ximelagatran-a direct thrombin inhibitor rapidly converted to its active form, melagatran, after oral administration-is being developed for the prevention and treatment of thromboembolic disease. The pharmacokinetics, pharmacodynamics, and tolerability/safety of ximelagatran following a single 36-mg oral dose of ximelagatran +/- a single oral dose of alcohol (0.5 and 0.6 g ethanol/kg to women and men, respectively) were assessed in a randomized, open-label, two-way crossover study (n = 26). The 90% confidence intervals (CIs) and least squares mean estimates for the ratio of ximelagatran plus alcohol to ximelagatran alone for melagatran AUC (1.04 [90% CI = 1.00-1.08]) and C(max) (1.08 [90% CI = 1.03-1.14]) fell within the bounds demonstrating no interaction. Alcohol did not alter the melagatran-induced prolongation of the activated partial thromboplastin time or the good tolerability/safety profile of ximelagatran. In conclusion, the pharmacokinetics, pharmacodynamics, and tolerability/safety of oral ximelagatran were not affected by alcohol.


Subject(s)
Azetidines/pharmacokinetics , Ethanol/pharmacology , Prodrugs/pharmacokinetics , Thrombin/antagonists & inhibitors , Adult , Area Under Curve , Azetidines/adverse effects , Azetidines/pharmacology , Benzylamines , Drug Interactions , Ethanol/administration & dosage , Female , Humans , Male , Partial Thromboplastin Time , Prodrugs/adverse effects , Prodrugs/pharmacology
20.
Curr Med Res Opin ; 20(3): 325-31, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15025841

ABSTRACT

OBJECTIVE: To investigate whether crushed or dissolved tablets of the oral direct thrombin inhibitor ximelagatran are bioequivalent to whole tablet administration. Ximelagatran is currently under development for the prevention and treatment of thromboembolic disorders. RESEARCH DESIGN AND METHODS: This was an open-label, randomised, three-period, three-treatment crossover study in which 40 healthy volunteers (aged 20-33 years) received a single 36-mg dose of ximelagatran administered in three different ways: I swallowed whole, II crushed, mixed with applesauce and ingested and III dissolved in water and administered via nasogastric tube. RESULTS: The plasma concentrations of ximelagatran, its intermediates and the active form melagatran were determined. Ximelagatran was rapidly absorbed and the bioavailability of melagatran was similar after the three different administrations, fulfilling the criteria for bioequivalence. The mean area under the plasma concentration-versus-time curve (AUC) of melagatran was 1.6 micromol.h/L (ratio 1.01 for treatment II/I and 0.97 for treatment III/I), the mean peak concentration (C(max)) was 0.3 micromol/L (ratio 1.04 for treatment II/I and 1.02 for treatment III/I) and the mean half-life (t(1/2)) was 2.8 h for all treatments. The time to C(max) (t(max)) was 2.2h for the whole tablet and approximately 0.5 h earlier when the tablet was crushed or dissolved (1.7-1.8 h), due to a more rapid absorption. The study drug was well tolerated as judged from the low incidence and type of adverse events reported. CONCLUSION: The present study showed that the pharmacokinetics (AUC and C(max)) of melagatran were not significantly altered whether ximelagatran was given orally as a crushed tablet mixed with applesauce or dissolved in water and given via nasogastric tube.


Subject(s)
Azetidines/administration & dosage , Azetidines/pharmacokinetics , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/pharmacokinetics , Adult , Benzylamines , Cross-Over Studies , Dosage Forms , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL