Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Ann Noninvasive Electrocardiol ; 14 Suppl 1: S42-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19143742

ABSTRACT

BACKGROUND: PC-based ECG measurements must cope with normal as well as pathological ECGs in a reliable manner. EClysis, a software for ECG measurements was tested against reference values from the Common Standards for Quantitative Electrocardiography (CSE) database. METHODS: Digital ECGs (12 leads, 500 Hz) were recorded by the CSE project. Data Set 3 contains reference values for 125 ECGs (33 normal and 92 pathological). Median values of measurements by 11 computer programs and by five cardiologists, respectively, refer to the earliest P and QRS onsets and to the latest P, QRS, and T offsets in any lead of a selected (index) beat. EClysis automatically measured all ECGs, without user interference. RESULTS: The PQRST points were correctly detected but in two ECGs with AV block II-III. The software was not designed to detect atrial activity in atrial fibrillation (n = 9) and flutter (n = 1). In one case of atrial fibrillation, atrial activity interfered with positioning of QRS and T offsets. Regression coefficients between EClysis and CSE (software-generated and human) were above 0.95 (P < 0.0001). The confidence intervals were 95% for the slope and the intercept of the regression lines. CONCLUSIONS: The PC-based detection and analysis of PQRST points showed a high level of agreement with the CSE database reference values.


Subject(s)
Electrocardiography , Pattern Recognition, Automated , Software Validation , Databases, Factual , Electrocardiography/standards , Humans
2.
Nephrol Dial Transplant ; 20(9): 1889-97, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15928099

ABSTRACT

BACKGROUND: This study assessed the feasibility of replacing intravenous (i.v.) dalteparin with the direct thrombin inhibitor (DTI) melagatran administered via dialysis fluid in patients undergoing haemodialysis, and also examined the pharmacokinetics of melagatran with and without dialysis. METHODS: During two 4 h haemodialysis sessions, 10 adult patients were administered i.v. dalteparin. During two subsequent sessions, melagatran was administered as an i.v. bolus before dialysis, and in the dialysis fluid. The pharmacokinetics of melagatran administered as a bolus before dialysis, and of i.v. melagatran during a dialysis-free day, were studied. Dialysis performances were evaluated from clinical criteria including clot formation in the dialyzer and bloodlines, pre-post dialyzer pressures and iohexol clearance. Anticoagulant efficacy was evaluated from dialysis success. RESULTS: All dialysis sessions were successful, with no apparent difference in clot formation between the two treatments. Median iohexol clearance was similar with dalteparin (99-103 ml/min) and melagatran in the dialysis fluid (98-100 ml/min). There was no difference in pre- and post-dialyzer bloodline pressures between the two treatments. During dialysis sessions with melagatran in dialysis fluid, melagatran concentrations in plasma rapidly equilibrated to approximately 70% of those in dialysis fluid. While the clearance of melagatran was low in patients with renal failure (mean+/-SD, 0.93+/-0.36 l/h), haemodialysis provided efficient clearance of melagatran (7.20+/-0.76 l/h). Melagatran clearance by dialysis (104+/-10 ml/min) was comparable to iohexol clearance. CONCLUSIONS: The DTI melagatran administered via dialysis fluid may provide sufficient anticoagulation for haemodialysis. Melagatran is rapidly cleared from plasma by haemodialysis, suggesting that this method may be used to decrease drug levels in patients with renal impairment.


Subject(s)
Dalteparin/therapeutic use , Glycine/analogs & derivatives , Renal Dialysis/adverse effects , Uremia/blood , Uremia/therapy , Adult , Aged , Anticoagulants/therapeutic use , Antithrombins/therapeutic use , Azetidines , Benzylamines , Chronic Disease , Glycine/pharmacokinetics , Glycine/therapeutic use , Humans , International Normalized Ratio , Kidney Failure, Chronic/therapy , Middle Aged
3.
Int J Biochem Cell Biol ; 36(11): 2226-41, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15313468

ABSTRACT

The platelet-derived growth factor (PDGF) family comprises important mitogens for mesenchymal cells. The active dimeric form of PDGF consists of four structurally related A, B, C, and D chains. All PDGF-variants bind to PDGF-receptors. The A and B chains occur with and without basic C-terminal amino acid extensions as long (A(L) and B(L)) and short (A(S) and B(S)) isoforms. PDGF-A and -B form homo- or heterodimers. The biological relevance of short and long isoforms is unknown, although it may relate to different affinities for glycosaminoglycans of the cell glycocalix and intercellular matrix. Commercially available anti-PDGF-A and anti-PDGF-B antibodies cannot discriminate between the short and the long isoforms. Thus, to investigate the function of the long and short isoforms, we raised antibodies specific for the long A and B chain isoforms. The antibodies were affinity-purified and their properties analysed by surface plasmon resonance. Inhibition studies with different PDGF homodimers and dot-blot studies proved their high specificity for the respective isoforms. Both antibodies recognised the target PDGF homodimers complexed to the glycocalix of human arterial smooth muscle cells and human monocyte-derived macrophages. By using these specific antibodies, we were able to confirm at the protein level the synthesis of PDGF-A and -B during differentiation of human monocyte-derived macrophages and to demonstrate the presence of the PDGF-A(L) and PDGF-B(L) isoforms in human arterial tissue.


Subject(s)
Carotid Arteries/metabolism , Cell Differentiation/physiology , Monocytes/metabolism , Myocytes, Smooth Muscle/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Antibodies/immunology , Cells, Cultured , Glycocalyx/metabolism , Humans , Monocytes/cytology , Myocytes, Smooth Muscle/cytology , Platelet-Derived Growth Factor/immunology , Protein Binding , Protein Isoforms/immunology , Protein Isoforms/metabolism , Rabbits , Receptors, Platelet-Derived Growth Factor/metabolism , Surface Plasmon Resonance
4.
Histochem Cell Biol ; 120(5): 371-82, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14557886

ABSTRACT

The platelet-derived growth factor (PDGF) family comprises disulfide-bonded dimeric isoforms and plays a key role in the proliferation and migration of mesenchymal cells. Traditionally, it consists of homo- and heterodimers of A and B polypeptide chains that occur as long (A(L) and B(L)) or short (A(S) and B(S)) isoforms. Short isoforms lack the basic C-terminal extension that mediates binding to heparin. In the present study, we show that certain PDGF isoforms bind in a specific manner to glycosaminoglycans (GAGs). Experiments performed with wild-type and mutant Chinese hamster ovary cells deficient in the synthesis of GAGs revealed that PDGF long isoforms bind to heparan sulfate and chondroitin sulfate, while PDGF short isoforms only bind to heparan sulfate. This was confirmed by digestion of cell surface GAGs with heparitinase and chondroitinase ABC and by incubation with sodium chloride to prevent GAG sulfation. Furthermore, exogenous GAGs inhibited the binding of long isoforms to the cell membrane more efficiently than that of short isoforms. Additionally, we performed surface plasmon resonance experiments to study the inhibition of PDGF isoforms binding to low molecular weight heparin by GAGs. These experiments showed that PDGF-AA(L) and PDGF-BB(S) isoforms bound to GAGs with the highest affinity. In conclusion, PDGF activity at the cell surface may depend on the expression of various cellular GAG species.


Subject(s)
Glycosaminoglycans/metabolism , Platelet-Derived Growth Factor/metabolism , Animals , Antibody Specificity , Becaplermin , CHO Cells , Chondroitin Sulfates/genetics , Chondroitin Sulfates/metabolism , Cricetinae , Dermatan Sulfate/genetics , Dermatan Sulfate/metabolism , Glycosaminoglycans/genetics , Glycosaminoglycans/pharmacology , Heparitin Sulfate/genetics , Heparitin Sulfate/metabolism , Isomerism , Lipoprotein Lipase/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutagenesis , Platelet-Derived Growth Factor/chemistry , Platelet-Derived Growth Factor/immunology , Protein Binding/drug effects , Protein Binding/physiology , Proto-Oncogene Proteins c-sis
5.
Eur J Clin Pharmacol ; 59(4): 283-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12845508

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the effect of acetylsalicylic acid (ASA or aspirin) on the pharmacokinetics (PK) and pharmacodynamics (PD) of melagatran in healthy volunteers. Melagatran is the active form of the oral direct thrombin inhibitor, ximelagatran. METHODS: This was a double-blind, randomised, two-way, crossover study consisting of two treatment periods separated by a washout period of at least 2 weeks. Twelve subjects received, in a randomised order, either melagatran plus ASA in the first treatment period and melagatran plus placebo in the second treatment period or vice versa. Two single doses of ASA were given, first 450 mg on the day before (day 1) and then 150 mg just before administration of melagatran on day 2. Melagatran 4.12 mg was administered as an intravenous (i.v.) infusion over 4 h on day 2 of both treatment periods. Serial blood samples were collected over the course of the study for the determination of melagatran plasma concentration and coagulation analyses [activated partial thromboplastin time (APTT) and activated clotting time (ACT)]. Capillary bleeding time was measured before ASA/placebo on day 1 and before and after the start of the melagatran infusion on day 2. RESULTS: The plasma concentration of melagatran during the i.v. infusion was maintained at about 0.2 micro mol/l, and ASA did not influence the PK parameters of melagatran. APTT and ACT increased with increasing melagatran plasma concentration, and the observed increases were similar whether melagatran was administered on top of ASA or placebo. Administration of ASA significantly prolonged the capillary bleeding time (by 41% relative to placebo). Melagatran also prolonged the bleeding time significantly (by 25% relative to placebo alone), but this prolongation was not significantly different from the observed prolongation when melagatran was administered on top of ASA (by 17% relative to ASA alone). CONCLUSION: In young healthy volunteers, ASA had no effect on the PK or PD properties of melagatran at the studied dose. That the combination of ximelagatran with ASA may be used with acceptable safety must be verified in the relevant patient populations.


Subject(s)
Aspirin/pharmacology , Glycine/analogs & derivatives , Glycine/pharmacology , Glycine/pharmacokinetics , Platelet Aggregation Inhibitors/pharmacology , Thrombin/antagonists & inhibitors , Administration, Oral , Adult , Azetidines/administration & dosage , Benzylamines , Blood Coagulation/drug effects , Cross-Over Studies , Double-Blind Method , Drug Interactions , Glycine/blood , Humans , Infusions, Intravenous , Male , Partial Thromboplastin Time , Prodrugs/administration & dosage , Whole Blood Coagulation Time
6.
Clin Pharmacokinet ; 42(8): 755-64, 2003.
Article in English | MEDLINE | ID: mdl-12846596

ABSTRACT

BACKGROUND: The oral direct thrombin inhibitor ximelagatran is a new class of anticoagulant currently in clinical development for the prevention and treatment of thromboembolic disease. After oral administration, ximelagatran is rapidly absorbed and bioconverted to its active form melagatran. OBJECTIVE: To investigate the influence of mild-to-moderate hepatic impairment on the pharmacokinetic and pharmacodynamic properties of ximelagatran. STUDY DESIGN: Nonblinded, nonrandomised study. PARTICIPANTS: Twelve volunteers with mild-to-moderate hepatic impairment (classified as Child-Pugh A or B) and 12 age-, weight-, and sex-matched control volunteers with normal hepatic function. METHODS: Volunteers received a single oral dose of ximelagatran 24mg. Plasma and urine samples were collected for pharmacokinetic and pharmacodynamic analyses. RESULTS: The absorption and bioconversion of ximelagatran to melagatran were rapid in both groups. The maximum plasma concentration of melagatran (Cmax) was achieved 2-3 hours after administration; the mean elimination half-life (t1/2z) was 3.6 hours for hepatically impaired volunteers and 3.1 hours for the control volunteers. The area under the plasma concentration-time curve (AUC) and Cmax of melagatran in volunteers with hepatic impairment were 11 and 25% lower than in control volunteers, respectively. However, after correcting for the higher renal function (i.e. higher calculated creatinine clearance) in the hepatically impaired volunteers, the ratio of melagatran AUC for hepatically impaired/control volunteers was 0.98 (90% CI 0.80, 1.22), suggesting that mild-to-moderate hepatic impairment had no influence on the pharmacokinetics of ximelagatran. Melagatran was the predominant compound in urine, accounting for 13-14% of the ximelagatran dose. Renal clearance of melagatran was 13% higher in hepatically impaired than in control volunteers. There were no significant differences between the two groups in the concentration-response relationship between plasma melagatran concentration and activated partial thromboplastin time (APTT). Baseline prothrombin time (PT) was slightly longer in the hepatically impaired patients than in the control volunteers, probably reflecting a slight decrease in the activity of coagulation factors. However, when concentrations of melagatran were at their peak, the increase in PT from baseline values was the same in both groups. Capillary bleeding time was measured in the hepatically impaired patients only, and was not increased by ximelagatran. Ximelagatran was well tolerated in both groups. CONCLUSION: There were no differences in the pharmacokinetic or pharmacodynamic properties of melagatran following oral administration of ximelagatran between the hepatically impaired and control volunteers. These findings suggest that dose adjustment for patients with mild-to-moderate impairment of hepatic function is not necessary.


Subject(s)
Anticoagulants/pharmacokinetics , Azetidines/pharmacokinetics , Glycine/analogs & derivatives , Liver Diseases/metabolism , Prodrugs/pharmacokinetics , Thrombin/antagonists & inhibitors , Administration, Oral , Adult , Aged , Amidines/blood , Anticoagulants/blood , Anticoagulants/pharmacology , Area Under Curve , Azetidines/blood , Azetidines/pharmacology , Benzylamines , Glycine/blood , Half-Life , Humans , Metabolic Clearance Rate , Middle Aged , Partial Thromboplastin Time , Prodrugs/metabolism , Prodrugs/pharmacology , Prothrombin Time
7.
Clin Pharmacokinet ; 42(8): 743-53, 2003.
Article in English | MEDLINE | ID: mdl-12846595

ABSTRACT

BACKGROUND: Ximelagatran is an oral direct thrombin inhibitor currently in clinical development as an anticoagulant for the prevention and treatment of thromboembolic disease. After oral administration, ximelagatran is rapidly absorbed and bioconverted to its active form, melagatran. OBJECTIVE: To investigate the effect of severe renal impairment on the pharmacokinetics and pharmacodynamics of melagatran following administration of subcutaneous melagatran and oral ximelagatran. STUDY DESIGN: This was a nonblinded randomised crossover study with 2 study days, separated by a washout period of 1-3 weeks. Twelve volunteers with severe renal impairment and 12 controls with normal renal function were included, with median (range) glomerular filtration rates (GFR) of 13 (5-24) and 86 (70-105) mL/min, respectively. All volunteers received, in a randomised sequence, a 3mg subcutaneous injection of melagatran and a 24mg immediate-release tablet of ximelagatran. Blood samples were collected up to 12 and 14 hours after administration of the subcutaneous and oral doses, respectively, for determination of melagatran plasma concentrations and the activated partial thromboplastin time (APTT), an ex vivo measurement of coagulation time. Urine was collected for 24 hours after each dose for determination of melagatran concentration. RESULTS: For the volunteers with severe renal impairment, the area under the plasma concentration-time curve (AUC) and the half-life of melagatran were significantly higher than in the control group with normal renal function. Least-squares mean estimates of the ratios of the mean AUC for volunteers with severe renal impairment and controls (95% confidence intervals) were 4.03 (3.29-4.93) after subcutaneous melagatran and 5.33 (3.76-7.56) after oral ximelagatran. This result was related to the decreased renal clearance (CL(R)) of melagatran, which was linearly correlated with GFR. In the severe renal impairment and control groups, respectively, the mean CL(R) of melagatran was 12.5 and 81.3 mL/min after subcutaneous administration of melagatran and 14.3 and 107 mL/min after oral administration of ximelagatran. There was a nonlinear relationship between the APTT ratio (postdose/predose APTT value) and melagatran plasma concentration. A statistically significant higher slope of the concentration-effect relationship, described by linear regression of the APTT ratio versus the square root of melagatran plasma concentrations, was estimated for the group with severe renal impairment compared to the control group; however, the increase in slope was minor and the estimated differences in APTT ratio between the groups in the studied concentration range was less than 10% and not considered clincially relevant. Ximelagatran and melagatran were well tolerated in both groups. CONCLUSIONS: After administration of subcutaneous melagatran and oral ximelagatran, subjects with severe renal impairment had significantly higher melagatran exposure and longer half-life because of lower CL(R) of melagatran compared with the control group with normal renal function, suggesting that a decrease in dose and/or an increase in the administration interval in patients with severe renal impairment would be appropriate.


Subject(s)
Anticoagulants/pharmacokinetics , Azetidines/pharmacokinetics , Glycine/analogs & derivatives , Glycine/pharmacokinetics , Kidney Failure, Chronic/metabolism , Prodrugs/pharmacokinetics , Administration, Oral , Adult , Aged , Anticoagulants/administration & dosage , Anticoagulants/adverse effects , Area Under Curve , Azetidines/administration & dosage , Azetidines/adverse effects , Benzylamines , Cross-Over Studies , Dose-Response Relationship, Drug , Female , Glycine/administration & dosage , Glycine/adverse effects , Half-Life , Humans , Injections, Subcutaneous , International Normalized Ratio , Male , Metabolic Clearance Rate , Middle Aged , Partial Thromboplastin Time , Prodrugs/metabolism , Thrombin/antagonists & inhibitors , Time Factors
8.
Clin Pharmacokinet ; 42(5): 475-84, 2003.
Article in English | MEDLINE | ID: mdl-12739985

ABSTRACT

OBJECTIVE: To determine the influence of ethnic origin on the pharmacokinetic and pharmacodynamic properties of melagatran after oral administration of ximelagatran, a novel oral direct thrombin inhibitor. STUDY DESIGN: This was an open-label, non-randomised study with a single study session. SUBJECTS: Thirty-six young healthy male subjects living in France were divided equally according to their ethnic origin (African, Asian and Caucasian). METHODS: All subjects received a single 50mg oral dose of ximelagatran in solution. Blood and urine samples for pharmacokinetic evaluation were collected up to 12 and 24 hours after administration, respectively. Blood samples were also collected to determine the activated partial thromboplastin time (APTT), an ex vivo coagulation time measurement used to demonstrate inhibition of thrombin, up to 24 hours after administration. RESULTS: The absorption of ximelagatran, and its bioconversion to melagatran, was rapid in all three ethnic groups. The metabolite pattern in plasma and urine was similar in all groups, with melagatran being the dominant compound. For ximelagatran, the mean area under the plasma concentration-time curve (AUC) was similar in the three groups, suggesting that there was no difference in the extent to which ximelagatran was absorbed. Melagatran AUC was higher in the Asian subjects, with a mean Asian/Caucasian ratio (95% CI) of 1.23 (1.04, 1.45). This was presumably because of their lower bodyweight, which is correlated to lower renal function. Following normalisation for bodyweight, there were no statistically significant differences between the three ethnic groups. This finding suggests that renal elimination was lower for Asian subjects, whereas there were no differences in the conversion of ximelagatran to melagatran. The interindividual variability of melagatran AUC was low (coefficient of variation 19-26%), and the mean bioavailability of melagatran, estimated using a mean value for melagatran clearance obtained from Caucasian subjects in a previous study, was approximately 20% in all groups (range of mean values 19-23%). APTT increased nonlinearly with increasing melagatran plasma concentration, and no difference in the concentration-response relationship was observed between the groups. CONCLUSIONS: After oral administration of ximelagatran, the pharmacokinetic and pharmacodynamic properties of melagatran are independent of ethnic origin. The elimination of melagatran is correlated with renal function.


Subject(s)
Azetidines/administration & dosage , Glycine , Glycine/analogs & derivatives , Prodrugs/administration & dosage , Racial Groups , Thrombin/antagonists & inhibitors , Administration, Oral , Adult , Area Under Curve , Asian People , Benzylamines , Black People , Glycine/blood , Glycine/pharmacology , Glycine/urine , Humans , Inactivation, Metabolic/physiology , Male , White People
9.
Pacing Clin Electrophysiol ; 26(1P2): 361-6, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12687846

ABSTRACT

The ECG may vary during the day (intra-day), and between days (interday), for the same subject. Variability in ECG characteristic measurements between different investigators is well documented and is often large. During days 1-6 of each placebo period of a two-way crossover Phase I study, digital ECGs were recorded at about 8 and 12 AM in 16 healthy volunteers (8 men, 8 women). Two observers independently analyzed leads V2 and V6 using EClysis software. The durations and amplitudes of major ECG waves and the intervals between major electrocardiographic events were analyzed in a mixed model ANOVA, in which subject, observer, time, and day were treated as random factors. The influence of various corrections for heart rate on the variability of QT intervals was investigated. The difference among subjects explained between 44-81% of the total variability in ECG intervals and amplitudes. Overall, inter- and intraday variability was not statistically significant for any variable. The individualized exponential correction of the QT interval for heart rate eliminated the QT interval dependence on the RR interval in all subjects. Changes in T wave morphology and shortening of the QT interval from morning to noon were observed in ten subjects. The interobserver variability was close to zero (SD < 0.005 ms) for all variables except the PQ interval (SD 1.4 ms). The various sources of variability in determinations of ECG wave characteristics should be considered in the design of clinical studies. The use of EClysis software for ECG measurements is this study made the results highly observer independent.


Subject(s)
Circadian Rhythm , Electrocardiography , Adult , Cross-Over Studies , Female , Heart Rate , Humans , Male , Reference Values , Reproducibility of Results , Signal Processing, Computer-Assisted
10.
Clin Pharmacokinet ; 42(4): 381-92, 2003.
Article in English | MEDLINE | ID: mdl-12648028

ABSTRACT

OBJECTIVE: To investigate the influence of age on the pharmacokinetics and pharmacodynamics of ximelagatran. STUDY DESIGN: This was an open-label, randomised, 3 x 3 crossover study with 4 study days, separated by washout periods of 7 days. SUBJECTS: Subjects comprised 6 healthy young men (aged 20-27 years) and 12 healthy older men and women (aged 56-70 years). METHODS: All subjects received a 2mg intravenous infusion of melagatran over 10 minutes followed, in randomised sequence, by a 20 mg immediate-release tablet of ximelagatran with breakfast, a 20 mg immediate-release tablet of ximelagatran while fasting, and a 7.5 mg subcutaneous injection of ximelagatran. The primary variables were the plasma concentration of melagatran, the active form of ximelagatran, and the activated partial thromboplastin time (APTT), an ex vivo coagulation time measurement used to demonstrate inhibition of thrombin. RESULTS: After oral and subcutaneous administration, ximelagatran was rapidly absorbed and biotransformed to melagatran, its active form and the dominant compound in plasma. The metabolite pattern in plasma and urine was similar in young and older subjects after both oral and subcutaneous administration of ximelagatran clearance of melagatran was correlated with renal function, resulting in about 40% (after intravenous melagatran) to 60% (after oral and subcutaneous ximelagatran) higher melagatran exposure in the older than in the young subjects. Renal clearance of melagatran, was 7.7 L/h and 4.9 L/h in the younger and older subjects, respectively. The interindividual variability inn the area under the melagatran plasma concentration-time curve was low following all regimens (coefficient variation 12-25%). The mean bioavailability of melagatran in young and older subjects was approximately 18 and 12% , respectively, following oral administration of ximalagratan, and 38 and 45%, respectively, following subcutaneous administration of ximelagatran. The bioavailability of melagatran following oral administration of ximelagatran was unaffected by whether subjects were fed or fasting, although the plasma concentration of melagatran peaked about 1 hour later under fed than fasting conditions, due to gastric emptying of the immediate-release tablet formulation used. The APTT as prolonged with increasing melagatran plasma concentration-effect relationship was independent of age. CONCLUSIONS: There were no age-dependent differences in the absorption and biotransformation of ximelagatran, and the observed differences in exposure to melagatran can be explained by differences in renal function between the young and older subjects.


Subject(s)
Azetidines/pharmacology , Fibrinolytic Agents/pharmacology , Glycine/analogs & derivatives , Prodrugs/pharmacology , Thrombin/antagonists & inhibitors , Administration, Oral , Adult , Age Factors , Aged , Area Under Curve , Azetidines/pharmacokinetics , Benzylamines , Biological Availability , Cross-Over Studies , Female , Fibrinolytic Agents/pharmacokinetics , Food-Drug Interactions , Glycine/blood , Glycine/pharmacokinetics , Glycine/urine , Humans , Infusions, Intravenous , Injections, Subcutaneous , Male , Metabolic Clearance Rate , Middle Aged , Prodrugs/pharmacokinetics
11.
Drug Metab Dispos ; 31(3): 294-305, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12584156

ABSTRACT

The absorption, metabolism, and excretion of the oral direct thrombin inhibitor, ximelagatran, and its active form, melagatran, were separately investigated in rats, dogs, and healthy male human subjects after administration of oral and intravenous (i.v.) single doses. Ximelagatran was rapidly absorbed and metabolized following oral administration, with melagatran as the predominant compound in plasma. Two intermediates (ethyl-melagatran and OH-melagatran) that were subsequently metabolized to melagatran were also identified in plasma and were rapidly eliminated. Melagatran given i.v. had relatively low plasma clearance, small volume of distribution, and short elimination half-life. The oral absorption of melagatran was low and highly variable. It was primarily renally cleared, and the renal clearance agreed well with the glomerular filtration rate. Ximelagatran was extensively metabolized, and only trace amounts were renally excreted. Melagatran was the major compound in urine and feces after administration of ximelagatran. Appreciable quantities of ethyl-melagatran were also recovered in rat, dog, and human feces after oral administration, suggesting reduction of the hydroxyamidine group of ximelagatran in the gastrointestinal tract, as demonstrated when ximelagatran was incubated with feces homogenate. Polar metabolites in urine and feces (all species) accounted for a relatively small fraction of the dose. The bioavailability of melagatran following oral administration of ximelagatran was 5 to 10% in rats, 10 to 50% in dogs, and about 20% in humans, with low between-subject variation. The fraction of ximelagatran absorbed was at least 40 to 70% in all species. First-pass metabolism of ximelagatran with subsequent biliary excretion of the formed metabolites account for the lower bioavailability of melagatran.


Subject(s)
Azetidines/pharmacokinetics , Thrombin/antagonists & inhibitors , Absorption/physiology , Administration, Oral , Adult , Animals , Area Under Curve , Azetidines/blood , Azetidines/chemistry , Azetidines/urine , Benzylamines , Dogs , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged , Rats , Rats, Sprague-Dawley , Thrombin/metabolism , Tissue Distribution/physiology
12.
J Am Coll Cardiol ; 41(4): 557-64, 2003 Feb 19.
Article in English | MEDLINE | ID: mdl-12598065

ABSTRACT

OBJECTIVES: The effects of ximelagatran, an oral direct thrombin inhibitor (DTI), recombinant hirudin (r-hirudin) and enoxaparin on thrombin generation and platelet activation were studied in humans. BACKGROUND: Recombinant hirudin (parenteral DTI) and enoxaparin (low molecular weight heparin) have been demonstrated to be clinically effective in acute coronary syndromes. Ximelagatran is currently under investigation for the prevention and treatment of thromboembolism. The shed blood model allows for the study of thrombin generation and platelet activation in humans in vivo. METHODS: This was an open-label, parallel-group study involving 120 healthy male volunteers randomized to receive one of three oral doses of ximelagatran (15, 30 or 60 mg), r-hirudin (intravenous) or enoxaparin (subcutaneous) at doses demonstrated to be clinically effective in acute coronary syndromes, or to serve as a control. Thrombin generation (prothrombin fragment 1+2 [F1+2] and thrombin-antithrombin complex [TAT]) and platelet activation (beta-thromboglobulin [beta-TG]) biomarkers were studied using a shed blood model involving blood collection from skin incisions made using standardized bleeding time devices. RESULTS: Oral ximelagatran, intravenous r-hirudin and subcutaneous enoxaparin rapidly and significantly (p < 0.05) decreased F1+2, TAT and beta-TG levels in shed blood, indicating inhibition of thrombin generation and platelet activation. Statistically significant concentration (melagatran, the active form of ximelagatran)-response relationships for F1+2 (p = 0.005), TAT (p = 0.005) and beta-TG (p < 0.001) levels, with IC(50)s of 0.376 (F1+2), 0.163 (TAT) and 0.115 (beta-TG) micromol/l, were detected. Melagatran showed dose-proportional pharmacokinetics with low variability. All drugs were well tolerated. CONCLUSIONS: Oral administration of the DTI ximelagatran resulted in a rapid inhibition of both thrombin generation and platelet activation in a concentration-dependent manner using a human shed blood model. The inhibition of thrombin generation by 60 mg ximelagatran was comparable to that observed with doses of r-hirudin and enoxaparin demonstrated to be effective for the treatment of acute coronary syndromes.


Subject(s)
Azetidines/administration & dosage , Azetidines/pharmacology , Enoxaparin/pharmacology , Fibrinolytic Agents/pharmacology , Hirudins/pharmacology , Platelet Activation/drug effects , Thrombin/biosynthesis , Thrombin/drug effects , Administration, Oral , Adult , Benzylamines , Dose-Response Relationship, Drug , Humans , Male , Recombinant Proteins/pharmacology , Reference Values , Thrombin/antagonists & inhibitors
13.
Ann Noninvasive Electrocardiol ; 7(4): 289-301, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12431306

ABSTRACT

BACKGROUND: New strategies are needed to improve the results of automatic measurement of the various parts of the ECG signal and their dynamic changes. METHODS: The EClysis software processes digitally-recorded ECGs from up to 12 leads at 500 Hz, using strictly defined algorithms to detect the PQRSTU points and to measure ECG intervals and amplitudes. Calculations are made on the averaged curve of each sampling period (beat group) or as means +/- SD for beat groups, after being analyzed at the individual beat level in each lead. Resulting data sets can be exported for further statistical analyses. Using QT and R-R measured on beat level, an individual correction for the R-R dependence can be performed. RESULTS: EClysis assigns PQRSTU points and intervals in a sensitive and highly reproducible manner, with coefficients of variation in ECG intervals corresponding to ca. 2 ms in the simulated ECG. In the normal ECG, the CVs are 2% for QRS, 0.8% for QT, and almost 6% for PQ intervals. EClysis highlights the increase in QT intervals and the decrease of T-wave amplitudes during almokalant infusion versus placebo. Using the observed linear or exponential relationships to adjust QT for R-R dependence in healthy subjects, one can eliminate this dependence almost completely by individualized correction. CONCLUSIONS: The EClysis system provides a precise and reproducible method to analyze ECGs.


Subject(s)
Electrocardiography , Signal Processing, Computer-Assisted , Adult , Anti-Arrhythmia Agents/pharmacology , Electrocardiography/drug effects , Female , Heart Rate/drug effects , Humans , Male , Models, Statistical , Models, Theoretical , Propanolamines/pharmacology , Reference Values , Reproducibility of Results , Software
14.
Thromb Haemost ; 87(2): 300-5, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11858491

ABSTRACT

Ximelagatran, an oral direct thrombin inhibitor, whose active form is melagatran, was studied using a model of thrombin generation in humans. Healthy male volunteers (18 per group) received ximelagatran (60 mg p.o.), dalteparin (120 IU/kg s.c.) or a control (water p.o.). Shed blood, collected after incision of the forearm with standardised bleeding time devices at pre-dose, and at 2, 4 and 10 h post-dosing, was analysed for markers of thrombin generation. Statistically significant reductions (p < 0.05) in levels of prothrombin fragment 1+2 (F1+2) and thrombin-antithrombin complex (TAT) in shed blood were detected at 2 and 4 h post-dosing in both the ximelagatran and dalteparin groups. Shed blood F1+2 and TAT levels had returned to pre-dose levels at 10 h post-dosing. Using a shed blood model, we demonstrate that the reversible thrombin inhibitor melagatran and, therefore, oral administration of ximelagatran, inhibits thrombin generation in humans after acute activation of coagulation.


Subject(s)
Anticoagulants/pharmacology , Azetidines/pharmacology , Thrombin/antagonists & inhibitors , Administration, Oral , Adult , Anticoagulants/administration & dosage , Anticoagulants/pharmacokinetics , Antithrombin III/analysis , Azetidines/administration & dosage , Azetidines/pharmacokinetics , Benzylamines , Dalteparin/administration & dosage , Dalteparin/pharmacokinetics , Dalteparin/pharmacology , Feedback , Humans , Male , Peptide Fragments/analysis , Peptide Hydrolases/analysis , Prothrombin/analysis , Thrombin/biosynthesis
15.
Thromb Haemost ; 87(2): 231-7, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11858482

ABSTRACT

The novel, oral direct thrombin inhibitor, ximelagatran (formerly H 376/95), represents an advance in antithrombotic therapy through its oral availability. After oral administration, ximelagatran is converted to its active form, melagatran. Melagatran can also be administered subcutaneously (s.c.). The results from the first clinical study with ximelagatran are reported. In this randomized, parallel-group, controlled study, 103 patients scheduled for elective total hip or total knee replacement received s.c. melagatran (1, 2 or 4 mg bid) for 2 days commencing immediately before surgery, followed by oral ximelagatran (6, 12 or 24 mg bid) for 6-9 days. Another 33 patients received dalteparin 5000 IU s.c. once daily, started the evening before surgery, for 8-11 days. At bilateral venography, deep vein thrombosis was found in 20.5% (16/78) of patients who had received s.c. melagatran and oral ximelagatran and in 18.5% (5/27) of patients in the dalteparin group. The study did not evaluate a dose-response for efficacy, and no differences between the three dose levels of melagatran and ximelagatran were shown. No pulmonary embolism was diagnosed during treatment. Total bleeding in the s.c. melagatran plus oral ximelagatran groups showed no dose-response and was similar to that seen in the dalteparin group. The pharmacokinetic properties of melagatran in the surgery patients were consistent with those observed for healthy subjects, and the APTT ratio, which increased non-linearly with plasma melagatran concentration, showed a consistent concentration-effect relationship during the treatment period. Ximelagatran and melagatran were well tolerated. In conclusion, ximelagatran and its active form melagatran appear to be promising agents for the prevention of venous thromboembolism following orthopaedic surgery.


Subject(s)
Anticoagulants/administration & dosage , Arthroplasty, Replacement, Hip , Arthroplasty, Replacement, Knee , Azetidines/administration & dosage , Glycine/analogs & derivatives , Glycine/administration & dosage , Postoperative Complications/prevention & control , Prodrugs/administration & dosage , Thrombin/antagonists & inhibitors , Thromboembolism/prevention & control , Administration, Oral , Adolescent , Aged , Aged, 80 and over , Anticoagulants/therapeutic use , Azetidines/pharmacokinetics , Azetidines/therapeutic use , Benzylamines , Dalteparin/therapeutic use , Female , Glycine/pharmacokinetics , Glycine/therapeutic use , Humans , Incidence , Injections, Subcutaneous , Male , Middle Aged , Partial Thromboplastin Time , Postoperative Complications/epidemiology , Prodrugs/pharmacokinetics , Prodrugs/therapeutic use , Safety , Sweden/epidemiology , Treatment Outcome , Venous Thrombosis/epidemiology , Venous Thrombosis/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...