Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters

Publication year range
1.
Br J Clin Pharmacol ; 90(1): 220-231, 2024 01.
Article in English | MEDLINE | ID: mdl-37567779

ABSTRACT

AIMS: Recombinant factor IX Fc fusion protein (rFIX-Fc) is an extended half-life factor concentrate administered to haemophilia B patients. So far, a population pharmacokinetic (PK) model has only been published for patients aged ≥12 years. The aim was to externally evaluate the predictive performance of the published rFIX-Fc population PK model for patients of all ages and develop a model that describes rFIX-Fc PK using real-world data. METHODS: We collected prospective and retrospective data from patients with haemophilia B treated with rFIX-Fc and included in the OPTI-CLOT TARGET study (NTR7523) or United Kindom (UK)-EHL Outcomes Registry (NCT02938156). Predictive performance was assessed by comparing predicted with observed FIX activity levels. A new population PK model was constructed using nonlinear mixed-effects modelling. RESULTS: Real-world data were obtained from 37 patients (median age: 16 years, range 2-71) of whom 14 were aged <12 years. Observed FIX activity levels were significantly higher than levels predicted using the published model, with a median prediction error of -48.8%. The new model showed a lower median prediction error (3.4%) and better described rFIX-Fc PK, especially for children aged <12 years. In the new model, an increase in age was correlated with a decrease in clearance (P < .01). CONCLUSIONS: The published population PK model significantly underpredicted FIX activity levels. The new model better describes rFIX-Fc PK, especially for children aged <12 years. This study underlines the necessity to strive for representative population PK models, thereby avoiding extrapolation outside the studied population.


Subject(s)
Factor IX , Hemophilia B , Child , Humans , Child, Preschool , Adolescent , Young Adult , Adult , Middle Aged , Aged , Factor IX/therapeutic use , Factor IX/pharmacokinetics , Hemophilia B/drug therapy , Retrospective Studies , Prospective Studies , Recombinant Fusion Proteins/therapeutic use , Recombinant Fusion Proteins/pharmacokinetics , Half-Life
2.
Blood ; 129(4): 520-524, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28064200

ABSTRACT

The von Willebrand receptor complex, which is composed of the glycoproteins Ibα, Ibß, GPV, and GPIX, plays an essential role in the earliest steps in hemostasis. During the last 4 decades, it has become apparent that loss of function of any 1 of 3 of the genes encoding these glycoproteins (namely, GP1BA, GP1BB, and GP9) leads to autosomal recessive macrothrombocytopenia complicated by bleeding. A small number of variants in GP1BA have been reported to cause a milder and dominant form of macrothrombocytopenia, but only 2 tentative reports exist of such a variant in GP1BB By analyzing data from a collection of more than 1000 genome-sequenced patients with a rare bleeding and/or platelet disorder, we have identified a significant association between rare monoallelic variants in GP1BB and macrothrombocytopenia. To strengthen our findings, we sought further cases in 2 additional collections in the United Kingdom and Japan. Across 18 families exhibiting phenotypes consistent with autosomal dominant inheritance of macrothrombocytopenia, we report on 27 affected cases carrying 1 of 9 rare variants in GP1BB.


Subject(s)
Blood Platelets/metabolism , Hemorrhage/genetics , Mutation , Platelet Glycoprotein GPIb-IX Complex/genetics , Thrombocytopenia/genetics , Alleles , Blood Platelets/pathology , Case-Control Studies , Female , Gene Expression , Genes, Dominant , Genome, Human , Hemorrhage/diagnosis , Hemorrhage/metabolism , Hemorrhage/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Pedigree , Platelet Count , Thrombocytopenia/diagnosis , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
3.
JAMA ; 321(1): 56-68, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30620371

ABSTRACT

Importance: Microvascular obstruction commonly affects patients with acute ST-segment elevation myocardial infarction (STEMI) and is associated with adverse outcomes. Objective: To determine whether a therapeutic strategy involving low-dose intracoronary fibrinolytic therapy with alteplase infused early after coronary reperfusion will reduce microvascular obstruction. Design, Setting, and Participants: Between March 17, 2016, and December 21, 2017, 440 patients presenting at 11 hospitals in the United Kingdom within 6 hours of STEMI due to a proximal-mid-vessel occlusion of a major coronary artery were randomized in a 1:1:1 dose-ranging trial design. Patient follow-up to 3 months was completed on April 12, 2018. Interventions: Participants were randomly assigned to treatment with placebo (n = 151), alteplase 10 mg (n = 144), or alteplase 20 mg (n = 145) by manual infusion over 5 to 10 minutes. The intervention was scheduled to occur early during the primary PCI procedure, after reperfusion of the infarct-related coronary artery and before stent implant. Main Outcomes and Measures: The primary outcome was the amount of microvascular obstruction (% left ventricular mass) demonstrated by contrast-enhanced cardiac magnetic resonance imaging (MRI) conducted from days 2 through 7 after enrollment. The primary comparison was the alteplase 20-mg group vs the placebo group; if not significant, the alteplase 10-mg group vs the placebo group was considered a secondary analysis. Results: Recruitment stopped on December 21, 2017, because conditional power for the primary outcome based on a prespecified analysis of the first 267 randomized participants was less than 30% in both treatment groups (futility criterion). Among the 440 patients randomized (mean age, 60.5 years; 15% women), the primary end point was achieved in 396 patients (90%), 17 (3.9%) withdrew, and all others were followed up to 3 months. In the primary analysis, the mean microvascular obstruction did not differ between the 20-mg alteplase and placebo groups (3.5% vs 2.3%; estimated difference, 1.16%; 95% CI, -0.08% to 2.41%; P = .32) nor in the analysis of 10-mg alteplase vs placebo groups (2.6% vs 2.3%; estimated difference, 0.29%; 95% CI, -0.76% to 1.35%; P = .74). Major adverse cardiac events (cardiac death, nonfatal MI, unplanned hospitalization for heart failure) occurred in 15 patients (10.1%) in the placebo group, 18 (12.9%) in the 10-mg alteplase group, and 12 (8.2%) in the 20-mg alteplase group. Conclusions and Relevance: Among patients with acute STEMI presenting within 6 hours of symptoms, adjunctive low-dose intracoronary alteplase given during the primary percutaneous intervention did not reduce microvascular obstruction. The study findings do not support this treatment. Trial Registration: ClinicalTrials.gov Identifier: NCT02257294.


Subject(s)
Coronary Occlusion/drug therapy , ST Elevation Myocardial Infarction/drug therapy , Tissue Plasminogen Activator/administration & dosage , Aged , Area Under Curve , Cardiac Catheters , Combined Modality Therapy , Coronary Angiography , Coronary Occlusion/surgery , Coronary Vessels , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intra-Arterial , Magnetic Resonance Imaging , Male , Middle Aged , Percutaneous Coronary Intervention , Quality of Life , ST Elevation Myocardial Infarction/surgery , Tissue Plasminogen Activator/adverse effects , Treatment Failure , Troponin T/blood
4.
Stroke ; 46(12): 3543-6, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26514192

ABSTRACT

BACKGROUND AND PURPOSE: We compared the fibrinolytic activity of tenecteplase and alteplase in patients with acute ischemic stroke, and explored the association between hypofibrinogenaemia and intracerebral hemorrhage. METHODS: Venous blood samples from a subgroup of participants in the Alteplase-Tenecteplase Trial Evaluation for Stroke Thrombolysis (ATTEST) study were obtained at pretreatment, 3 to 12 hours, and 24±3 hours post-intravenous thrombolysis for analyses of plasminogen, plasminogen activator inhibitor-1, d-dimer, factor V, fibrinogen, and fibrin(ogen) degradation products, in addition to routine coagulation assays. Related sample Wilcoxon signed-rank tests were used to test the within-group changes, and independent Mann-Whitney tests for between-group differences. RESULTS: Thirty patients were included (alteplase=14 and tenecteplase=16) with similar baseline demographics. Compared with baseline, alteplase caused significant hypofibrinogenaemia (P=0.002), prolonged prothrombin time (P=0.011), hypoplasminogenaemia (P=0.001), and lower factor V (P=0.002) at 3 to 12 hours after administration with persistent hypofibrinogenaemia at 24 hours (P=0.011), whereas only minor hypoplasminogenaemia (P=0.029) was seen in the tenecteplase group. Tenecteplase consumed less plasminogen (P<0.001) and fibrinogen (P=0.002) compared with alteplase. CONCLUSIONS: In patients with acute ischemic stroke, alteplase 0.9 mg/kg caused significant disruption of the fibrinolytic system, whereas tenecteplase 0.25 mg/kg did not, consistent with the trend toward lower intracerebral hemorrhage incidence with tenecteplase in the ATTEST study. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifier: NCT01472926.


Subject(s)
Blood Coagulation/drug effects , Brain Ischemia/drug therapy , Fibrinolytic Agents/therapeutic use , Stroke/drug therapy , Tissue Plasminogen Activator/therapeutic use , Aged , Aged, 80 and over , Blood Coagulation/physiology , Brain Ischemia/blood , Brain Ischemia/diagnosis , Female , Humans , Male , Middle Aged , Stroke/blood , Stroke/diagnosis , Tenecteplase , Tissue Plasminogen Activator/pharmacology , Treatment Outcome
5.
Eur J Haematol ; 89(4): 336-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22775476

ABSTRACT

OBJECTIVE: Cardiovascular disease (CVD) mortality is reported to be lower in haemophilia patients than in the general population, but information on the occurrence of non-fatal CVD is lacking. The aim of our study was to assess CVD history in a cohort of living haemophilia patients. METHODS: Retrospective data on the occurrence of myocardial infarction, angina pectoris, ischaemic stroke and intracranial bleeding in 709 living Dutch and British haemophilia patients aged 30 yr or older were analysed and compared with the general age-matched male population. RESULTS: There was a trend towards a lower cumulative incidence of myocardial infarction (1.7% vs. 4.0%) and ischaemic stroke (0% vs. 1.5%) in patients with severe haemophilia than in the general population, while the occurrence of angina pectoris was similar (3.2 vs. 3.7%). As expected, the cumulative incidence of intracranial bleeding was, on the other hand, significantly increased in haemophilia patients (1.6% vs. 0.4% in the general population). CONCLUSION: Our results suggest a protective effect of severe haemophilia against acute ischaemic CVD.


Subject(s)
Cardiovascular Diseases/complications , Hemophilia A/complications , Adult , Aged , Aged, 80 and over , Cohort Studies , Humans , Male , Middle Aged , Netherlands , United Kingdom
7.
Ann Intern Med ; 153(8): 523-31, 2010 Oct 19.
Article in English | MEDLINE | ID: mdl-20956709

ABSTRACT

BACKGROUND: In patients with a first unprovoked venous thromboembolism (VTE), an elevated d-dimer level after anticoagulation is stopped is a risk factor for recurrent VTE. However, questions remain about the utility of measuring d-dimer in clinical practice. PURPOSE: To determine whether the timing of testing, patient age, and the cut point used to define a positive or negative result affect the ability of d-dimer testing to distinguish risk for recurrent disease. DATA SOURCES: Comprehensive search of electronic databases (MEDLINE, EMBASE, CINAHL, and the Cochrane Central Register of Controlled Trials) until July 2010, supplemented by reviewing conference abstracts and contacting content experts. STUDY SELECTION: 7 prospective studies that investigated an association between d-dimer, measured after stopping anticoagulation, and disease recurrence in patients with a first unprovoked VTE (proximal deep venous thrombosis, pulmonary embolism, or both). DATA EXTRACTION: Patient-level databases were obtained, transferred to a central database, checked, completed with further information provided by study investigators, and pooled into a single database. DATA SYNTHESIS: 1818 patients with a first unprovoked VTE were followed for a mean of 26.9 months (SD, 19.1). A study-stratified multivariate Cox regression model, which included patient age, sex, hormone therapy use at the time of the index event, body mass index, timing of postanticoagulation d-dimer testing, and inherited thrombophilia as possible confounders, indicated that the hazard ratio for d-dimer status (positive vs. negative) was 2.59 (95% CI, 1.90 to 3.52). Only male sex had a significant effect on risk for recurrent VTE independent of d-dimer status. The Cox regression model and the log-rank test confirmed that the risk for recurrent VTE was higher in patients with a positive d-dimer result than in those with a negative result, regardless of the timing of postanticoagulation d-dimer testing or patient age. No study- or assay-specific d-dimer effect was found, and reassessing the analysis after recoding data according to specific quantitative d-dimer cut points (500 µg/L and 250 µg/L) did not change the results. LIMITATIONS: Unmeasured variables could have affected the risk for recurrent VTE. The study population was predominantly white. CONCLUSION: In patients with a first unprovoked VTE who have their d-dimer level measured after stopping anticoagulation, the timing of d-dimer testing, patient age, and the assay cut point used do not affect the ability of d-dimer to distinguish patients with a higher or lower risk for recurrent VTE.


Subject(s)
Anticoagulants/therapeutic use , Fibrin Fibrinogen Degradation Products/analysis , Venous Thromboembolism/blood , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Biomarkers/blood , Female , Humans , Male , Middle Aged , Proportional Hazards Models , Recurrence , Risk Factors , Sex Factors , Time Factors , Venous Thromboembolism/drug therapy , Withholding Treatment , Young Adult
9.
Medicine (Baltimore) ; 99(9): e19339, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32118768

ABSTRACT

Ageing people with hemophilia (PWH) have a higher prevalence of hypertension than the general population. This study aimed to determine whether macroscopic hematuria was associated with hypertension in PWH in a post hoc analysis using data from a cross-sectional study conducted by the ADVANCE Working Group (the H3 study), which included PWH ≥ 40 years of age. Data from 16 contributing centers, located in 13 European countries and Israel, were analyzed using logistic regression models. Of 532 recruited PWH in the H3 study, 117 had hypertension and a positive family history of hypertension (hypertension FH+), 75 had hypertension and a negative family history of hypertension (hypertension FH-), 290 had no diagnosis of hypertension, and the remaining 50 had missing hypertension data. Logistic regressions showed that macroscopic hematuria was associated with hypertension FH+, both in the univariate (OR = 1.84 [1.17-2.90], P = .01) and in the multivariate model (OR = 1.80 [1.03-3.16], P = .04). Macroscopic hematuria was not associated with hypertension FH-. Moreover, in a multivariate logistic regression the odds of hypertension FH+ were increased with the number of macroscopic hematuria episodes. The association between macroscopic hematuria and hypertension was significant for PWH with a family history of hypertension.


Subject(s)
Hematuria/etiology , Hemophilia A/complications , Hypertension/complications , Adult , Aged , Aged, 80 and over , Aging , Cross-Sectional Studies , Female , Hematuria/epidemiology , Hematuria/physiopathology , Hemophilia A/epidemiology , Hemophilia A/physiopathology , Humans , Hypertension/epidemiology , Hypertension/physiopathology , Israel/epidemiology , Logistic Models , Male , Middle Aged , Risk Factors
10.
Res Pract Thromb Haemost ; 4(2): 309-317, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32110762

ABSTRACT

INTRODUCTION: Macroscopic hematuria is considered a significant risk factor for urologic disease, and it is highly prevalent in people with hemophilia. AIM: To determine whether prophylactic factor replacement therapy is associated with reduced occurrence of macroscopic hematuria in people with hemophilia in a post hoc analysis using data from a cross-sectional study conducted by the Age-Related Developments and Comobordities in Hemophilia (ADVANCE) Working Group that included males with hemophilia ≥40 years of age. METHODS: Data from 16 contributing centers, in 13 European countries and Israel, were analyzed using logistic regression. Of 532 recruited individuals, this analysis included 370 patients with moderate or severe hemophilia who received on-demand or prophylactic therapy. RESULTS: For patients with a history of macroscopic hematuria, we analyzed the association between prophylaxis and reoccurrence of macroscopic hematuria within the past 5 years (n = 235 patients). Frequent (≥3 times/wk) prophylaxis was negatively associated with a recent episode of macroscopic hematuria (odds ratio [OR], 0.38; 95% confidence interval [CI], 0.18-0.76). We also analyzed whether prophylaxis corresponded to a lower lifetime number of macroscopic hematuria episodes (n = 285 patients). Frequent prophylaxis for >15 years was associated with a lower number of episodes compared to on-demand treatment (OR, 0.29; 95% CI, 0.16-0.54), whereas nonsteroidal anti-inflammatory drugs (NSAIDs) and severe hemophilia were associated with a higher number. There was no association of prophylaxis <3 times/wk with hematuria. CONCLUSION: Frequent prophylaxis was negatively associated with the number of episodes of macroscopic hematuria in people with hemophilia. Prevalence of macroscopic hematuria was higher among individuals with severe hemophilia and those regularly using NSAIDs.

11.
J Thromb Haemost ; 18(9): 2209-2214, 2020 09.
Article in English | MEDLINE | ID: mdl-32634856

ABSTRACT

BACKGROUND: Thrombomodulin-associated coagulopathy (TM-AC) is a rare bleeding disorder in which a single reported p.Cys537* variant in the thrombomodulin gene THBD causes high plasma thrombomodulin (TM) levels. High TM levels attenuate thrombin generation and delay fibrinolysis. OBJECTIVES: To report the characteristics of pedigree with a novel THBD variant causing TM-AC, and co-inherited deficiency of thrombin-activatable fibrinolysis inhibitor (TAFI). PATIENTS/METHODS: Identification of pathogenic variants in hemostasis genes by next-generation sequencing and case recall for deep phenotyping. RESULTS: Pedigree members with a previously reported THBD variant predicting p.Pro496Argfs*10 and chain truncation in TM transmembrane domain had abnormal bleeding and greatly increased plasma TM levels. Affected cases had attenuated thrombin generation and delayed fibrinolysis similar to previous reported TM_AC cases with THBD p.Cys537*. Coincidentally, some pedigree members also harbored a stop-gain variant in CPB2 encoding TAFI. This reduced plasma TAFI levels but was asymptomatic. Pedigree members with TM-AC caused by the p.Pro496Argfs*10 THBD variant and also TAFI deficiency had a partially attenuated delay in fibrinolysis, but no change in the defective thrombin generation. CONCLUSIONS: These data extend the reported genetic repertoire of TM-AC and establish a common molecular pathogenesis arising from high plasma levels of TM extra-cellular domain. The data further confirm that the delay in fibrinolysis associated with TM-AC is directly linked to increased TAFI activation. The combination of the rare variants in the pedigree members provides a unique genetic model to develop understanding of the thrombin-TM system and its regulation of TAFI.


Subject(s)
Blood Coagulation Disorders , Carboxypeptidase B2 , Carboxypeptidase B2/genetics , Fibrinolysis/genetics , Humans , Pedigree , Thrombin , Thrombomodulin/genetics
12.
J Am Heart Assoc ; 9(3): e014066, 2020 02 04.
Article in English | MEDLINE | ID: mdl-31986989

ABSTRACT

Background Impaired microcirculatory reperfusion worsens prognosis following acute ST-segment-elevation myocardial infarction. In the T-TIME (A Trial of Low-Dose Adjunctive Alteplase During Primary PCI) trial, microvascular obstruction on cardiovascular magnetic resonance imaging did not differ with adjunctive, low-dose, intracoronary alteplase (10 or 20 mg) versus placebo during primary percutaneous coronary intervention. We evaluated the effects of intracoronary alteplase, during primary percutaneous coronary intervention, on the index of microcirculatory resistance, coronary flow reserve, and resistive reserve ratio. Methods and Results A prespecified physiology substudy of the T-TIME trial. From 2016 to 2017, patients with ST-segment-elevation myocardial infarction ≤6 hours from symptom onset were randomized in a double-blind study to receive alteplase 20 mg, alteplase 10 mg, or placebo infused into the culprit artery postreperfusion, but prestenting. Index of microcirculatory resistance, coronary flow reserve, and resistive reserve ratio were measured after percutaneous coronary intervention. Cardiovascular magnetic resonance was performed at 2 to 7 days and 3 months. Analyses in relation to ischemic time (<2, 2-4, and ≥4 hours) were prespecified. One hundred forty-four patients (mean age, 59±11 years; 80% male) were prospectively enrolled, representing 33% of the overall population (n=440). Overall, index of microcirculatory resistance (median, 29.5; interquartile range, 17.0-55.0), coronary flow reserve(1.4 [1.1-2.0]), and resistive reserve ratio (1.7 [1.3-2.3]) at the end of percutaneous coronary intervention did not differ between treatment groups. Interactions were observed between ischemic time and alteplase for coronary flow reserve (P=0.013), resistive reserve ratio (P=0.026), and microvascular obstruction (P=0.022), but not index of microcirculatory resistance. Conclusions In ST-segment-elevation myocardial infarction with ischemic time ≤6 hours, there was overall no difference in microvascular function with alteplase versus placebo. Clinical Trial Registration URL: https://www.clinicaltrials.gov. Unique identifier: NCT02257294.


Subject(s)
Fibrinolytic Agents/administration & dosage , Fractional Flow Reserve, Myocardial/drug effects , Microcirculation/drug effects , Percutaneous Coronary Intervention , ST Elevation Myocardial Infarction/therapy , Thrombolytic Therapy , Tissue Plasminogen Activator/administration & dosage , Aged , Double-Blind Method , Female , Fibrinolytic Agents/adverse effects , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Prospective Studies , ST Elevation Myocardial Infarction/diagnostic imaging , ST Elevation Myocardial Infarction/physiopathology , Thrombolytic Therapy/adverse effects , Time Factors , Tissue Plasminogen Activator/adverse effects , Treatment Outcome , United Kingdom
13.
Ann Intern Med ; 149(7): 481-90, W94, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18838728

ABSTRACT

BACKGROUND: The optimal duration of anticoagulation for a first episode of unprovoked venous thromboembolism (VTE) is uncertain. Methods for predicting risk for recurrence may identify low-risk patients who are less likely to benefit from prolonged anticoagulation. PURPOSE: To synthesize evidence evaluating the value of D-dimer as a predictor of recurrent disease in patients who have stopped anticoagulant therapy after a first unprovoked VTE. DATA SOURCES: The MEDLINE, EMBASE, CINAHL, and Cochrane databases were searched until March 2008 without language restrictions. The strategy was supplemented with manual review of reference lists and contact with content experts. STUDY SELECTION: Randomized, controlled trials or prospective cohort studies that measured D-dimer after anticoagulant therapy in patients who received at least 3 months of anticoagulant treatment of unprovoked VTE. DATA EXTRACTION: Two authors independently reviewed articles and extracted data. DATA SYNTHESIS: Seven studies, totaling 1888 patients with a first unprovoked VTE, were eligible for analysis. During 4500 person-years of follow up, annual rates of recurrent VTE differed statistically significantly: 8.9% (95% CI, 5.8% to 11.9%) in patients with positive D-dimer results and 3.5% (CI, 2.7% to 4.3%) in patients with negative D-dimer results. LIMITATION: The duration of anticoagulation, timing of D-dimer testing, and D-dimer assay varied across studies. CONCLUSION: In patients who have completed at least 3 months of anticoagulation for a first episode of unprovoked VTE and after approximately 2 years of follow-up, a negative D-dimer result was associated with a 3.5% annual risk for recurrent disease, whereas a positive D-dimer result was associated with an 8.9% annual risk for recurrence. These rates should inform decisions about the balance of risks and benefits of prolonging anticoagulation.


Subject(s)
Anticoagulants/administration & dosage , Fibrin Fibrinogen Degradation Products/analysis , Venous Thromboembolism/blood , Venous Thromboembolism/drug therapy , Drug Administration Schedule , Female , Humans , Male , Recurrence , Risk Factors , Withholding Treatment
14.
Thromb Haemost ; 99(3): 616-22, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18327412

ABSTRACT

During surgery and childbirth, patients with hereditary antithrombin (AT) deficiency are at high risk for thrombosis, and heparin prophylaxis may not be sufficiently efficacious. In these patients, exogenous AT may be used in association with heparin. A recombinant human AT (generic name: antithrombin alfa) has been developed. This multi-center study assessed the efficacy and safety of prophylactic intravenous administration of antithrombin alfa to hereditary AT deficient patients in high risk situations, including elective surgery, childbirth, or cesarean section. Antithrombin alfa was administered prior to and during the high risk period for restoration and maintenance of AT activity at 100% of normal. Heparin, low-molecular-weight heparin, and/or vitamin K antagonists were used according to standard of care. The primary efficacy endpoint was the incidence of acute deep vein thrombosis (DVT) from baseline up to day 30 post dosing as assessed by independent central review of duplex ultrasonograms and/or venograms. Safety was assessed based on adverse events (AEs) and laboratory evaluations. Five surgical and nine obstetrical hereditary AT deficiency patients received antithrombin alfa for a mean period of seven days. No clinically overt DVT occurred. Central review of ultrasonograms identified signs of acute DVT in two out of 13 evaluable patients. No antithrombin alfa-related AEs were reported. No patient developed anti-antithrombin alfa antibodies. In conclusion, this study suggests that antithrombin alfa is a safe and effective alternative to human plasma-derived AT for treating hereditary AT deficiency patients at high risk for thromboembolic events.


Subject(s)
Anticoagulants/administration & dosage , Antithrombins/administration & dosage , Blood Coagulation Disorders, Inherited/drug therapy , Venous Thrombosis/prevention & control , Adult , Aged , Anticoagulants/adverse effects , Antithrombins/adverse effects , Antithrombins/deficiency , Antithrombins/genetics , Arthroplasty, Replacement, Hip/adverse effects , Blood Coagulation/drug effects , Blood Coagulation Disorders, Inherited/blood , Blood Coagulation Disorders, Inherited/complications , Blood Coagulation Disorders, Inherited/genetics , Cesarean Section/adverse effects , Delivery, Obstetric/adverse effects , Drug Administration Schedule , Europe , Female , Heparin/therapeutic use , Humans , Infusions, Intravenous , Male , Mammaplasty/adverse effects , Middle Aged , Phlebography , Prospective Studies , Recombinant Proteins/administration & dosage , Time Factors , Treatment Outcome , Ultrasonography, Doppler, Duplex , United States , Venous Thrombosis/blood , Venous Thrombosis/etiology , Venous Thrombosis/genetics , Venous Thrombosis/pathology
15.
Drugs ; 67(10): 1429-40, 2007.
Article in English | MEDLINE | ID: mdl-17600391

ABSTRACT

Antithrombin is a glycoprotein critical to the regulation of coagulation. Its primary action is the inhibition of the activated coagulation factors IIa (thrombin) and Xa. In addition there is growing evidence to suggest that antithrombin also plays a role in the inhibition of inflammation within the environment of the vascular endothelium. Reduced plasma antithrombin may result from congenital deficiency or arise secondarily from a range of disorders such as liver dysfunction, premature infancy and sepsis, or as a result of interventions such as major surgery or cardiopulmonary bypass. Congenital antithrombin deficiency is the most clinically important of the inherited thrombophilias resulting in thrombosis in the majority of those affected. The challenge in managing these patients is preventing potentially life-threatening thrombosis, while minimising the equally significant risk of haemorrhage associated with long-term anticoagulation. This is achieved in the first instance by identifying high-risk episodes such as surgery, immobility and pregnancy for which prophylactic anticoagulation can be used in the short term. Prophylaxis for such periods is best provided by the use of low molecular weight heparin (LMWH) with substitution by or addition of antithrombin concentrate in particularly high-risk circumstances. In the case of pregnancy, antithrombin concentrate is often used around the time of birth when LMWH may increase the risk of post-partum haemorrhage. As patients with congenital antithrombin deficiency get older so their thrombotic risk gradually increases and for many patients long-term anticoagulation becomes unavoidable because of recurrent episodes of venous thromboembolism. There has been much interest in the role of antithrombin deficiency in the setting of sepsis and the critically ill patient where there is a clear correlation between severity of illness and degree of antithrombin reduction. It is not clear yet, however, to what extent the depletion of antithrombin affects the clinical condition of such patients. A number of trials have investigated the use of antithrombin as a treatment in the intensive care setting with the overall conclusion being that there is some benefit to its use but only if large supra-physiological doses are used. It has also become clear that the concurrent use of any form of heparin removes whatever benefit may be derived from antithrombin treatment in this setting. Until recently, antithrombin replacement was only available as a pooled plasma-derived product, which despite effective viral inactivation still carries an uncertain risk of transfusion transmitted infection. A recombinant antithrombin product now under investigation, and recently licensed in Europe, may provide a useful alternative treatment option.


Subject(s)
Antithrombin III Deficiency , Antithrombin III/physiology , Anticoagulants/therapeutic use , Antithrombin III/therapeutic use , Antithrombin III Deficiency/epidemiology , Antithrombin III Deficiency/etiology , Antithrombin III Deficiency/genetics , Antithrombin III Deficiency/therapy , Blood Component Transfusion , Female , Heparin, Low-Molecular-Weight/therapeutic use , Humans , Pregnancy , Pregnancy Complications, Hematologic/drug therapy , Prevalence , Recombinant Proteins/therapeutic use , Sepsis/complications , Sepsis/therapy
16.
Clin Pharmacokinet ; 56(12): 1555-1566, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28409488

ABSTRACT

BACKGROUND: Warfarin acts by inhibiting the reduction of vitamin K (VK) to its active form, thereby decreasing the production of VK-dependent coagulation proteins. The aim of this research is to develop a joint model for the VK-dependent clotting factors II, VII, IX and X, and the anticoagulation proteins, proteins C and S, during warfarin initiation. METHODS: Data from 18 patients with atrial fibrillation who had warfarin therapy initiated were available for analysis. Nine blood samples were collected from each subject at baseline, and at 1-5, 8, 15 and 29 days after warfarin initiation and assayed for factors II, VII, IX and X, and proteins C and S. Warfarin concentration-time data were not available. The coagulation proteins data were modelled in a stepwise manner using NONMEM® Version 7.2. In the first stage, each of the coagulation proteins was modelled independently using a kinetic-pharmacodynamic model. In the subsequent step, the six kinetic-pharmacodynamic models were combined into a single joint model. RESULTS: One patient was administered VK and was excluded from the analysis. Each kinetic-pharmacodynamic model consisted of two parts: (1) a common one-compartment pharmacokinetic model with first-order absorption and elimination for warfarin; and (2) an inhibitory E max model linked to a turnover model for coagulation proteins. In the joint model, an unexpected pharmacodynamic lag was identified and the estimated degradation half-life of VK-dependent coagulation proteins were in agreement with previously published values. The model provided an adequate fit to the observed data. CONCLUSION: The joint model represents the first work to quantify the influence of warfarin on all six VK-dependent coagulation proteins simultaneously. Future work will expand the model to predict the influence of exogenously administered VK on the time course of clotting factor concentrations after warfarin overdose and during perioperative warfarin reversal procedures.


Subject(s)
Anticoagulants/pharmacology , Blood Coagulation/drug effects , Models, Biological , Vitamin K/metabolism , Aged , Aged, 80 and over , Anticoagulants/pharmacokinetics , Atrial Fibrillation/drug therapy , Blood Coagulation Factors/drug effects , Blood Coagulation Factors/metabolism , Blood Proteins/drug effects , Blood Proteins/metabolism , Female , Half-Life , Humans , Male , Middle Aged , Nonlinear Dynamics , Warfarin/pharmacokinetics , Warfarin/pharmacology
17.
Leuk Lymphoma ; 57(11): 2568-74, 2016 11.
Article in English | MEDLINE | ID: mdl-27078747

ABSTRACT

Thrombosis is a well-recognized complication of asparaginase therapy for acute lymphoblastic leukemia (ALL), associated with the depletion of antithrombin (AT). Following a high incidence of thrombotic episodes during induction therapy for ALL in our tertiary referral center, we prospectively instituted a protocol of AT replacement. Forty-five consecutive adolescents and adults with ALL treated with asparaginase-containing phase I induction protocols were included in this observational study. Fifteen received standard therapy with no replacement; the subsequent 30 were managed with the protocol described. One or more low AT levels (<70 iu/dl) were recorded in 76% of patients in the cohort managed using the protocol, resulting them in receiving an AT replacement. There was a significant reduction in the incidence of thrombosis with this strategy (0/30 vs. 5/15, p < 0.001). We suggest that such a strategy should be studied in a prospective randomized sub-study within the context of a national ALL trial.


Subject(s)
Antineoplastic Agents/adverse effects , Antithrombins/therapeutic use , Asparaginase/adverse effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Thrombosis/drug therapy , Thrombosis/etiology , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/therapeutic use , Drug Therapy, Combination , Female , Humans , Male , Middle Aged , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Retrospective Studies , Thrombosis/prevention & control , Treatment Outcome , Young Adult
18.
Am J Clin Nutr ; 82(6): 1320-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16332666

ABSTRACT

BACKGROUND: Homocysteine is an independent risk factor for vascular disease and is associated with dementia in older people. Potential mechanisms include altered endothelial and hemostatic function. OBJECTIVE: We aimed to determine the effects of folic acid plus vitamin B-12, riboflavin, and vitamin B-6 on homocysteine and cognitive function. DESIGN: This was a factorial 2 x 2 x 2, randomized, placebo-controlled, double-blind study with 3 active treatments: folic acid (2.5 mg) plus vitamin B-12 (500 microg), vitamin B-6 (25 mg), and riboflavin (25 mg). We studied 185 patients aged >or=65 y with ischemic vascular disease. Outcome measures included plasma homocysteine, fibrinogen, and von Willebrand factor at 3 mo and cognitive change (determined with the use of the Letter Digit Coding Test and on the basis of the Telephone Interview of Cognitive Status) after 1 y. RESULTS: The mean (+/-SD) baseline plasma homocysteine concentration was 16.5 +/- 6.4 micromol/L. This value was 5.0 (95% CI: 3.8, 6.2) micromol/L lower in patients given folic acid plus vitamin B-12 than in patients not given folic acid plus vitamin B-12 but did not change significantly with vitamin B-6 or riboflavin treatment. Homocysteine lowering with folic acid plus vitamin B-12 had no significant effect, relative to the 2 other treatments, on fibrinogen, von Willebrand factor, or cognitive performance as measured by the Letter Digit Coding Test (mean change: -1; 95% CI: -2.3, 1.4) and the Telephone Interview of Cognitive Status (-0.7; 95% CI: -1.7, 0.4). CONCLUSION: Oral folic acid plus vitamin B-12 decreased homocysteine concentrations in elderly patients with vascular disease but was not associated with statistically significant beneficial effects on cognitive function over the short or medium term.


Subject(s)
Cognition/drug effects , Folic Acid/therapeutic use , Homocysteine/blood , Vascular Diseases/drug therapy , Vitamin B 12/therapeutic use , Vitamin B Complex/therapeutic use , Aged , Analysis of Variance , Double-Blind Method , Drug Synergism , Female , Fibrinogen/analysis , Folic Acid/blood , Humans , Male , Riboflavin/therapeutic use , Treatment Outcome , Vascular Diseases/blood , Vitamin B 12/blood , Vitamin B 6/therapeutic use , von Willebrand Factor/analysis
19.
Blood Coagul Fibrinolysis ; 25(5): 444-50, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24686101

ABSTRACT

Recombinant human antithrombin (rhAT; ATryn), isolated from the milk of transgenic goats, provides an alternative to human plasma-derived antithrombin (AT) concentrate for perioperative and peripartum prophylaxis of venous thromboembolism (VTE) in patients with hereditary AT deficiency. Optimized rhAT dosing algorithms and improved plasma AT monitoring protocol were used in an open-label, single-arm, multinational, pivotal safety and efficacy study that was conducted in patients with hereditary AT deficiency in perioperative and peripartum settings. Loading and maintenance doses were calculated on the basis of pretreatment AT activity levels. Specific dosing regimens were used for pregnant and surgical patients; rhAT was to be given for at least 3 days and for 14 days or less. The primary efficacy end point was the incidence of any thromboembolic event during rhAT therapy or within 7 days of rhAT discontinuation. Safety and AT activity levels were secondary end points. Six surgical and 12 pregnant patients were treated for a median of 3.2 days (range 0.9-14 days). With the optimized dosing regimens, a median of 1 dose adjustment (range 0-6 dose adjustments) was needed to maintain AT activity levels within 80-120% of normal. No confirmed VTEs occurred during treatment or in the subsequent 7 days. Overall, rhAT was well tolerated, but some bleeding complications occurred after rhAT discontinuation and anticoagulation reinstitution. No antibodies to rhAT or goat milk proteins were detected. Perioperative and peripartum prophylactic rhAT therapy in patients with hereditary AT deficiency is well tolerated and effective in preventing VTE.


Subject(s)
Anticoagulants/therapeutic use , Antithrombin III Deficiency/complications , Antithrombins/therapeutic use , Perioperative Period/adverse effects , Peripartum Period/blood , Venous Thromboembolism/drug therapy , Venous Thromboembolism/prevention & control , Adult , Anticoagulants/administration & dosage , Antithrombins/administration & dosage , Female , Humans , Male , Middle Aged , Pregnancy , Young Adult
20.
Thromb Haemost ; 109(1): 16-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23138324

ABSTRACT

Cardiovascular disease (CVD) mortality is reported to be decreased in haemophilia patients, but reports on the prevalence of CVD risk factors are conflicting. A cross-sectional assessment of CVD risk profiles was performed in a large cohort of haemophilia patients. Baseline data on CVD risk factors of 709 Dutch and UK haemophilia patients aged ≥30 years were analysed and compared with the general age-matched male population. CVD risk profiles were assessed using the QRISK®2-2011 and SCORE algorithms. Although QRISK® 2 was only validated in the UK, comparison with SCORE indicated similar properties of QRISK®2 in both Dutch and UK patients (correlation 0.86). Mean age was 49.8 years. Hypertension was more common in haemophilia patients than in the general population (49% vs. 40%), while the prevalences of obesity and hypercholesterolaemia were lower (15 vs. 20% and 44 vs. 68%, respectively), and those of diabetes and smoking were similar. The predicted 10-year QRISK®2 risk was significantly higher in haemophilia patients than in the general population (8.9 vs. 6.7%), indicating more unfavourable cardiovascular disease risk profiles. This increased risk became apparent after the age of 40 years. Our results indicate an increased prevalence of hypertension and overall more unfavourable CVD risk profiles in haemophilia patients compared with the general age-matched male population.


Subject(s)
Cardiovascular Diseases/epidemiology , Hemophilia A/epidemiology , Adult , Age Factors , Aged , Aged, 80 and over , Algorithms , Cardiovascular Diseases/diagnosis , Case-Control Studies , Cross-Sectional Studies , Diabetes Mellitus/epidemiology , Hemophilia A/diagnosis , Humans , Hypercholesterolemia/epidemiology , Hypertension/epidemiology , Linear Models , Male , Middle Aged , Multivariate Analysis , Netherlands/epidemiology , Obesity/epidemiology , Prevalence , Prognosis , Prospective Studies , Risk Assessment , Risk Factors , Smoking/adverse effects , Smoking/epidemiology , Time Factors , United Kingdom/epidemiology
SELECTION OF CITATIONS
SEARCH DETAIL