Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Haemophilia ; 24(4): 675-682, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28124445

RESUMEN

INTRODUCTION: Postinfusion ReFacto AF levels can be difficult to measure accurately due to discrepancies between one-stage and chromogenic FVIII assays. To overcome this, the use of the ReFacto AF laboratory standard (RAFLS) is recommended, but there are discordant reports regarding its usefulness. AIM: We investigated whether calibration with RAFLS and measurement of ReFacto AF levels are influenced by the choice of reagents and patient-specific factors in one-stage FVIII assays. METHODS: Calibration curves were generated with both the RAFLS and a plasma standard using different F8DPs and one-stage FVIII assay reagents. This selection of reagents was then used to determine FVIII levels in the plasma of patients repeatedly treated with ReFacto AF. Results were compared with those obtained with a chromogenic assay. RESULTS: F8DP devoid of von Willebrand factor (VWF) falsely increased the values of RAFLS pro-coagulant activity generated using the APTT reagent. The resulting RAFLS calibration curve underestimated ReFacto AF levels to be half of their true concentration. The use of RAFLS with F8DP containing VWF reduced the discrepancy observed between the one-stage and chromogenic FVIII assays. However, the mean difference between the two assays still varied up to 50% depending on the patient. CONCLUSIONS: The RAFLS is a suitable calibrator for one-stage FVIII assays carried out with F8DP containing VWF. However, calibration with the RAFLS does not avoid the effect of patient-specific variables that contribute to discrepancies between the measurements of ReFacto AF levels with one-stage and chromogenic FVIII assays.


Asunto(s)
Pruebas de Coagulación Sanguínea/normas , Factor VIII/genética , Factor VIII/farmacología , Eliminación de Secuencia , Calibración , Factor VIII/química , Humanos , Indicadores y Reactivos/química , Dominios Proteicos , Estándares de Referencia
2.
Ann Oncol ; 24(7): 1892-1899, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23553060

RESUMEN

BACKGROUND: Heparin has been used for years as a locking solution in totally implantable venous access devices. Normal saline (NS) might be a safe alternative for heparin. However, evidence of non-inferiority of NS versus heparin is lacking. PATIENTS AND METHODS: We randomly allocated 802 cancer patients with a newly inserted port either to heparin lock (300 U/3 ml) or to NS lock groups in a 1:1 assignment ratio. The primary outcome was the number of functional complications, which was defined as 'easy injection, impossible aspiration' at port access. Secondary outcomes included all functional problems and catheter-related bacteraemia. We hypothesised that NS locks do not cause more functional problems and catheter-related bacteraemia than heparin locks. Non-inferiority is established if the upper limit of the confidence interval (CI) for the relative risk of NS versus heparin is <1.4. RESULTS: Three hundred and eighty-two patients from the NS group and 383 from the heparin lock group were included in the analysis. The incidence rate of our primary outcome (easy injection, impossible aspiration) was 3.70% (95% CI 2.91%-4.69%) and 3.92% (95% CI 3.09%-4.96%) of accesses in the NS and heparin groups, respectively. The relative risk was 0.94% (95% CI 0.67%-1.32%). Catheter-related bloodstream infection was 0.03 per 1000 catheter days in the NS group and 0.10 per 1000 catheter days in the heparin group. CONCLUSION: NS is a safe and effective locking solution in implantable ports if combined with a strict protocol for device insertion and maintenance.


Asunto(s)
Cateterismo Venoso Central/métodos , Heparina/química , Neoplasias/tratamiento farmacológico , Cloruro de Sodio/química , Adolescente , Adulto , Anciano , Bacteriemia/etiología , Infecciones Relacionadas con Catéteres/etiología , Cateterismo Venoso Central/efectos adversos , Catéteres de Permanencia/microbiología , Femenino , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Soluciones , Adulto Joven
3.
Haemophilia ; 19(3): e133-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23374099

RESUMEN

The assessment of recombinant FVIII (rFVIII) activity (FVIII:C) in plasma of patients is dependent on the assay. Notably, a calibration with a product-specific laboratory standard is recommended when measuring Refacto-AF(R) activity in plasma with a one-stage assay. The objective of this study was to facilitate the measurement of rFVIII, taking into account the recent demonstration that a calibration curve does not have to be included in each run. FVIII:C was measured in patients' samples after infusion of different types of rFVIII with a one-stage and a chromogenic assay calibrated either with pooled normal plasma or a product-specific laboratory standard. Results obtained with the one-stage coagulation assay were compared with these provided by a chromogenic assay. We confirmed that a calibration curve can be used for a prolonged period of time without loss of precision and accuracy. In such conditions, a stable relation between the calibration curves generated with a product-specific laboratory standard and plasma can be established. In patients' plasma, Refacto-AF levels measured with a one-stage FVIII assay calibrated with plasma or a product-specific laboratory standard diverged from -58% to -17% and from -25% to +18%, respectively, from the activity determined with a chromogenic substrate assay. By comparison, FVIII:C levels of full-length rFVIII measured with the one-stage assay calibrated with plasma were 6-49% lower than with the chromogenic assay. In a monocentric setting, the long-term stability of the calibration curves allows the implementation of a practical and cost-effective approach to determine rFVIII:C levels.


Asunto(s)
Pruebas de Enzimas , Factor VIII/análisis , Proteínas Recombinantes/análisis , Calibración , Compuestos Cromogénicos/química , Compuestos Cromogénicos/metabolismo , Pruebas de Enzimas/normas , Factor VIII/biosíntesis , Factor VIII/normas , Hemofilia A/sangre , Humanos , Laboratorios , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/normas , Estándares de Referencia
4.
Phys Med Biol ; 66(17)2021 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-33571977

RESUMEN

To further improve the understanding ofin vitrobiological effects of incorporated radionuclides, it is essential to accurately determine cellular absorbed doses. In the case ofßemitters, the cross-dose is a major contribution, and can involve up to millions of cells. Realistic and efficient computational models are needed for that purpose. Conventionally, distances between each cell are calculated and the related dose contributions are cumulated to get the total cross-dose (standard method). In this work, we developed a novel approach for the calculation of the cross-absorbed dose, based on the use of the radial distribution function (rdf)) that describes the spatial properties of the cellular model considered. The dynamic molecular tool LAMMPS was used to create 3D cellular models and computerdfsfor various conditions of cell density, volume size, and configuration type (lattice and randomized geometry). The novel method is suitable for any radionuclide of nuclear medicine. Here, the model was applied for the labeling of cells with18F-FDG used for PET imaging, and first validated by comparison with other reference methods. MeanScrossvalues calculated with the novel approach versus the standard method agreed very well (relative differences less that 0.1%). Implementation of therdf-based approach with LAMMPS allowed to achieved results considerably faster than with the standard method, the computing time decreasing from hours to seconds for 106cells. Therdf-based approach was also faster and easier to accommodate more complex cellular models than the standard and other published methods. Finally, a comparative study of the meanScrossfor different types of configuration was carried out, as a function of the cell density and the volume size, allowing to better understand the impact of the configuration on the cross-absorbed dose.


Asunto(s)
Análisis de la Célula Individual , Fluorodesoxiglucosa F18 , Método de Montecarlo , Medicina Nuclear , Tomografía de Emisión de Positrones , Radioisótopos
5.
Haemophilia ; 16(102): 16-9, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20536982

RESUMEN

The mechanism of action of antibodies inhibiting partially factor VIII (FVIII) activity (type II inhibitor) is still poorly understood. We produced an unusual type II monoclonal antibody, called LE2E9, derived from a patient with mild haemophilia A. The antibody displayed several unexpected structural and functional properties such as glycosylation in the variable region, binding to the FVIII C1 domain, inhibition of maximum 80-90% FVIII activity when in excess over FVIII, and prevention of FVIII binding to von Willebrand factor (VWF). Those unusual characteristics of the antibody prompted multidisciplinary studies to determine its mechanism of action and the role of the FVIII C1 domain. Enzymatic deglycosylation and site-directed mutagenesis indicated that the oligosaccharides do not determine the affinity of the antibody but enhanced its FVIII neutralizing activity. Modification of glycosylation in the variable region of antibodies therefore contributes to the diversity of FVIII type II inhibition and provides a novel strategy with which to modulate the functional activity of antibodies. Investigation of the FVIII C1 domain function led to identification of mutations located in that domain and impairing FVIII binding to VWF as a common cause of mild/moderate haemophilia A. Finally, the cloning of human monoclonal antibodies inhibiting partially FVIII activity opened the way to evaluate such antibodies as a novel type of anticoagulant drug. This concept was validated in animal models of thrombosis. Those studies are expected to have a significant impact on the optimization of treatments for patients with inhibitor as well as for patients with thrombophilia.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticoagulantes/farmacología , Coagulación Sanguínea/inmunología , Factor VIII/inmunología , Fibrinolíticos/farmacología , Glicosilación , Hemofilia A/tratamiento farmacológico , Trombosis/prevención & control , Animales , Anticuerpos Monoclonales/inmunología , Anticoagulantes/inmunología , Factor VIII/química , Fibrinolíticos/inmunología , Hemofilia A/complicaciones , Humanos , Ratones
6.
Haemophilia ; 16 Suppl 5: 100-6, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20590864

RESUMEN

SUMMARY: Despite major advances in diagnosis and treatment, the management of patients with mild haemophilia (MH) remains a major challenge. Mild haemophilia is defined by factor levels between 0.05 and 0.40 IU mL(-1). The bleeding associated with mild haemophilia is most frequently episodic, occurring during surgery or following trauma. Spontaneous bleeding is rare. Diagnosis is sometimes delayed because of insensitivity of screening clotting assays or discrepancies in factor VIII activity as measured by different assays. The treatment of choice in mild haemophilia A is desmopressin, which typically induces a 2-6-fold increase of factor VIII over baseline. However, desmopressin has its limitations in this setting such as the occurrence of tachyphylaxis and failure to respond in an undetermined proportion of patients. Factors underlying poor biological response or magnitude of response to desmopressin are incompletely understood. Inhibitor development in mild haemophilia is particularly distressing. This complication arises at an older age in this patient group because of infrequent need for factor VIII replacement. Inhibitors in mild haemophilia patients often cross-react with endogenous factor VIII resulting in severe spontaneous bleeding frequently in a postoperative setting. Intensive perioperative use of factor VIII and some specific mutations induce a particularly high risk for inhibitor development, but risk factors are incompletely understood. For reasons of the older age of the patients, treatment of bleeding with bypassing agents may cause major thrombotic complications. Data on therapeutic options for inhibitor eradication in patients with mild haemophilia are particularly scarce. With increased life-expectancy for all haemophilia patients, the group of elderly patients with mild haemophilia requiring major surgery will further increase. Prevention of inhibitors, particularly in this patient group, should be a major topic of interest in both clinic and research.


Asunto(s)
Factores de Coagulación Sanguínea/análisis , Pruebas de Coagulación Sanguínea/métodos , Desamino Arginina Vasopresina/uso terapéutico , Hemofilia A/diagnóstico , Hemofilia A/terapia , Hemostáticos/uso terapéutico , Inhibidores de Factor de Coagulación Sanguínea/sangre , Hemofilia A/sangre , Humanos
7.
Haemophilia ; 14 Suppl 3: 138-46, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18510534

RESUMEN

In mild/moderate haemophilia A (MHA) patients, many factor VIII (FVIII) gene defects, mainly missense mutations, have been identified and greatly improved the understanding of the structure and function of FVIII molecule. Characterization of the molecular mechanisms involved in MHA has helped to identify regions critical for proper FVIII biosynthesis, thrombin activation, intramolecular stability as well as binding regions for important intermolecular interactions with von Willebrand factor, factor IXa and the phospholipid surface. Some missense mutations were also recognized as contributing factors to inhibitor development in MHA, in parallel to acquired factors such as inflammatory state or intensity of treatment. Treatment of MHA with inhibitor patients raises questions on how best to stop or prevent bleeding episodes and eradicate the inhibitor. Longitudinal data collection is currently being conducted in France and Belgium to enhance our knowledge in this field and to further help make treatment decision. The description of mutations in MHA finally contributed to the identification of epitopes involved in the immune response to FVIII. In some patients, the epitope specificity of inhibitor antibodies recognizing normal exogenous FVIII alone and not patient ('self') FVIII was described. This distinguished epitope specificity could also be demonstrated at the T-cell clonal level. One might expect that these molecular studies will have a major impact on development of new FVIII products in the future.


Asunto(s)
Epítopos/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Tolerancia Inmunológica/inmunología , Mutación Missense/inmunología , Anticuerpos/inmunología , Desamino Arginina Vasopresina/uso terapéutico , Factor IXa/inmunología , Factor VIII/genética , Genotipo , Hemofilia A/tratamiento farmacológico , Hemofilia A/genética , Hemorragia/tratamiento farmacológico , Hemostáticos/uso terapéutico , Humanos , Mutación Missense/genética , Trombina/inmunología
8.
Haemophilia ; 14(2): 295-302, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18081826

RESUMEN

A retrospective chart review of 11 subjects with severe haemophilia A and high-titre inhibitors who received a von Willebrand factor-containing FVIII concentrate (VWF/FVIII) for immune tolerance induction (ITI) was accompanied by B cell inhibitor epitope mapping during 10/11 treatment courses. ITI was successful or partially successful in all seven subjects who received VWF/FVIII for initial ITI, and failed in all four subjects whose ITI with this product was initiated following treatment failure using recombinant factor VIII. Variables including age at inhibitor development and age at ITI initiation, interval between inhibitor detection and ITI initiation, titre at start of ITI, and peak historical titres prior to and during ITI were not statistically significant outcome predictors in this cohort. However, the B cell epitope specificity in all four successful and in one of two partially successful ITI subjects for whom information was available included the C2 and excluded the A2 domains. Conversely, FVIII B cell epitopes in one partially successful ITI and in all three failed ITI subjects for whom data were available mapped to both the C2 and the A2 domains. The FVIII B cell epitope profile was associated with ITI outcome in this VWF/FVIII-treated cohort. Its role in predicting ITI outcome and guiding choice of FVIII product for ITI requires further study.


Asunto(s)
Autoanticuerpos/sangre , Factor VIII/inmunología , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Hemostáticos/uso terapéutico , Factor de von Willebrand/uso terapéutico , Adolescente , Adulto , Linfocitos B/inmunología , Niño , Preescolar , Combinación de Medicamentos , Mapeo Epitopo , Epítopos/análisis , Factor VIII/uso terapéutico , Genotipo , Hemofilia A/genética , Humanos , Tolerancia Inmunológica , Lactante , Recién Nacido , Estudios Retrospectivos , Resultado del Tratamiento
9.
Int J Lab Hematol ; 40 Suppl 1: 21-29, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29741261

RESUMEN

Several recombinant factor VIII and factor IX concentrates with extended half-life (EHL) have recently been validated by clinical studies. The availability of these novel concentrates is expected to significantly facilitate the treatment of patients with hemophilia A and B. However, the modification applied to these molecules has introduced variations in their activity measurement in routine coagulation assays. Depending on the assays, underestimations of up to 10-fold or overestimations of up to approximately 30-fold in the measurements of the recovery have been reported in some factor concentrates. Such biases in monitoring may lead to major under- or overtreatment, as well as unnecessary searching for inhibitor antibodies. In this review, we discuss the guidelines and recommendations that allow the selection of optimal strategies to monitor patients treated with these novel factor concentrates. Based on the specificities of the assays and on local regulations, different chromogenic substrate assays in addition to one-stage clotting assays may be validated to allow the accurate measurement of all novel products. An efficient communication between the clinical laboratory and the clinicians is essential to ensure that the appropriate assays are carried out in laboratories and that the clinicians correctly evaluate the data. Further laboratory and clinical studies are still required for the optimization of the laboratory assays that can be used in the measurement of novel factor VIII and factor IX concentrates with EHL.


Asunto(s)
Técnicas de Laboratorio Clínico/tendencias , Factor IX/uso terapéutico , Factor VIII/uso terapéutico , Técnicas de Laboratorio Clínico/métodos , Monitoreo de Drogas/métodos , Factor IX/farmacocinética , Factor VIII/farmacocinética , Semivida , Hemofilia A/tratamiento farmacológico , Hemofilia B/tratamiento farmacológico , Humanos
10.
Int J Lab Hematol ; 40(4): 442-447, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29573340

RESUMEN

INTRODUCTION: The direct thrombin inhibitor dabigatran interferes with thrombophilia screening and with the diagnosis of hemostasis disorders that develop during treatment with the anticoagulant. In vitro addition of idarucizumab, a humanized antibody fragment that binds dabigatran, to plasma samples containing dabigatran fully neutralizes the drug. This study was carried out to determine whether binding of dabigatran on selected insoluble commercial adsorbent material, DOAC-STOPR , was as efficient as idarucizumab to neutralize the anticoagulant activity of the drug in vitro. METHODS: Coagulation assays sensitive to dabigatran were carried out with patient and control plasma samples spiked with dabigatran and supplemented with idarucizumab or incubated with adsorbent material. RESULTS: In samples containing upto 10 000 ng/mL dabigatran, the adsorption procedure was at least as efficient as the addition of idarucizumab to neutralize the activity of the anticoagulant drug. Neither the adsorption procedure nor the addition of idarucizumab did impair routine coagulation assays carried out with plasma devoid of dabigatran, such as the activated partial thromboplastin time, prothrombin time, fibrinogen Clauss, and the thrombophilia screening assays used to detect antiphospholipid antibodies or activated protein C resistance. In addition, the adsorption procedure did not interfere with the detection of lupus anticoagulant samples. CONCLUSIONS: Adsorption of dabigatran in plasma samples containing the drug neutralizes its activity as efficiently as the addition of idarucizumab. This method allows the evaluation of thrombophilia markers without interruption of anticoagulation therapy or the detection of hemostasis disorders in patients treated with the drug.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Pruebas de Coagulación Sanguínea/normas , Dabigatrán/aislamiento & purificación , Trombofilia/diagnóstico , Adsorción , Anticoagulantes/uso terapéutico , Interacciones Farmacológicas , Humanos
11.
J Clin Invest ; 85(4): 1024-35, 1990 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-2318962

RESUMEN

Antigen-antibody complexes were made from allergens of the common house dust mite, Dermatophagoides pteronyssinus (Dpt) and an excess of purified autologous specific antibodies. These complexes have been used to treat Dpt-hypersensitive patients who suffered from chronic bronchial asthma. Clinical symptoms and medication intake were followed by filling in diary cards. Peak expiratory flow, measured four times a day, was also followed. Intradermal skin tests and bronchial challenge tests were performed with allergen together with an evaluation of nonspecific bronchial reactivity. Specific IgE and IgG antibodies were assayed after separation from the bulk of serum immunoglobulins by immunoadsorption. The study was carried out over two years according to a double-blind protocol. Intradermal inoculation of antigen-antibody complexes resulted in a marked reduction of both clinical and medication scores. No systemic side-effects were observed and only mild wheal and flare reactions were noted at the injection site. The treatment showed a drastic reduction of specific skin and bronchial reactivities with only marginal effects on nonspecific bronchial reactivity. Concentrations of specific IgE antibodies decreased significantly during the first weeks of treatment and remained at these lower values throughout the study. Specific IgG antibodies actually decreased in the majority of treated patients. The total amount of allergen used in this study was less than 1% of the amount currently used for conventional hyposensitization with the same allergen. These findings show that antigen-antibody complex inoculation is an efficient and safe means of treating allergic bronchial asthma and that the mechanism of action is likely to differ from conventional hyposensitization.


Asunto(s)
Alérgenos/inmunología , Complejo Antígeno-Anticuerpo/inmunología , Asma/terapia , Hipersensibilidad/terapia , Inmunoterapia/métodos , Ácaros/inmunología , Adolescente , Adulto , Animales , Asma/inmunología , Asma/fisiopatología , Pruebas de Provocación Bronquial , Femenino , Humanos , Inmunoglobulina E/análisis , Inmunoglobulina G/análisis , Masculino , Flujo Espiratorio Máximo , Persona de Mediana Edad , Cooperación del Paciente , Pruebas Cutáneas
12.
Int J Lab Hematol ; 39(3): 301-307, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28318107

RESUMEN

INTRODUCTION: Thrombin time (TT) tests are useful for diagnosing coagulation disorders involving abnormal fibrinogen but do not allow us to distinguish between qualitative and quantitative defects. However, with the widening availability of optical coagulation automates, more information about the coagulation process is becoming increasingly accessible. METHODS: In this study, we compared the coagulation curves of TT tests carried out with plasma from healthy donors with those from patients with acquired low Clauss fibrinogen levels or with dysfibrinogenemia caused by a heterozygous point mutation in the fibrinogen γ-chain that results in a p.Arg301(275)Cys substitution. The functional fibrinogen levels of these three groups of samples were also measured with the Clauss method, and their fibrinogen protein levels were determined by ELISA. RESULTS: Our data indicate that the amplitude and maximal velocity of coagulation curves from plasma samples from FGG p.Arg301(275)Cys dysfibrinogenemic patients were comparable to those from plasma samples with fibrinogen in the normal range, whereas the amplitude of coagulation curves from patients with acquired low fibrinogen levels was lower. CONCLUSIONS: Examination of the amplitude of coagulation curves generated during TT tests may provide additional information to enable the differential diagnoses of diseases following a low fibrinogen measurement by the Clauss method.


Asunto(s)
Afibrinogenemia/sangre , Afibrinogenemia/genética , Fibrinógenos Anormales/genética , Fibrinógenos Anormales/metabolismo , Mutación Missense , Sustitución de Aminoácidos , Femenino , Humanos , Masculino , Tiempo de Trombina/métodos
13.
Int J Lab Hematol ; 39(4): 402-408, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28304137

RESUMEN

INTRODUCTION: The Belgian national External Quality Assessment Scheme performed a survey to assess the effect of the direct oral anticoagulant apixaban on the coagulation assays prothrombin time (PT), activated partial thromboplastin time (aPTT), fibrinogen and antithrombin as performed with a large number of reagent/instrument combinations. METHODS: Four lyophilized plasma samples spiked with apixaban (0, 41, 94 and 225 ng/mL) were sent to the 195 Belgian and Luxembourg clinical laboratories performing coagulation testing. RESULTS: PT and aPTT were barely influenced at the concentrations tested. At 225 ng/mL apixaban, PT and aPTT clotting times were only 1.15 times longer than at 0 ng/mL. Among PT reagents, RecombiPlasTin 2G® showed a slightly higher sensitivity with 225 ng/mL apixaban prolonging the PT clotting time 1.3-fold. Among aPTT reagents, there was no appreciable difference in sensitivity. Fibrinogen results were unaffected by the presence of apixaban, but antithrombin activity was considerably overestimated when measured with a FXa-based assay. At 225 ng/mL apixaban, the median percentage increase in antithrombin level was 31% when measured with the Liquid Antithrombin® reagent and 44% with the Innovance Antithrombin® reagent. CONCLUSION: Our data provide clinical laboratories with useful information on the impact of apixaban on their routine coagulation assays.


Asunto(s)
Pruebas de Coagulación Sanguínea/normas , Coagulación Sanguínea/efectos de los fármacos , Inhibidores del Factor Xa/farmacología , Pirazoles/farmacología , Piridonas/farmacología , Antitrombinas/sangre , Bélgica , Pruebas de Coagulación Sanguínea/métodos , Monitoreo de Drogas , Inhibidores del Factor Xa/uso terapéutico , Fibrinógeno/biosíntesis , Humanos , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , Pirazoles/uso terapéutico , Piridonas/uso terapéutico , Garantía de la Calidad de Atención de Salud
14.
J Thromb Haemost ; 4(5): 1047-55, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16689758

RESUMEN

BACKGROUND: N-glycosylation occurs in the variable region of about 10% of antibodies but the role of carbohydrate at this location is still poorly understood. OBJECTIVES: We investigated the function of N-glycosylation in the variable region of the heavy chain of a human monoclonal antibody, mAb-LE2E9, that partially inhibits factor VIII (FVIII) activity during coagulation. METHODS AND RESULTS: Enzymatic deglycosylation indicated that the oligosaccharides do not determine the affinity of the antibody but enhance its FVIII neutralizing activity. A mutant antibody lacking the N-glycosylation site in the variable region of the heavy chain inhibited FVIII activity by up to 40%, while inhibition by the native antibody was 80%. To evaluate the physiological effect of such a FVIII inhibition, we investigated the ability of the mutant antibody devoid of N-glycosylation in the variable region to prevent thrombosis in mice with a strong prothombotic phenotype resulting from a type II deficiency mutation in the heparin binding site of antithrombin. Despite its moderate inhibition of FVIII activity, the mutant antibody significantly prevented thrombosis in treated animals. We also carried out glycan analysis of native and mutant antibodies. CONCLUSIONS: Modification of glycosylation in the variable region of antibodies contributes to the diversity of FVIII type II inhibition possibly by steric hindrance of the active site of FVIII by glycans, and may provide a novel strategy to modulate the functional activity of therapeutic antibodies.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Anticoagulantes/farmacología , Factor VIII/inmunología , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Anticoagulantes/química , Anticoagulantes/inmunología , Secuencia de Bases , Células CHO , Cromatografía en Gel , Cricetinae , Cartilla de ADN , Glicosilación , Humanos , Resonancia por Plasmón de Superficie
15.
J Thromb Haemost ; 13(11): 2087-92, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26347330

RESUMEN

BACKGROUND: The anticoagulant effect of dabigatran can be approximated by its prolongation of routine coagulation assays. Consequently, dabigatran also interferes with thrombophilia screening or with diagnosing hemostasis disorders that have developed after the initiation of anticoagulant treatment, such as vitamin K deficiency or acquired hemophilia A. OBJECTIVES: This study was carried out to determine whether idarucizumab, a humanized antibody fragment that binds dabigatran, could fully neutralize dabigatran in routine diagnostic coagulation assays conducted in vitro, thereby preventing false-positive or false-negative diagnostic readouts. METHODS: Preliminary experiments identified coagulation assays that were sensitive to dabigatran, and identified a concentration of idarucizumab that neutralized the effects of dabigatran. These assays were then carried out with patient and control plasma samples spiked with dabigatran, with or without a molar excess of idarucizumab. RESULTS: Dabigatran altered the prothrombin time, activated partial thromboplastin time and thrombin time, and the measurement of intrinsic and extrinsic factor levels. Screening and confirmation tests used for lupus anticoagulant detection were prolonged by dabigatran, falsely suggesting the presence of lupus anticoagulant. Conversely, the addition of dabigatran falsely corrected an abnormal activated protein C resistance ratio. Addition of idarucizumab completely normalized these measurements, and allowed the correct identification of normal and abnormal samples with these assays. CONCLUSIONS: In vitro addition of idarucizumab to plasma samples containing dabigatran fully neutralizes the drug, and facilitates the use of routine coagulation assays to allow the diagnosis of hemostasis disorders that may be concurrently present in patients taking dabigatran.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Neutralizantes/farmacología , Antitrombinas/sangre , Trastornos de la Coagulación Sanguínea/sangre , Pruebas de Coagulación Sanguínea , Dabigatrán/sangre , Resistencia a la Proteína C Activada/sangre , Anticuerpos Monoclonales Humanizados/inmunología , Anticuerpos Neutralizantes/inmunología , Especificidad de Anticuerpos , Antitrombinas/inmunología , Antitrombinas/farmacología , Trastornos de la Coagulación Sanguínea/tratamiento farmacológico , Dabigatrán/antagonistas & inhibidores , Dabigatrán/inmunología , Dabigatrán/farmacología , Relación Dosis-Respuesta Inmunológica , Reacciones Falso Negativas , Reacciones Falso Positivas , Hemofilia A/sangre , Humanos , Inhibidor de Coagulación del Lupus/sangre
16.
Curr Med Chem ; 11(17): 2291-6, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15379713

RESUMEN

Current treatments for preventing thrombotic diseases are associated with a significant risk of bleeding. Improved anticoagulant agents are therefore still required. The specificity and pharmacokinetics properties of monoclonal antibodies to coagulation factors allow novel anticoagulation approaches. Treatment with human antibodies or humanized mouse monoclonal antibodies should avoid unacceptable side effects due to immune response to the drug. Such antibodies were developed against three coagulation factor: Tissue factor (TF), Factor IX (FIX) and Factor VIII (FVIII). A fully humanized antibody was successfully derived from a mouse monoclonal antibodies to TF. In vivo studies with monoclonal antibodies to TF demonstrated efficient antithrombotic activity. Anti-TF antibodies may also prove useful in cardiovascular disorders and cancer, given the role of TF in these diseases. Mouse and human monoclonal antibodies to FIX were also efficient to prevent thrombosis in animal models of venous and arterial thrombosis and in stroke. A humanized anti-FIX antibody was tested in phase I study in healthy volunteers. The pharmacokinetics of the antibody were determined by the rapid formation of stable complexes with newly synthesised FIX. Human anti-FVIII antibodies inhibiting only partially FVIII activity were recently described. Investigations in mice have established that treatment with such anti-FVIII antibodies is efficient to prevent deep vein thrombosis. Given the low concentration of FVIII in plasma and the long half-life of antibody, treatment with anti-FVIII antibody could be very convenient, allowing one administration every month. Altogether, monoclonal antibodies to coagulation factor appear as promising novel antithrombotic drugs.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Anticoagulantes/uso terapéutico , Factores de Coagulación Sanguínea/inmunología , Trombina/antagonistas & inhibidores , Trombosis/prevención & control , Animales , Sitios de Unión , Coagulación Sanguínea/efectos de los fármacos , Enfermedades Cardiovasculares/tratamiento farmacológico , Modelos Animales de Enfermedad , Humanos , Neoplasias/tratamiento farmacológico , Trombina/química , Trombosis/tratamiento farmacológico
17.
J Thromb Haemost ; 2(1): 77-84, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14717970

RESUMEN

Venous thromboembolic disease is a major cause of morbidity and mortality, necessitating antithrombotic therapy. A human monoclonal anti-factor (F)VIII antibody, LCL-mAb-LE2E9, produced by a lymphoblastoid cell line derived from a hemophilia A patient with inhibitor to wild-type but not mutant self FVIII, was previously reported to achieve efficient inhibition of thrombosis in an experimental vena cava thrombosis model in mice. Here, the antithrombotic efficacy of a recombinant DNA-derived version of this anti-FVIII antibody (rec-mAb-LE2E9) was tested in mice which carry a type II heparin binding site antithrombin deficiency mutation and display spontaneous chronic thrombosis in several sites including the penile vein of sexually active males. The recombinant anti-FVIII antibody (100 microg, repeated after 3 days) prevented thrombotic priapism in all treated males, whereas all control animals treated with saline (group of four animals) developed priapism within 6 days after mating (P < 0.05 for treated vs. saline). The rec-mAb-LE2E9 and the original LCL-mAb-LE2E9 were equally effective (five and seven males/group, respectively). These results confirm that FVIII inhibition represents a potent antithrombotic strategy, and show that both LCL-mAb-LE2E9 and rec-mAb-LE2E9 efficiently prevent thrombosis in a physiological model representative of thrombosis in patients with a severe prothrombotic risk.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Deficiencia de Antitrombina III/tratamiento farmacológico , Factor VIII/antagonistas & inhibidores , Fibrinolíticos/farmacología , Trombosis/prevención & control , Animales , Anticuerpos Monoclonales/farmacocinética , Antitrombina III/genética , Deficiencia de Antitrombina III/sangre , Deficiencia de Antitrombina III/genética , Sitios de Unión/genética , Factor VIII/inmunología , Femenino , Fibrinolíticos/farmacocinética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Priapismo/etiología , Priapismo/patología , Priapismo/prevención & control , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Trombosis/etiología , Trombosis/patología
18.
J Thromb Haemost ; 1(3): 456-63, 2003 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-12871450

RESUMEN

Mutations responsible for mild/moderate hemophilia A were extensively characterized over the last 15 years and more than 200 mutations have been identified. However, most of the molecular mechanisms responsible for the reduced factor (F)VIII levels in patients' plasma were determined only recently. Recent progresses in the study of the FVIII molecule three-dimensional structure provided a major insight for understanding molecular events leading to mild/moderate hemophilia A. This allowed prediction of mutations impairing FVIII folding and intracellular processing, which result in reduced FVIII secretion. Mutations potentially slowing down FVIII activation by thrombin were also identified. A number of mutations were also predicted to result in altered stability of activated FVIII. Biochemical analyses allowed identification of mutations reducing FVIII production. Mutations impairing FVIII stability in plasma, by reducing FVIII binding to von Willebrand factor (VWF) were also characterized. Defects in FVIII activity, notably slow activation by thrombin, or abnormal interaction with FIXa, were also recently demonstrated. Biochemical analysis of FVIII variants provided information regarding the structure/function relationship of the FVIII molecule and validated predictions of the three-dimensional structure of the molecule. These observations also contributed to explain the discrepant activities recorded for some FVIII variants using different types of FVIII assays. Altogether, the study of the biochemical properties of FVIII variants and the evaluation of the effects of mutations in three-dimensional models of FVIII identified molecular mechanisms potentially explaining reduced FVIII levels for a majority of patients with mild/moderate hemophilia A. It is expected that these studies will improve diagnosis and treatment of this disease.


Asunto(s)
Hemofilia A/etiología , Factor IXa/metabolismo , Factor VIII/química , Factor VIII/genética , Factor VIII/metabolismo , Hemofilia A/genética , Humanos , Mutación , Trombina/biosíntesis , Factor de von Willebrand/metabolismo
19.
Thromb Haemost ; 77(1): 80-6, 1997 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9031454

RESUMEN

Antibodies to factor VIII (inhibitors) are usually produced at the beginning of treatment with factor VIII and are rare in multitransfused patients. Such antibodies are deemed to be patient-related, as supported by the description of a number of associated risk factors. However, a second category of inhibitors has recently been identified, namely antibodies occurring in multitransfused patients as a result of exposure to a particular factor VIII concentrate. A first outbreak of product-related inhibitors was recently described. The present paper describes the second well-documented occurrence of such inhibitors. Eight out of 140 multitransfused patients with severe haemophilia A developed an inhibitor to factor VIII shortly after changing treatment to a double-virus inactivated plasma-derived factor VIII concentrate. In addition to solvent-detergent treatment, this concentrate was pasteurised at 63 degrees C for 10 hours. Exposure to the pasteurised product before inhibitor detection ranged from 9 to 45 days. Inhibitor titers varied between 2.2 and 60 Bethesda Units and recovery of transfused factor VIII ranged from 0.21 to 0.68 (expressed as i.u./dl factor VIII rise per i.u./kg administered). In contrast to usual inhibitors in haemophilia A patients, these product-related inhibitors showed complex inhibition kinetics. They were found specific for the factor VIII light chain. The inhibitors gradually declined when exposure to the pasteurised product was stopped, despite further treatment with other factor VIII concentrates. The present data stress the importance of carefully monitored clinical studies, both in previously treated and previously untreated patients, before introduction of a new or modified clotting factor concentrate.


Asunto(s)
Anticuerpos/sangre , Factor VIII/inmunología , Hemofilia A/inmunología , Adolescente , Adulto , Factor VIII/aislamiento & purificación , Factor VIII/uso terapéutico , Femenino , Hemofilia A/sangre , Humanos , Masculino , Virus
20.
Thromb Haemost ; 82(1): 40-5, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10456452

RESUMEN

A mild haemophilia A patient (LE) with an Arg2150His mutation in the C1 domain of the factor VIII (FVIII) light chain was shown to have anti-FVIII antibodies inhibiting wild type but not self FVIII. Polyclonal anti-FVIII antibodies of this patient were purified by affinity adsorption using recombinant FVIII (rFVIII) and/or plasma-derived FVIII-von Willebrand factor (vWF) complexes. A distinct population of antibodies was obtained that bound to FVIII-vWF complexes but not to rFVIII, indicating that an epitope was created by the association of FVIII to vWF. Such antibodies belonged to the IgG2 isotype, but the FVIII epitopes to which they bind could not be mapped with precision due to vWF dependency. Depletion experiments showed that anti-FVIII antibodies recognising FVIII-vWF complex also distinguished wildtype from mutated self FVIII, indicating that the Arg2150His mutation alters the B cell epitope formed by the association of FVIII to vWF. To determine whether the Arg2150His substitution also alters the formation of the FVIII-vWF complex, the interaction between mutated or normal FVIII with vWF was evaluated in plasma. The dissociation rate of mutated FVIII from vWF was found to be significantly increased. The presence of an Arg2150His mutation therefore results in the disappearance of a FVIII B cell epitope generated by the association of FVIII with vWF. Patients carrying such an Arg2150His mutation and receiving infusion of wild-type FVIII may therefore be at risk of developing inhibitors to allogeneic FVIII only.


Asunto(s)
Anticuerpos/inmunología , Epítopos/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Factor de von Willebrand/inmunología , Especificidad de Anticuerpos , Mapeo Epitopo , Factor VIII/metabolismo , Hemofilia A/tratamiento farmacológico , Humanos , Masculino , Unión Proteica , Factor de von Willebrand/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA