Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Blood Adv ; 8(6): 1392-1404, 2024 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-38286442

RESUMEN

ABSTRACT: Congenital fibrinogen deficiency (CFD) is a rare bleeding disorder caused by mutations in FGA, FGB, and FGG. We sought to comprehensively characterize patients with CFD using PRO-RBDD (Prospective Rare Bleeding Disorders Database). Clinical phenotypes, laboratory, and genetic features were investigated using retrospective data from the PRO-RBDD. Patients were classified from asymptomatic to grade 3 based on their bleeding severity. In addition, FGA, FGB, and FGG were sequenced to find causative variants. A total of 166 CFD cases from 16 countries were included, of whom 123 (30 afibrinogenemia, 33 hypofibrinogenemia, 55 dysfibrinogenemia, and 5 hypodysfibrinogenemia) were well characterized. Considering the previously established factor activity and antigen level thresholds, bleeding severity was correctly identified in 58% of the cases. The rates of thrombotic events among afibrinogenemic and hypofibrinogenemic patients were relatively similar (11% and 10%, respectively) and surprisingly higher than in dysfibrinogenemic cases. The rate of spontaneous abortions among 68 pregnancies was 31%, including 86% in dysfibrinogenemic women and 14% with hypofibrinogenemia. Eighty-six patients received treatment (69 on-demand and/or 17 on prophylaxis), with fibrinogen concentrates being the most frequently used product. Genetic analysis was available for 91 cases and 41 distinct variants were identified. Hotspot variants (FGG, p.Arg301Cys/His and FGA, p.Arg35Cys/His) were present in 51% of dysfibrinogenemia. Obstetric complications were commonly observed in dysfibrinogenemia. This large multicenter study provided a comprehensive insight into the clinical, laboratory, and genetic history of patients with CFDs. We conclude that bleeding severity grades were in agreement with the established factor activity threshold in nearly half of the cases with quantitative defects.


Asunto(s)
Afibrinogenemia , Hemostáticos , Humanos , Femenino , Fibrinógeno/genética , Afibrinogenemia/epidemiología , Afibrinogenemia/genética , Afibrinogenemia/complicaciones , Estudios Prospectivos , Estudios Retrospectivos , Hemorragia/genética
2.
Haemophilia ; 30(2): 306-319, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38239180

RESUMEN

AIM: For people with haemophilia A (PwHA), bleeding in the joints leads to joint damage and haemophilia-related arthropathy, impacting range of motion and life expectancy. Existing guidelines for managing haemophilia A support healthcare professionals (HCPs) and PwHA in their efforts to preserve joint health. However, such guidance should be reviewed, considering emerging evidence and consensus as presented in this manuscript. METHODS: Fifteen HCPs experienced in the management of PwHA in the UK participated in a three-round Delphi panel. Consensus was defined at ≥70% of panellists agreeing or disagreeing for Likert-scale questions, and ≥70% selecting the same option for multiple- or single-choice questions. Questions not reaching consensus were revised for the next round. RESULTS: 26.8% (11/41), 44.8% (13/29) and 93.3% (14/15) of statements reached consensus in Rounds 1, 2 and 3, respectively. HCPs agreed that prophylaxis should be offered to patients with a baseline factor VIII (FVIII) level of ≤5 IU/dL and that, where there is no treatment burden, the aim of prophylaxis should be to achieve a trough FVIII level ≥15 IU/dL and maintain a longer period with FVIII levels of ≥20-30 IU/dL to provide better bleed protection. The aspirational goal for PwHA is to prevent all joint bleeds, which may be achieved by maintaining normalised (50-150 IU/dL) FVIII levels. CONCLUSION: The panel of experts were largely aligned on approaches to preserving joint health in PwHA, and this consensus may help guide HCPs.


Asunto(s)
Hemofilia A , Humanos , Hemofilia A/tratamiento farmacológico , Factor VIII/uso terapéutico , Consenso , Hemartrosis/prevención & control , Hemorragia/prevención & control , Reino Unido
4.
Res Pract Thromb Haemost ; 6(2): e12690, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35356667

RESUMEN

Background: The Hemophilia Joint Health Score (HJHS) was developed and validated to detect arthropathy in children. Additional evidence is required to show validity in adults. We studied the convergent and discriminant construct validity of the HJHS version 2.1(HJHSv2.1) in adults with hemophilia. A secondary aim was to define age-related normative adult HJHSv2.1 reference values. Methods: We studied 192 adults with hemophilia, and 120 healthy adults in four age-matched groups-18 to 29, 30 to 40, 41 to 50, and >50 years-at nine centers. Trained physiotherapists scored the HJHS and World Federation of Hemophilia (WFH) joint score. Health history, the Functional Independence Scale of Hemophilia (FISH), Hemophilia Activities List (HAL), and Short-Form McGill Pain Questionnaire (SF-MPQ) were also collected. Results: The median age was 35.0 years. Of participants with hemophilia, 68% had severe, 14% moderate, and 18% mild disease. The HJHS correlated strongly with WFH score (Spearman's rho [rs ] = .95, P < .001). Moderate correlations were seen between the FISH (rs  = .50, P < .001) and SF-MPQ Present Pain Intensity (rs  = .50, P < .001), while a modest correlation was found with the HAL (rs  = -.37, P < .001). The HJHS significantly differentiated between age groups (Kruskal-Wallis T = 35.02, P < .001) and disease severity in participants with hemophilia. The HJHS had high internal reliability (Cronbach's α = .88). We identified duration of swelling as a redundant item in the HJHS. Conclusions: The HJHS shows evidence of strong convergent and discriminant construct validity to detect arthropathy in adults with hemophilia and is well suited for use in this population.

5.
Thromb Haemost ; 120(5): 747-757, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32369846

RESUMEN

BACKGROUND: The currently published population pharmacokinetic (PK) models used for PK-guided dosing in hemophilia patients are based on clinical trial data and usually not externally validated in clinical practice. The aim of this study was to validate a published model for recombinant factor VIII-Fc fusion protein (rFVIII-Fc) concentrate and to develop an enriched model using independently collected clinical data if required. METHODS: Clinical data from hemophilia A patients treated with rFVIII-Fc concentrate (Elocta) participating in the United Kingdom Extended Half-Life Outcomes Registry were collected. The predictive performance of the published model was assessed using mean percentage error (bias) and mean absolute percentage error (inaccuracy). An extended population PK model was developed using nonlinear mixed-effects modeling (NONMEM). RESULTS: A total of 43 hemophilia A patients (FVIII ≤ 2 IU/dL), aged 5 to 70 years, were included. The prior model was able to predict the collected 244 rFVIII-Fc levels without significant bias (-1.0%, 95% CI: -9.4 to 7.3%) and with acceptable accuracy (12.9%). However, clearance and central distribution volume were under predicted in patients <12 years, which was expected as this age group was not represented in the previous model population. An enriched population PK model was constructed, which was able to successfully characterize PK profiles of younger children. CONCLUSION: We concluded that the existing rFVIII-Fc population PK model is valid for patients ≥ 12 years. However, it is not reliable in younger patients. Our alternative model, constructed from real world patient data including children, allows for better description of patients ≥5 years.


Asunto(s)
Factor VIII/farmacocinética , Hemofilia A/tratamiento farmacológico , Hemostáticos/farmacocinética , Modelos Biológicos , Proteínas Recombinantes de Fusión/farmacocinética , Adolescente , Adulto , Factores de Edad , Anciano , Niño , Factor VIII/administración & dosificación , Factor VIII/efectos adversos , Hemofilia A/sangre , Hemofilia A/diagnóstico , Hemostáticos/administración & dosificación , Hemostáticos/efectos adversos , Humanos , Fragmentos Fc de Inmunoglobulinas/administración & dosificación , Fragmentos Fc de Inmunoglobulinas/efectos adversos , Masculino , Persona de Mediana Edad , Países Bajos , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Sistema de Registros , Reproducibilidad de los Resultados , Reino Unido , Adulto Joven
6.
Haemophilia ; 26(1): 33-40, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31742871

RESUMEN

The management of haemophilia-associated pseudotumours presents an ongoing challenge to the haematologist, surgeon and interventional radiologist alike. There is a range of therapeutic approaches including factor replacement, embolization, radiotherapy and a variety of surgical interventions. However, there remains little evidence regarding the most appropriate treatment. We aimed to evaluate the available options of management for the haemophilia-associated pseudotumour. A literature review was performed using relevant terminology and reviewed for treatment approaches and outcomes. The results demonstrated that most of the data is from single case reports with a small number of single- and multicentre case series. In total, 133 patients with 134 described pseudotumours were identified. Adequate haemostatic control with factor replacement was a key component to successful treatment. Surgical excision was the most commonly reported surgical intervention with various composites used for filling of the surgical cavity. The use of radiotherapy has been described particularly in the paediatric population and sites of difficult surgical access. Embolization can be considered as a method of presurgical optimization. Patients with both factor inhibitors and pseudotumours have poorer postoperative outcomes. This review demonstrates that although a lack of large-centre, randomized studies, timely surgical intervention with adequate haemostatic support and the consideration adjuvant therapies in selected cases can achieve acceptable outcomes in this cohort of patients.


Asunto(s)
Hemofilia A/complicaciones , Neoplasias/etiología , Factores de Coagulación Sanguínea/uso terapéutico , Embolización Terapéutica , Hemofilia A/tratamiento farmacológico , Humanos , Neoplasias/radioterapia , Neoplasias/cirugía , Complicaciones Posoperatorias/etiología
7.
Expert Rev Hematol ; 11(2): 87-97, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29258406

RESUMEN

INTRODUCTION: The development of inhibitors against factor VIII (FVIII) replacement therapy remains the most important challenge for clinicians in the treatment of hemophilia patients. This review focusses on risk factors and management of FVIII inhibitors, particularly in light of SIPPET study findings and subsequent analyses. Areas covered: A brief history and evolution of hemophilia therapies is provided, including an overview of conventional and new (including investigational) therapeutic approaches for the treatment of hemophilia. The SIPPET study, the first randomized clinical trial to demonstrate a lower incidence of inhibitors in previously untreated patients treated with plasma-derived FVIII products compared with recombinant FVIII products, has generated much debate. We review the SIPPET trial and reactions, in addition to preliminary observations from a single center's experience, the cost impact of inhibitors, recent findings from SIPPET subanalyses, and inhibitor development in previously-treated patients. Expert commentary: Despite recent advances in potential new treatment options for hemophilia, conventional factor replacement concentrates currently remain the cornerstone of treatment. It is paramount that clinicians familiarize themselves with the findings from the SIPPET trial and substudies, in order to better inform their patients and families on inhibitor risk factors and to aid the treatment decision-making process.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea , Factor VIII , Hemofilia A , Animales , Inhibidores de Factor de Coagulación Sanguínea/sangre , Inhibidores de Factor de Coagulación Sanguínea/inmunología , Factor VIII/efectos adversos , Factor VIII/inmunología , Factor VIII/uso terapéutico , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Recombinantes/efectos adversos , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico
8.
N Engl J Med ; 377(9): 819-828, 2017 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-28691885

RESUMEN

BACKGROUND: Current hemophilia treatment involves frequent intravenous infusions of clotting factors, which is associated with variable hemostatic protection, a high treatment burden, and a risk of the development of inhibitory alloantibodies. Fitusiran, an investigational RNA interference (RNAi) therapy that targets antithrombin (encoded by SERPINC1), is in development to address these and other limitations. METHODS: In this phase 1 dose-escalation study, we enrolled 4 healthy volunteers and 25 participants with moderate or severe hemophilia A or B who did not have inhibitory alloantibodies. Healthy volunteers received a single subcutaneous injection of fitusiran (at a dose of 0.03 mg per kilogram of body weight) or placebo. The participants with hemophilia received three injections of fitusiran administered either once weekly (at a dose of 0.015, 0.045, or 0.075 mg per kilogram) or once monthly (at a dose of 0.225, 0.45, 0.9, or 1.8 mg per kilogram or a fixed dose of 80 mg). The study objectives were to assess the pharmacokinetic and pharmacodynamic characteristics and safety of fitusiran. RESULTS: No thromboembolic events were observed during the study. The most common adverse events were mild injection-site reactions. Plasma levels of fitusiran increased in a dose-dependent manner and showed no accumulation with repeated administration. The monthly regimen induced a dose-dependent mean maximum antithrombin reduction of 70 to 89% from baseline. A reduction in the antithrombin level of more than 75% from baseline resulted in median peak thrombin values at the lower end of the range observed in healthy participants. CONCLUSIONS: Once-monthly subcutaneous administration of fitusiran resulted in dose-dependent lowering of the antithrombin level and increased thrombin generation in participants with hemophilia A or B who did not have inhibitory alloantibodies. (Funded by Alnylam Pharmaceuticals; ClinicalTrials.gov number, NCT02035605 .).


Asunto(s)
Antitrombina III/antagonistas & inhibidores , Hemofilia A/terapia , Hemofilia B/terapia , Tratamiento con ARN de Interferencia , Adulto , Antitrombinas/sangre , Relación Dosis-Respuesta a Droga , Voluntarios Sanos , Hemofilia A/sangre , Hemofilia B/sangre , Humanos , Inyecciones Subcutáneas , Masculino , Persona de Mediana Edad , Método Simple Ciego , Trombina/biosíntesis , Adulto Joven
9.
Blood ; 130(4): 466-471, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28576877

RESUMEN

Immune-mediated thrombotic thrombocytopenic purpura (TTP) is a life-threatening disorder caused by antibodies against ADAMTS13. From the United Kingdom TTP registry, we undertook a prospective study investigating the impact of the presenting anti-ADAMTS13 IgG antibody and ADAMTS13 antigen on mortality. A total of 312 episodes involving 292 patients over 87 months were included; 68% were female, median age 46 (range, 11-88 years), and median presenting ADAMTS13 of <5% (range, <5%-18%). The mortality rate was 10.3% (n = 32); 68% of patients had a raised troponin at presentation conferring a sixfold increase in mortality compared with those with normal troponin levels (12.1% vs 2.0%, P = .04). Twenty-four percent had a reduced Glasgow Coma Score (GCS) at presentation with a ninefold increase in mortality (20% vs 2.2% for normal GCS at presentation, P < .0001). Mortality increased with higher anti-ADAMTS13 antibody levels and lower ADAMTS13 antigen levels. Those with antibody levels in the upper quartile (antibody >77%) had a mortality of 16.9% compared with 5.0% for the lowest quartile (antibody <20%) (P = .004). Those with an antigen level in the lowest quartile (antigen <1.5%) had a mortality of 18% compared with 3.8% for the highest quartile (antigen >11%) (P = .005). The synergistic effect of anti-ADAMTS13 IgG antibody in the upper quartile and ADAMTS13 antigen in the lowest quartile had the highest mortality of 27.3%. We conclude that both anti-ADAMTS13 IgG antibody and ADAMTS13 antigen levels correlate with outcome in TTP with increased cardiac and neurological involvement and increased mortality.


Asunto(s)
Proteína ADAMTS13 , Autoanticuerpos , Inmunoglobulina G , Púrpura Trombocitopénica Trombótica , Proteína ADAMTS13/sangre , Proteína ADAMTS13/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Autoanticuerpos/sangre , Autoanticuerpos/inmunología , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Masculino , Persona de Mediana Edad , Púrpura Trombocitopénica Trombótica/sangre , Púrpura Trombocitopénica Trombótica/inmunología , Púrpura Trombocitopénica Trombótica/mortalidad , Tasa de Supervivencia
10.
Br J Haematol ; 176(5): 796-804, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28198996

RESUMEN

Inhibitor formation in non-severe haemophilia A is a life-long risk and associated with morbidity and mortality. There is a paucity of data to understand real-world inhibitor screening practice. We evaluated the treatment burden, haemostatic strategies, F8 genotyping and inhibitor screening practices in non-severe haemophilia A in seven London haemophilia centres. In the 2-year study period, 44% (377/853) patients received at least one haemostatic treatment. Seventy-nine percent of those treated (296/377) received factor VIII (FVIII) concentrate. F8 genotype was known in 88% (331/377) of individuals. Eighteen per cent (58/331) had 'high-risk' F8 genotypes. In patients with 'standard-risk' F8 genotypes treated on-demand with FVIII concentrate, 51·3% episodes (243/474) were screened within 1 year. However, poor screening compliance was observed after 'high-risk' treatment episodes. In patients with 'standard-risk' F8 genotypes, 12·3% (28/227) of treatment episodes were screened in the subsequent 6 weeks after surgery or a bleed requiring ≥5 exposure days. Similarly, in the context of 'high-risk' F8 genotypes after any FVIII exposure, only 13·6% (12/88) of episodes were screened within 6 weeks. Further study is required to assess optimal practice of inhibitor screening in non-severe haemophilia A to inform subsequent clinical decisions and provide more robust prevalence data to further understand the underlying immunological mechanism.


Asunto(s)
Factor VIII/genética , Genotipo , Hemofilia A/inmunología , Hemofilia A/terapia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Hemofilia A/genética , Hemostáticos/uso terapéutico , Humanos , Lactante , Tamizaje Masivo/métodos , Persona de Mediana Edad , Estudios Retrospectivos , Medición de Riesgo , Adulto Joven
12.
Hamostaseologie ; 37(2): 104-106, 2017 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27598535

RESUMEN

Haemophilic arthropathy is a complex multifactorial disorder that poses significant challenges to both the treating haematologist and arthroplasty surgeon. Its pathogenesis is incompletely understood. Recent literature has concentrated on the toxic effects of iron and the characteristic inflammatory synovitis. Discussion of the role of subchondral bleeding in joint damage has been neglected. A case of haemophilic arthropathy with extensive evidence of subchondral bleeding and related osteochondral destruction is presented. RESULT: The relevance of this mechanical pathway in the future management of haemophilic arthropathy is discussed with reference to recent literature. CONCLUSION: Clinicians should consider its importance when deciding whether to manage patients expectantly or with prophylactic factor replacement.


Asunto(s)
Artroplastia de Reemplazo de Rodilla , Hemartrosis/diagnóstico por imagen , Hemartrosis/cirugía , Hemofilia A/diagnóstico , Hemofilia A/cirugía , Articulación de la Rodilla/diagnóstico por imagen , Articulación de la Rodilla/cirugía , Anciano , Diagnóstico Diferencial , Hemartrosis/etiología , Hemofilia A/complicaciones , Humanos , Masculino , Resultado del Tratamiento
13.
Hamostaseologie ; 37(2): 104-106, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29582910

RESUMEN

Haemophilic arthropathy is a complex multifactorial disorder that poses significant challenges to both the treating haematologist and arthroplasty surgeon. Its pathogenesis is incompletely understood. Recent literature has concentrated on the toxic effects of iron and the characteristic inflammatory synovitis. Discussion of the role of subchondral bleeding in joint damage has been neglected. A case of haemophilic arthropathy with extensive evidence of subchondral bleeding and related osteochondral destruction is presented. RESULT: The relevance of this mechanical pathway in the future management of haemophilic arthropathy is discussed with reference to recent literature. CONCLUSION: Clinicians should consider its importance when deciding whether to manage patients expectantly or with prophylactic factor replacement.


Asunto(s)
Enfermedades Óseas/diagnóstico , Fémur , Hemartrosis/diagnóstico , Hematoma/diagnóstico , Hemofilia A/diagnóstico , Articulación de la Rodilla , Anciano , Artroplastia de Reemplazo de Rodilla , Enfermedades Óseas/sangre , Enfermedades Óseas/cirugía , Comorbilidad , Factor VIII/análisis , Fémur/cirugía , Hemartrosis/sangre , Hemartrosis/cirugía , Hematoma/sangre , Hematoma/cirugía , Hemofilia A/sangre , Hemofilia A/complicaciones , Hepatitis C/sangre , Hepatitis C/diagnóstico , Humanos , Articulación de la Rodilla/cirugía , Imagen por Resonancia Magnética , Masculino
14.
Sci Transl Med ; 8(328): 328ra30, 2016 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-26936507

RESUMEN

The Src family kinase (SFK) member SRC is a major target in drug development because it is activated in many human cancers, yet deleterious SRC germline mutations have not been reported. We used genome sequencing and Human Phenotype Ontology patient coding to identify a gain-of-function mutation in SRC causing thrombocytopenia, myelofibrosis, bleeding, and bone pathologies in nine cases. Modeling of the E527K substitution predicts loss of SRC's self-inhibitory capacity, which we confirmed with in vitro studies showing increased SRC kinase activity and enhanced Tyr(419) phosphorylation in COS-7 cells overexpressing E527K SRC. The active form of SRC predominates in patients' platelets, resulting in enhanced overall tyrosine phosphorylation. Patients with myelofibrosis have hypercellular bone marrow with trilineage dysplasia, and their stem cells grown in vitro form more myeloid and megakaryocyte (MK) colonies than control cells. These MKs generate platelets that are dysmorphic, low in number, highly variable in size, and have a paucity of α-granules. Overactive SRC in patient-derived MKs causes a reduction in proplatelet formation, which can be rescued by SRC kinase inhibition. Stem cells transduced with lentiviral E527K SRC form MKs with a similar defect and enhanced tyrosine phosphorylation levels. Patient-derived and E527K-transduced MKs show Y419 SRC-positive stained podosomes that induce altered actin organization. Expression of mutated src in zebrafish recapitulates patients' blood and bone phenotypes. Similar studies of platelets and MKs may reveal the mechanism underlying the severe bleeding frequently observed in cancer patients treated with next-generation SFK inhibitors.


Asunto(s)
Huesos/patología , Hemorragia/genética , Mutación/genética , Mielofibrosis Primaria/genética , Trombocitopenia/genética , Familia-src Quinasas/genética , Animales , Plaquetas/patología , Células COS , Chlorocebus aethiops , Femenino , Hematopoyesis , Hemorragia/complicaciones , Humanos , Masculino , Linaje , Fenotipo , Mielofibrosis Primaria/complicaciones , Trombocitopenia/complicaciones , Transfección , Pez Cebra
15.
Blood ; 127(23): 2903-14, 2016 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-26912466

RESUMEN

Macrothrombocytopenia (MTP) is a heterogeneous group of disorders characterized by enlarged and reduced numbers of circulating platelets, sometimes resulting in abnormal bleeding. In most MTP, this phenotype arises because of altered regulation of platelet formation from megakaryocytes (MKs). We report the identification of DIAPH1, which encodes the Rho-effector diaphanous-related formin 1 (DIAPH1), as a candidate gene for MTP using exome sequencing, ontological phenotyping, and similarity regression. We describe 2 unrelated pedigrees with MTP and sensorineural hearing loss that segregate with a DIAPH1 R1213* variant predicting partial truncation of the DIAPH1 diaphanous autoregulatory domain. The R1213* variant was linked to reduced proplatelet formation from cultured MKs, cell clustering, and abnormal cortical filamentous actin. Similarly, in platelets, there was increased filamentous actin and stable microtubules, indicating constitutive activation of DIAPH1. Overexpression of DIAPH1 R1213* in cells reproduced the cytoskeletal alterations found in platelets. Our description of a novel disorder of platelet formation and hearing loss extends the repertoire of DIAPH1-related disease and provides new insight into the autoregulation of DIAPH1 activity.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Pérdida Auditiva/genética , Mutación , Trombocitopenia/genética , Células A549 , Adolescente , Adulto , Anciano , Estudios de Casos y Controles , Células Cultivadas , Niño , Femenino , Forminas , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Células HEK293 , Pérdida Auditiva/complicaciones , Humanos , Masculino , Persona de Mediana Edad , Linaje , Polimorfismo de Nucleótido Simple , Síndrome , Trombocitopenia/complicaciones , Adulto Joven
16.
Genome Med ; 7(1): 36, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25949529

RESUMEN

BACKGROUND: Heritable bleeding and platelet disorders (BPD) are heterogeneous and frequently have an unknown genetic basis. The BRIDGE-BPD study aims to discover new causal genes for BPD by high throughput sequencing using cluster analyses based on improved and standardised deep, multi-system phenotyping of cases. METHODS: We report a new approach in which the clinical and laboratory characteristics of BPD cases are annotated with adapted Human Phenotype Ontology (HPO) terms. Cluster analyses are then used to characterise groups of cases with similar HPO terms and variants in the same genes. RESULTS: We show that 60% of index cases with heritable BPD enrolled at 10 European or US centres were annotated with HPO terms indicating abnormalities in organ systems other than blood or blood-forming tissues, particularly the nervous system. Cases within pedigrees clustered closely together on the bases of their HPO-coded phenotypes, as did cases sharing several clinically suspected syndromic disorders. Cases subsequently found to harbour variants in ACTN1 also clustered closely, even though diagnosis of this recently described disorder was not possible using only the clinical and laboratory data available to the enrolling clinician. CONCLUSIONS: These findings validate our novel HPO-based phenotype clustering methodology for known BPD, thus providing a new discovery tool for BPD of unknown genetic basis. This approach will also be relevant for other rare diseases with significant genetic heterogeneity.

17.
J Acquir Immune Defic Syndr ; 46(4): 498-507, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18077841

RESUMEN

OBJECTIVE: To comprehensively assess iron status and determine whether elevated iron status, like anemia, predicts mortality. METHODS: We followed 1362 Gambian adults (53% female) in an HIV-seroprevalent clinic-based cohort over 11.5 years to ascertain all-cause mortality. Baseline iron status (iron, soluble transferrin receptor [sTfR], transferrin, ferritin, transferrin saturation, log [transferrin receptor: ferritin]), age, gender, ethnicity, hemoglobin, body mass index, HIV type, absolute CD4 count, malaria status, and [alpha]-(1)-antichymotrypsin were measured. RESULTS: The mortality rate was 25.9/100 person-years. Elevated iron universally predicted greater mortality compared to normal iron status for all iron status indices, with the exception of sTfR in unadjusted models. In fully adjusted models, transferrin (elevated vs. normal, hazard ratio [HR]: 1.77; 95% confidence interval [CI]: 1.30 to 2.42; P < 0.001), ferritin (elevated vs. normal, HR: 1.40; 95% CI: 1.07 to 1.83; P = 0.014), and the combined iron status index (highly elevated vs. normal, HR: 2.20; 95% CI: 1.16 to 4.18; P = 0.016) remained significant predictors. As expected, hemoglobin (Hb) concentration and absolute CD4 counts were each inversely associated with mortality. CONCLUSIONS: Elevated iron status predicts mortality in HIV infection, even after adjustment for immunosuppression and other confounders. This finding has implications in the clinical monitoring of disease progression and for iron-supplementation practices in areas of high HIV prevalence.


Asunto(s)
Infecciones por VIH/sangre , Hierro/sangre , Adolescente , Adulto , Estudios de Cohortes , Demografía , Etnicidad/estadística & datos numéricos , Femenino , Gambia/epidemiología , Infecciones por VIH/mortalidad , Humanos , Masculino , Encuestas y Cuestionarios , Análisis de Supervivencia
18.
Blood ; 108(10): 3494-503, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16868253

RESUMEN

The receptor tyrosine kinase FLT3 is a promising molecular therapeutic target in acute myeloid leukemia (AML). Activating mutations of FLT3 are present in approximately one-third of patients, while many nonmutants show evidence of FLT3 activation, which appears to play a significant role in leukemogenesis. We studied the effects of lestaurtinib (CEP701) and PKC412, 2 small molecule inhibitors of FLT3, on 65 diagnostic AML blast samples. Both agents induced concentration-dependent cytotoxicity in most cases, although responses to PKC412 required higher drug concentrations. Cytotoxic responses were highly heterogeneous and were only weakly associated with FLT3 mutation status and FLT3 expression. Importantly, lestaurtinib induced cytotoxicity in a synergistic fashion with cytarabine, particularly in FLT3 mutant samples. Both lestaurtinib and PKC412 caused inhibition of FLT3 phosphorylation in all samples. Translation of FLT3 inhibition into cytotoxicity was influenced by the degree of residual FLT3 phosphorylation remaining and correlated with deactivation of STAT5 and MAP kinase. FLT3 mutant and wild-type cases both varied considerably in their dependence on FLT3 signaling for survival. These findings support the continued clinical assessment of FLT3 inhibitors in combination with cytotoxic chemotherapy: Entry to future clinical trials should include FLT3 wild-type patients and should remain unrestricted by FLT3 expression level.


Asunto(s)
Carbazoles/farmacología , Indoles/farmacología , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Estaurosporina/análogos & derivados , Enfermedad Aguda , Adolescente , Adulto , Anciano , Apoptosis/efectos de los fármacos , Crisis Blástica/patología , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Furanos , Humanos , Masculino , Persona de Mediana Edad , Mutación , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Transducción de Señal , Estaurosporina/farmacología , Células Tumorales Cultivadas , Tirosina Quinasa 3 Similar a fms/genética , Tirosina Quinasa 3 Similar a fms/fisiología
19.
Aust Vet J ; 82(1-2): 30, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15088951
20.
Leuk Res ; 26(11): 997-1005, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12363468

RESUMEN

We recorded elevated numbers of circulating myeloid and erythroid colony-forming cells in 15 adult patients with acute myeloid leukaemia (AML) who presented with high blood white cell counts. Since leukaemic blasts from three of these patients were Philadelphia chromosome-positive (Ph+), we were able to determine if blood progenitors from these particular patients arose from the leukaemic clone or from residual normal progenitors. Blasts and colonies were intensively investigated using a combination of cell surface marker analysis by flow cytometry, RT-PCR and interphase fluorescence in situ hybridization (FISH). FISH detected rearrangements within the major breakpoint BCR (M-BCR) region in blasts and in some myeloid and erythroid colonies from patients 1 and 2. The minor breakpoint (m-BCR) region was detected in blasts and in some myeloid and erythroid colonies from patient 3. RT-PCR detected long b2a2 BCR-ABL transcripts in blasts from patients 1 and 2, although misspliced short e1a2 transcripts were also seen in patient 1. Only e1a2 transcripts were found in blasts from patient 3. Flow sorting demonstrated the B-cell marker CD19 on blasts and on a proportion of myeloid and erythroid progenitors from patients 1 and 3. RT-PCR also detected IgH rearrangements, further evidence of B-cell differentiation, in blasts from these two patients. We conclude that both normal and clonal circulating progenitor numbers can be raised in both M-BCR and m-BCR Ph+ AML. The underlying cause, perhaps efflux from a congested marrow, may be common to AML patients with a high blood white cell count.


Asunto(s)
Células Precursoras Eritroides/metabolismo , Proteínas de Fusión bcr-abl/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Células Progenitoras Mieloides/metabolismo , Proteínas Tirosina Quinasas , Proteínas Proto-Oncogénicas , Enfermedad Aguda , Adulto , Biomarcadores de Tumor , Estudios de Casos y Controles , Ensayo de Unidades Formadoras de Colonias , ADN de Neoplasias/análisis , Citometría de Flujo , Proteínas de Fusión bcr-abl/sangre , Genes de Inmunoglobulinas/genética , Humanos , Hibridación Fluorescente in Situ , Cariotipificación , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Recuento de Leucocitos , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-bcr , ARN Mensajero/genética , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...