Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Haemophilia ; 23(2): e93-e98, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28124406

RESUMO

INTRODUCTION: High-titre factor VIII (FVIII) inhibitors complicate peri-operative haemostasis. Recombinant porcine FVIII (r-pFVIII) may provide an alternative haemostatic agent for high-risk procedures and allow FVIII activity monitoring. AIM: Devise an effective haemostatic plan for repair of a progressively symptomatic aortic coarctation in a 5-year-old male with immune tolerance induction (ITI) refractory high-titre FVIII inhibitors. METHODS: Preprocedure human FVIII inhibitor titre was 58 Bethesda Units mL-1 (BU) and cross-reacted to neutralize porcine FVIII at 30 BU. Daily ITI with plasma-derived FVIII concentrate was supplemented with anti-B-cell and anti-plasma cell immunotherapy to reduce FVIII inhibitor titres. Potential haemostatic agents were evaluated in comparative ex vivo thrombin generation assays (TGA). RESULTS: Four weeks after immunosuppression, human and porcine inhibitor titres declined to 16 and 2 BU respectively. TGA with r-pFVIII was less robust than with activated prothrombin complex concentrate (aPCC); however, r-pFVIII was selected for cardiac surgery to secure the ability to assay FVIII levels throughout this high-bleeding risk procedure. Haemostasis with r-pFVIII was excellent; initial trough FVIII activity levels ranged from 0.81-1.17 IU mL-1 . On postoperative day 3, peak and trough levels markedly declined suggesting a rising porcine inhibitor titre. Postprocedure prophylaxis was transitioned to aPCC, informed by TGA. CONCLUSIONS: R-pFVIII provided effective peri-procedural haemostasis with no adverse events. Rapid neutralization of r-pFVIII after the first 60 hours, despite intensive immune suppression, accentuates the importance of careful monitoring. Use of TGA can support bypassing agent selection for convalescence. The comparative cost of r-pFVIII may limit its use to high morbidity clinical scenarios.


Assuntos
Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Proteínas Recombinantes/uso terapêutico , Animais , Pré-Escolar , Humanos , Masculino , Proteínas Recombinantes/administração & dosagem , Suínos
2.
Haemophilia ; 20(1): 25-31, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23855900

RESUMO

Several risk factors for inhibitors have recently been described for haemophilia A. It has been assumed that similar risk factors are also relevant for haemophilia B, but there is limited data to confirm this notion. The aim of this study was to determine the prevalence of and risk factors associated with inhibitors in haemophilia B. The database of the Universal Data Collection (UDC) project of the Centers for Disease Control for the years 1998-2011 was queried to determine the prevalence of inhibitors in haemophilia B subjects. In addition, disease severity, race/ethnicity, age, factor exposure and prophylaxis usage were evaluated to determine their impact on inhibitor prevalence. Of the 3785 male subjects with haemophilia B enrolled in the UDC database, 75 (2%) were determined to have an inhibitor at some point during the study period. Severe disease (OR 13.1, 95% CI 6.2-27.7), black race (OR 2.2, 95% CI 1.2-4.1), and age <11 years (OR 2.5, 95% CI 1.5-4.0) were found to be significantly associated with having an inhibitor. There was insufficient data to determine if type of factor used and prophylaxis were associated with inhibitors. Inhibitors in haemophilia B are much less prevalent than haemophilia A, especially in patients with mild disease. Similar factors associated with inhibitors in haemophilia A also seem to be present for haemophilia B. The information collected by this large surveillance project did not permit evaluation of potential risk factors related to treatment approaches and exposures, and additional studies will be required.


Assuntos
Fator IX/antagonistas & inibidores , Hemofilia B/epidemiologia , Adolescente , Adulto , Criança , Pré-Escolar , Bases de Dados Factuais , Fator IX/uso terapêutico , Hemofilia B/sangue , Hemofilia B/tratamento farmacológico , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Prevalência , Fatores de Risco , Adulto Jovem
3.
Haemophilia ; 20(5): 716-22, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24712867

RESUMO

While chronic degenerative arthropathy is the main morbidity of haemophilia, a very high prevalence of low bone density is also seen in men and boys with haemophilia. This study investigates bone degradation in the knee joint of haemophilic mice resulting from haemarthrosis and the efficacy of aggressive treatment with factor VIII in the period surrounding injury to prevent bone pathology. Skeletally mature factor VIII knock-out mice were subjected to knee joint haemorrhage induced by puncture of the left knee joint capsule. Mice received either intravenous factor VIII treatment or placebo immediately prior to injury and at hours 4, 24, 48, 72 and 96 after haemorrhage. Mice were killed 2-weeks after injury and the joint morphology and loss of bone in the proximal tibia was assessed using microCT imaging. Quantitative microCT imaging of the knee joint found acute bone loss at the proximal tibia following injury including loss of trabecular bone volumetric density and bone mineral density, as well as trabecular connectivity density, number and thickness. Unexpectedly, joint injury also resulted in calcification of the joint soft tissues including the tendons, ligaments, menisci and cartilage. Treatment with factor VIII prevented this bone and soft tissue degeneration. Knee joint haemorrhage resulted in acute changes in adjacent bone including loss of bone density and mineralization of joint soft tissues. The rapid calcification and loss of bone has implications for the initiation and progression of osteoarthritic degradation following joint bleeding.


Assuntos
Calcinose/etiologia , Coagulantes/uso terapêutico , Fator VIII/uso terapêutico , Hemartrose/complicações , Hemofilia A/complicações , Osteoartrite do Joelho , Osteoporose/etiologia , Malha Trabecular , Doença Aguda , Animais , Modelos Animais de Doenças , Hemartrose/tratamento farmacológico , Hemofilia A/tratamento farmacológico , Masculino , Camundongos , Camundongos Knockout , Osteoartrite do Joelho/tratamento farmacológico , Osteoartrite do Joelho/etiologia , Osteoartrite do Joelho/patologia , Osteoporose/prevenção & controle , Tíbia , Microtomografia por Raio-X
4.
Gene Ther ; 19(3): 288-94, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21697954

RESUMO

Recombinant adeno-associated virus (rAAV) is a promising gene delivery vector and has recently been used in patients with hemophilia. One limitation of AAV application is that most humans have experienced wild-type AAV serotype 2 exposure, which frequently generates neutralizing antibodies (NAbs) that may inhibit rAAV2 vector transduction. Employing alternative serotypes of rAAV vectors may circumvent this problem. We investigated the development of NAbs in early childhood by examining sera gathered prospectively from 62 children with hemophilia A, participating in a multi-institutional hemophilia clinical trial (the Joint Outcome Study). Clinical applications in hemophilia therapy have been suggested for serotypes AAV2, AAV5 and AAV8, therefore NAbs against these serotypes were serially assayed over a median follow-up of 4 years. NAbs prevalence increased during early childhood for all serotypes. NAbs against AAV2 (43.5%) were observed more frequently and at higher titers compared with both AAV5 (25.8%) and AAV8 (22.6%). NAbs against AAV5 or AAV8 were rarely observed in the absence of co-prevalent and higher titer AAV2 NAbs, suggesting that NAbs to AAV5 and AAV8 were detected following AAV2 exposure due to partial cross-reactivity of AAV2-directed NAbs. The results may guide rational design of clinical trials using alternative AAV serotypes and suggest that younger patients who are given AAV gene therapy will benefit from the lower prevalence of NAbs.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Dependovirus/imunologia , Hemofilia A/imunologia , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Linhagem Celular , Criança , Seguimentos , Vetores Genéticos/imunologia , Hemofilia A/virologia , Humanos , Masculino , Infecções por Parvoviridae/epidemiologia , Infecções por Parvoviridae/imunologia , Estudos Soroepidemiológicos
5.
Haemophilia ; 18 Suppl 4: 112-9, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22726093

RESUMO

Blood in the joint causes a number of physiological and pathological events that eventually lead to haemophilic arthropathy. Animal models show that blood in the joint induces inflammation that continues long after blood has been cleared. TNF-alpha, IL-1 beta and IL-6 are inflammatory mediators that increase following haemarthrosis in haemophilic mice. Conventional anti-inflammatory drugs have failed to demonstrate a lasting effect in preventing haemophilic arthropathy. A new TNF-alpha antagonist has shown promising results in haemophilic mice. Similarly, the use of cyclo-oxygenase-2 inhibitors may reduce angiogenesis associated with the healing process following bleeding and the associated tissue damage. Animal models are useful for studying the pathophysiology of haemarthropathy, however, when applying results from animals to humans, the differences in matrix turnover rate, thickness of cartilage and joint biomechanics must be kept in mind. In people with haemophilia, there is a variable response to haemarthrosis as demonstrated by magnetic resonance imaging (MRI). Up to 30% of subjects have normal MRI despite having three or more haemarthroses into the same joint. Once bone damage is present, little can be done to restore anatomic integrity. Several molecules, including members of the bone morphogenic protein subfamily, have been injected into bone defects in non-haemophilic subjects with some evidence of benefit. To achieve the primary goal of reducing blood in the joint and the negative sequelae, it is questionable to use ice to treat haemarthrosis. Indeed low temperature is associated with impairment of coagulation enzyme activity and platelet function.


Assuntos
Hemartrose/terapia , Hemofilia A/complicações , Hemofilia B/complicações , Animais , Anti-Inflamatórios/uso terapêutico , Crioterapia , Inibidores de Ciclo-Oxigenase/farmacologia , Citocinas/metabolismo , Modelos Animais de Doenças , Hemartrose/complicações , Hemartrose/metabolismo , Hemofilia A/tratamento farmacológico , Hemofilia B/tratamento farmacológico , Humanos , Camundongos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Cicatrização/efeitos dos fármacos
6.
Gene Ther ; 18(8): 842-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21412285

RESUMO

Hemophilia B, a hereditary bleeding disorder caused by a deficiency of coagulation factor IX (FIX), is an excellent candidate for gene therapy. However, to date, success in hemophilia gene therapy clinical trials has been limited due to failure to achieve or sustain therapeutic levels of factor expression. The ΦC31 integrase system efficiently integrates plasmid DNA carrying a transgene and an attB site into a limited number of endogenous pseudo attP sites in mammalian genomes, leading to robust, sustained transgene expression. A strategy utilizing plasmid DNA integrated with ΦC31 integrase may offer a facile and safe alternative for sustained human FIX (hFIX) expression. Hydrodynamic tail vein injection was used for delivery of plasmids encoding ΦC31 integrase and hFIX to the liver of FIX knockout mice. We demonstrated prolonged therapeutic levels of hFIX in this knockout mouse model of hemophilia B over a 6-month time course when ΦC31 integrase was used. Additionally, we observed sustained FIX activity in plasma and phenotypic correction of bleeding after tail clip in ΦC31-treated mice. In the livers that received integrase, we also demonstrated prolonged hFIX expression in hepatocytes by immunohistochemistry and documented sequence-specific genomic integration of the hFIX plasmid. These studies suggest the possibility that a similar approach in large animals and humans could lead to a simple and successful gene therapy for hemophilia.


Assuntos
Bacteriófagos , Fator IX/genética , Técnicas de Transferência de Genes , Terapia Genética , Hemofilia B/terapia , Integrases , Animais , Bacteriófagos/genética , Modelos Animais de Doenças , Fator IX/metabolismo , Camundongos , Camundongos Knockout
7.
Haemophilia ; 16 Suppl 5: 29-34, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20590853

RESUMO

SUMMARY: Gene therapy of haemophilia has been initiated through a number of approaches including expression in muscle, liver and omental implanted fibroblasts, or i.v. injection of an expression construct under the control of a ubiquitous promoter. In all these approaches, the goal was to have factor VIII (FVIII) or factor IX (FIX) synthesized so that it restored the levels of the missing protein in blood. The three talks in this session are totally, or at least in part, directed at strategies that may be clinically effective even in the absence of correction of the missing plasma clotting factor, although the haematopoietic stem cell or blood outgrowth endothelial cell therapy could achieve plasma correction as well. Two of the approaches achieve localized coagulation factor expression without necessarily correcting the systemic defect--one is with synthesis of FVIII or FIX within the joint space and the other is with the local release of FVIII (or FIX) by platelets at the site of vascular injury. All of the three approaches have demonstrated efficacy in small animal models and are now the subject of larger animal studies. None has yet to progress to human trials.


Assuntos
Fator IX/uso terapêutico , Fator VIII/uso terapêutico , Fator X/uso terapêutico , Técnicas de Transferência de Genes , Terapia Genética/métodos , Hemofilia A/terapia , Hemofilia B/terapia , Fator IX/biossíntese , Fator IX/genética , Fator VIII/biossíntese , Fator VIII/genética , Fator X/biossíntese , Fator X/genética , Células-Tronco Hematopoéticas/metabolismo , Hemofilia A/genética , Hemofilia B/genética , Humanos
8.
Haemophilia ; 16(3): 460-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20059559

RESUMO

Prophylaxis is increasingly prescribed in treatment of haemophilia and its benefit is believed to be most significant for the youngest patients as haemophilic arthropathy may be prevented if prophylaxis is initiated prior to recurrent haemarthroses. While clinical prophylaxis data are readily available for haemophilia A, analogous data for haemophilia B are relatively limited. A prospective clinical study of recombinant factor IX (BeneFIX; rFIX), designed to allow investigator prescribed prophylaxis according to customary practices, was conducted in children <6 years old with severe haemophilia B. Nearly all children were prescribed prophylaxis (22/25; 88%) for all or part of their study participation. Favourable efficacy and safety profiles were reported. Routine prophylaxis with 1 or 2 rFIX infusions per week over an average of greater than 6 months of therapy resulted in near complete prevention of spontaneous breakthrough haemorrhages (<1 per year), with most children (77%) having none, including seven patients (32%) who had no bleeding episodes at all. Haemorrhages in joints were less common than those outside joints (27% vs. 73% of haemorrhages). In a patient population that included children with multiple prior haemarthroses, 68% of children had no joint bleeding. Breakthrough haemorrhages resolved with 1 or 2 infusions in 89% of episodes. The absence of changes in prophylaxis infusion schedules suggests that 1 or 2 rFIX infusion(s) per week were well-tolerated by these young patients, including those with (41%) and without (59%) central venous access devices. Safety was established by the low incidence of treatment-related adverse events.


Assuntos
Fator IX/uso terapêutico , Hemartrose/prevenção & controle , Hemofilia B/tratamento farmacológico , Hemorragia/prevenção & controle , Proteínas Recombinantes/uso terapêutico , Criança , Pré-Escolar , Fator IX/efeitos adversos , Fator IX/farmacocinética , Feminino , Humanos , Lactente , Masculino , Estudos Prospectivos , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/farmacocinética
9.
Haemophilia ; 14(6): 1209-13, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19141161

RESUMO

Combined deficiency of vitamin K-dependent clotting factors II, VII, IX and X (and proteins C, S, and Z) is usually an acquired clinical problem, often resulting from liver disease, malabsorption, or warfarin overdose. A rare inherited form of defective gamma-carboxylation resulting in early onset of bleeding was first described by McMillan and Roberts in 1966 and subsequently has been termed 'vitamin K-dependent clotting factor deficiency' (VKCFD). Biochemical and molecular studies identify two variants of this autosomal recessive disorder: VKCFD1, which is associated with point mutations in the gamma-glutamylcarboxylase gene (GGCX), and VKCFD2, which results from point mutations in the vitamin K epoxide reductase gene (VKOR). Bleeding ranges in severity from mild to severe. Therapy includes high oral doses of vitamin K for prophylaxis, usually resulting in partial correction of factor deficiency, and episodic use of plasma infusions or prothrombin complex concentrate. Recent molecular studies have the potential to further our understanding of vitamin K metabolism, gamma-carboxylation, and the functional role this post-translational modification has for other proteins. The results may also provide potential targets for molecular therapeutics and pharmacogenetics.


Assuntos
Transtornos da Coagulação Sanguínea/congênito , Proteínas Sanguíneas/deficiência , Vitamina K/metabolismo , Adulto , Transtornos da Coagulação Sanguínea/diagnóstico , Transtornos da Coagulação Sanguínea/tratamento farmacológico , Fatores de Coagulação Sanguínea/uso terapêutico , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Carbono-Carbono Ligases/genética , Feminino , Hemorragia/sangue , Humanos , Lactente , Recém-Nascido , Oxigenases de Função Mista/genética , Plasma , Gravidez , Vitamina K/uso terapêutico , Vitamina K Epóxido Redutases
10.
J Thromb Haemost ; 13 Suppl 1: S151-60, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26149016

RESUMO

Factor IX deficiency (hemophilia B) is less common than factor VIII deficiency (hemophilia A), and innovations in therapy for hemophilia B have generally lagged behind those for hemophilia A. Recently, the first sustained correction of the hemophilia bleeding phenotype by clotting factor gene therapy has been described using recombinant adeno-associated virus (AAV) to deliver factor IX. Despite this success, many individuals with hemophilia B, including children, men with active hepatitis, and individuals who have pre-existing natural immunity to AAV, are not eligible for the current iteration of hemophilia B gene therapy. In addition, recent advances in recombinant factor IX protein engineering have led some hemophilia treaters to reconsider the urgency of genetic cure. Current clinical and preclinical approaches to advancing AAV-based and alternative approaches to factor IX gene therapy are considered in the context of current demographics and treatment of the hemophilia B population.


Assuntos
Terapia Genética/métodos , Hemofilia B/terapia , Animais , Dependovirus/genética , Fator IX/biossíntese , Fator IX/genética , Fator IX/uso terapêutico , Técnicas de Transferência de Genes , Predisposição Genética para Doença , Vetores Genéticos , Hemofilia B/sangue , Hemofilia B/diagnóstico , Hemofilia B/genética , Hemostáticos/uso terapêutico , Humanos , Lentivirus/genética , Masculino , Proteínas Recombinantes/uso terapêutico , Resultado do Tratamento
11.
J Thromb Haemost ; 13(11): 1980-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26382916

RESUMO

BACKGROUND: Immune tolerance induction (ITI) in patients with congenital hemophilia A is successful in up to 70%. Although there is growing understanding of predictors of response to ITI, the probability and predictors of inhibitor recurrence after successful ITI are not well understood. OBJECTIVES: To determine the association of clinical characteristics, particularly adherence to factor VIII (FVIII) prophylaxis after ITI, with inhibitor recurrence in patients with hemophilia A who were considered tolerant after ITI. METHODS: In this multicenter retrospective cohort study, 64 subjects with FVIII level < 2% who were considered successfully tolerant after ITI were analyzed to estimate the cumulative probability of inhibitor recurrence using the Kaplan-Meier method. The association of clinical characteristics with inhibitor recurrence was assessed using logistic regression. RESULTS: A recurrent inhibitor titer ≥ 0.6 BU mL(-1) occurred at least once in 19 (29.7%) and more than once in 12 (18.8%). The probability of any recurrent inhibitor at 1 and 5 years was 12.8% and 32.5%, respectively. Having a recurrent inhibitor was associated with having received immune modulation during ITI (odds ratio [OR] 3.8, 95% confidence interval [CI] 1.2-22.4) and FVIII recovery of < 85% at the end of ITI (OR 2.6, 95% CI 1.3-5.9) but was not associated with adherence to post-ITI prophylactic FVIII infusion (OR 0.5, 95% CI 0.06-4.3). CONCLUSIONS: The use of immune modulation therapy during ITI and lower FVIII recovery at the end of ITI appear to be associated with an increased risk of inhibitor recurrence after successful ITI. Adherence to post-ITI prophylactic FVIII infusions is not a major determinant of recurrence.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Terapia de Imunossupressão , Isoanticorpos/biossíntese , Criança , Pré-Escolar , Fator VIII/administração & dosagem , Fator VIII/uso terapêutico , Feminino , Hemofilia A/tratamento farmacológico , Humanos , Imunossupressores/uso terapêutico , Lactente , Isoanticorpos/sangue , Isoanticorpos/imunologia , Estimativa de Kaplan-Meier , Modelos Logísticos , Masculino , Adesão à Medicação , Modelos Imunológicos , Plasmaferese , Pontuação de Propensão , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/uso terapêutico , Recidiva , Estudos Retrospectivos , Fatores de Tempo
12.
Endocrinology ; 128(2): 1029-35, 1991 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-1989845

RESUMO

To investigate the mechanism(s) during pubertal development by which androgens alter hypothalamic proopiomelanocortin (POMC) gene expression and beta-endorphin content, we used the technique of in situ hybridization histochemistry and the androgen-insensitive testicular feminized (Tfm) rat. We evaluated POMC mRNA levels in the arcuate nuclei and periarcuate regions of 12 coronal brain slices from prepubertal (age, 30 days) and adult (age, 60 days) normal male and Tfm rats (n = 4 for each group). Hybridizations were performed using an 35S-radiolabeled oligonucleotide probe complementary to a 30-base sequence within POMC mRNA. The tissue sections were sequentially exposed to x-ray film and photographic emulsion with subsequent analysis by both densitometry and computer-assisted grain counting. beta-Endorphin was measured in hypothalamic tissue blocks from similar animals in each of the four experimental groups. The results of densitometry and grain counting were consistent and revealed an increase in POMC mRNA with pubertal development in both the male and Tfm animals. The concentration of hypothalamic beta-endorphin was greater for the adult Tfm animals than for all other groups, which did not differ from each other. These results suggest that androgens may stimulate POMC gene transcription by their action through estrogen receptors after conversion by aromatase. Alternatively, additional pubertal factors may be responsible for act directly through their respective receptors to alter translation, posttranslational processing, or secretion of beta-endorphin, resulting in diminished intracellular hypothalamic peptide concentration.


Assuntos
Regulação da Expressão Gênica , Hipotálamo/metabolismo , Pró-Opiomelanocortina/genética , Receptores Androgênicos/fisiologia , Animais , Peso Corporal , Hormônios Esteroides Gonadais/sangue , Histocitoquímica , Masculino , Hibridização de Ácido Nucleico , Concentração Osmolar , Ratos , beta-Endorfina/metabolismo
13.
Curr Gene Ther ; 3(6): 545-65, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14683451

RESUMO

Recombinant AAV efficacy has been demonstrated in numerous gene therapy preclinical studies. As this vector is increasingly applied to human clinical trials, it is a priority to evaluate the risks of its use for workers involved in research and clinical trials as well as for the patients and their descendants. At high multiplicity of infection, wild-type AAV integrates into human chromosome 19 in approximately 60% of latently infected cell lines. However, it has been recently demonstrated that only approximately 1 out of 1000 infectious units can integrate. The mechanism of this site-specific integration involves AAV Rep proteins which are absent in vectors. Accordingly, recombinant AAV (rAAV) do not integrate site-specifically. Random integration of vector sequences has been demonstrated in established cell lines but only in some cases and at low frequency in primary cultures and in vivo. In contrast, episomal concatemers predominate.Therefore, the risks of insertional mutagenesis and activation of oncogenes are considered low. Biodistribution studies in non-human primates after intramuscular, intrabronchial, hepatic artery and subretinal administration showed low and transient levels of vector DNA in body fluids and distal organs. Analysis of patients body fluids revealed rAAV sequences in urine, saliva and serum at short-term. Transient shedding into the semen has been observed after delivery to the hepatic artery. However, motile germ cells seemed refractory to rAAV infection even when directly exposed to the viral particles, suggesting that the risk of insertion of new genetic material into the germ line is absent or extremely low. Risks related to viral capsid-induced inflammation also seem to be absent since immune response is restricted to generation of antibodies. In contrast, transgene products can elicit both cellular and humoral immune responses, depending on the nature of the expressed protein and of the route of vector administration. Finally, a correlation between early abortion as well as male infertility and the presence of wt AAV DNA in the genital tract has been suggested. Although no causal relationship has been established, this issue stresses the importance of using rAAV stocks devoid of contaminating replication-competent AAV. This review comprehensively examines virus integration, biodistribution, immune interactions, and other safety concerns regarding the wild-type AAV and recombinant AAV vectors.


Assuntos
Dependovirus/genética , Vetores Genéticos/efeitos adversos , Animais , Formação de Anticorpos , Dependovirus/fisiologia , Imunidade Celular , Recombinação Genética , Medição de Risco , Transgenes , Latência Viral
14.
Bone Marrow Transplant ; 28(4): 405-7, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11571515

RESUMO

Recombinant factor VIIa (rFVIIa, NovoSeven) is FDA-approved for the treatment of bleeding in patients with hemophilia A/B with inhibitors. A growing literature suggests that there may be expanded indications for the use of NovoSeven in patients with significant bleeding who do not have a known factor deficiency. Severe bleeding refractory to standard hematologic or hemostatic support is common in patients undergoing bone marrow transplantation (BMT). We review our experience with rFVIIa in three patients (8 years 8 months to 19 years, median 13 years) treated for pulmonary hemorrhage (n = 1), hemorrhagic cystitis (n = 3), and gastrointestinal bleeding (n = 2). Boluses of 90-270 microg/kg rVIIa with subsequent doses of 90 microg/kg every 4-24 h for 3-14 days were given, concurrent with maintaining platelet counts >50,000/mm(3). Transient clinical responses in gross hematuria (two patients) and in pulmonary hemorrhage were noted within several days of starting rFVIIa, but bleeding in a new site in two patients and renewed bleeding of the initial site in the third resulted in discontinuation of the drug. No toxicity or adverse events were observed while the patients were on rFVIIa treatment. Because of the substantial cost of this product, the lack of adequate monitoring methodology, and the variability of current dose and dosing intervals, large randomized studies are needed before definitive off-label use in the setting of BMT can be recommended.


Assuntos
Transplante de Medula Óssea/métodos , Fator VII/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Doença Aguda , Adolescente , Adulto , Anemia Aplástica/complicações , Anemia Aplástica/terapia , Transplante de Medula Óssea/efeitos adversos , Criança , Fator VIIa , Feminino , Hemorragia/tratamento farmacológico , Humanos , Leucemia Mieloide/complicações , Leucemia Mieloide/terapia , Estudos Retrospectivos
16.
J Thromb Haemost ; 11(5): 881-93, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23413986

RESUMO

BACKGROUND: The most common morbidity that results from hemophilia is bleeding-induced hemophilic arthropathy (HA), which once established may not be interrupted completely even by prophylactic clotting factor replacement. Specific therapies to oppose inflammatory cytokines, including Interleukin 6 (IL-6) receptor antagonists, have become important in the management of inflammatory arthritides. OBJECTIVES: We investigated combining therapy using MR16-1, a rat IgG antibody directed against mouse IL-6 receptor (anti-IL-6R), with factor VIII (FVIII) replacement to protect against bleeding-induced arthropathy in hemophilia A mice. METHODS: Three recurrent hemarthroses were induced in the knee joint capsule of FVIII knockout mice. Treatment at the time of each hemorrhage included either: no treatment; FVIII replacement given at the time of hemorrhage; FVIII replacement at hemorrhage plus anti-IL-6R as 4-weekly injections; FVIII replacement with non-specific control antibody (rat IgG); and anti-IL-6R alone without FVIII replacement. Six weeks following the first hemarthosis, joints were harvested and histopathology was scored for synovitis, for cartilage integrity and for macrophage infiltration. RESULTS: Animals that received anti-IL-6R as an adjunct to FVIII replacement demonstrated the best survival and the least acute joint swelling and pathology on histologic examination of the synovium and cartilage (P < 0.05 for each parameter). All histopathologic parameters in the mice receiving FVIII+anti-IL-6R were limited and were comparable to findings in injured hemostatically normal mice. The major benefits of adjunctive anti-IL-6R were decreasing synovial hyperplasia, hemosiderin deposition and macrophage infiltration. CONCLUSIONS: Short-course specific inhibition of inflammatory cytokines as an adjunct to replacement hemostasis may be an approach to minimize hemophilic joint degeneration.


Assuntos
Fator VIII/uso terapêutico , Hemofilia A/complicações , Hemorragia/complicações , Artropatias/prevenção & controle , Receptores de Interleucina-6/antagonistas & inibidores , Animais , Quimioterapia Combinada , Fator VIII/administração & dosagem , Fator VIII/antagonistas & inibidores , Hemofilia A/fisiopatologia , Humanos , Artropatias/etiologia , Camundongos , Camundongos Knockout
18.
J Thromb Haemost ; 7(11): 1843-51, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19583826

RESUMO

BACKGROUND: Factor IX binds to collagen type IV, but this binding has no known consequence. OBJECTIVES: To determine the effect of reduced binding of FIX to collagen IV. METHODS: We constructed and characterized 'knock-in' mice containing the mutation lysine 5 to alanine (K5A) in the Gla domain of their FIX. The K5A mutation dramatically reduced the affinity of FIX for collagen type IV, but had no measurable effect on platelet binding, phospholipid binding, or in vitro clotting activity. However, K5AFIX mice had a mild bleeding tendency, despite their in vitro clotting activity being normal. Hemostatic protection from delayed rebleeding was intermediate between wild-type and hemophilia B mice (which had no detectable clotting activity); moreover, survival of K5A FIX mice after nascent clot removal was dramatically improved as compared with hemophilia B mice. Importantly, there was no detectable difference between K5AFIX and wild-type mice in either a laser-induced thrombosis model or the chromogenic FIX activity assay. In contrast, after ferric chloride injury, which exposes collagen IV as well as other basement membrane proteins, intravital microscopy revealed that vessel occlusion was significantly slower in K5AFIX mice than in wild-type mice. CONCLUSIONS: Our results indicate that the FIX molecule with decreased affinity for collagen IV has altered hemostatic properties in vivo and that the binding of FIX to collagen IV probably plays a significant functional role in hemostasis.


Assuntos
Colágeno Tipo IV/metabolismo , Fator IX/genética , Variação Genética , Hemostasia/genética , Animais , Sítios de Ligação/genética , Fator IX/análise , Técnicas de Introdução de Genes , Hemofilia B , Hemorragia , Camundongos , Ligação Proteica/genética , Trombose
19.
Gene Ther ; 14(5): 429-40, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17066096

RESUMO

Immune responses leading to antibody-mediated elimination of the transgenic protein are a concern in gene replacement for congenital protein deficiencies, for which hemophilia is an important model. Although most hemophilia B patients have circulating non-functional but immunologically crossreactive factor IX (FIX) protein (CRM+ phenotype), inciting factors for FIX neutralizing antibody (inhibitor) development have been studied in crossreactive material-negative (CRM-) animal models. For this study, determinants of FIX inhibitor development were compared in hemophilia B mice, in which circulating FIX protein is absent (CRM- factor IX knockout (FIXKO) model) or present (CRM+ missense R333Q-hFIX model) modeling multiple potential therapies. The investigations compare for the first time different serotypes of adeno-associated virus (AAV) vectors (AAV2 and AAV1), each at multiple doses, in the setting of two different FIX mutations. The comparisons demonstrate in the FIXKO background (CRM- phenotype) that neither vector serotype nor vector particle number independently determine the inhibitor trigger, which is influenced primarily by the level and kinetics of transgene expression. In the CRM+ missense background, inhibitor development was never stimulated by AAV gene therapy or protein therapy, despite the persistence of lymphocytes capable of responding to FIX with non-inhibitory antibodies. This genotype/phenotype is strongly protective against antibody formation in response to FIX therapy.


Assuntos
Autoanticorpos/imunologia , Fator IX/genética , Terapia Genética/efeitos adversos , Vetores Genéticos/genética , Animais , Reações Cruzadas , Fator IX/imunologia , Expressão Gênica , Terapia Genética/métodos , Hemofilia A/sangue , Hemofilia A/imunologia , Hemofilia A/terapia , Humanos , Cinética , Camundongos , Camundongos Knockout , Camundongos Mutantes , Modelos Biológicos , Mutação de Sentido Incorreto , Risco , Transdução Genética/métodos , Transgenes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA