Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
N Engl J Med ; 387(3): 237-247, 2022 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-35857660

RESUMO

BACKGROUND: FLT180a (verbrinacogene setparvovec) is a liver-directed adeno-associated virus (AAV) gene therapy that uses a synthetic capsid and a gain-of-function protein to normalize factor IX levels in patients with hemophilia B. METHODS: In this multicenter, open-label, phase 1-2 trial, we assessed the safety and efficacy of varying doses of FLT180a in patients with severe or moderately severe hemophilia B (factor IX level, ≤2% of normal value). All the patients received glucocorticoids with or without tacrolimus for immunosuppression to decrease the risk of vector-related immune responses. After 26 weeks, patients were enrolled in a long-term follow-up study. The primary end points were safety and efficacy, as assessed by factor IX levels at week 26. RESULTS: Ten patients received one of four FLT180a doses of vector genomes (vg) per kilogram of body weight: 3.84×1011 vg, 6.40×1011 vg, 8.32×1011 vg, or 1.28×1012 vg. After receiving the infusion, all the patients had dose-dependent increases in factor IX levels. At a median follow-up of 27.2 months (range, 19.1 to 42.4), sustained factor IX activity was observed in all the patients except one, who resumed factor IX prophylaxis. As of the data-cutoff date (September 20, 2021), five patients had normal factor IX levels (range, 51 to 78%), three patients had levels from 23 to 43%, and one had a level of 260%. Of the reported adverse events, approximately 10% were related to FLT180a and 24% to immunosuppression. Increases in liver aminotransferase levels were the most common FLT180a-related adverse events. Late increases in aminotransferase levels occurred in patients who had received prolonged tacrolimus beyond the glucocorticoid taper. A serious adverse event of arteriovenous fistula thrombosis occurred in the patient with high factor IX levels. CONCLUSIONS: Sustained factor IX levels in the normal range were observed with low doses of FLT180a but necessitated immunosuppression with glucocorticoids with or without tacrolimus. (Funded by Freeline Therapeutics; ClinicalTrials.gov numbers, NCT03369444 and NCT03641703; EudraCT numbers, 2017-000852-24 and 2017-005080-40.).


Assuntos
Dependovirus , Terapia Genética , Glucocorticoides , Hemofilia B , Dependovirus/genética , Fator IX/análise , Fator IX/genética , Seguimentos , Terapia Genética/efeitos adversos , Terapia Genética/métodos , Vetores Genéticos/uso terapêutico , Glucocorticoides/efeitos adversos , Glucocorticoides/uso terapêutico , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/terapia , Humanos , Imunossupressores/efeitos adversos , Imunossupressores/uso terapêutico , Tacrolimo/efeitos adversos , Tacrolimo/uso terapêutico , Transaminases/análise
2.
Hum Gene Ther ; 33(3-4): 119-130, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34617445

RESUMO

Glucocorticoids have anti-inflammatory and immunosuppressive functions and have commonly been used for preventing liver toxicity after the systemic application of a high dose of adeno-associated virus (AAV) vector for gene therapy. Clinical studies have reported that glucocorticoids have rescued factor IX (FIX) expression in patients with hemophilia B who showed a reduced FIX expression at 6 to 10 weeks post-AAV vector administration. In this study, we explored whether glucocorticoids could affect transgene expression in AAV targeted livers in animal models. When dexamethasone was applied before AAV9/FIX vector administration in the wild-type C57BL/6 mice, FIX expression was much higher than that of the control mice at any time point. More importantly, FIX expression transiently increased after dexamethasone was administered at week 6 or later post-AAV injection regardless of the various dexamethasone treatments applied. The transient enhancement in transgene expression was observed once there were one to several consecutive dexamethasone treatments completed. A similar result was also achieved in other wild-type BALB/c and hemophilia B mice that were treated with AAV9/FIX and dexamethasone. This mechanism study demonstrated that the administration of dexamethasone did not change either AAV genome copy number or transgene expression at the transcription level but transiently decreased interferon beta (IFN-ß) and tumor necrosis factor alpha (TNF-α) expression in the livers of mice at a later time after AAV injection. Next, we studied the effect of dexamethasone on late transgene expression in hemophilia B dogs. Dexamethasone was administered 1 year after AAV9/FIX injection. Inconsistent with the results in mice, no significant change of FIX expression was observed in hemophilia B dogs. In summary, the results from this study indicate that dexamethasone may have various effects on transgene expression in AAV-transduced livers in different species, which provides valuable information about the rational application of dexamethasone in future clinical studies.


Assuntos
Dependovirus , Hemofilia B , Animais , Dependovirus/genética , Dependovirus/metabolismo , Dexametasona/farmacologia , Cães , Fator IX/genética , Vetores Genéticos/genética , Glucocorticoides/metabolismo , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/terapia , Humanos , Fígado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transgenes
3.
Int J Mol Sci ; 22(14)2021 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-34299267

RESUMO

Hemophilia is a monogenic mutational disease affecting coagulation factor VIII or factor IX genes. The palliative treatment of choice is based on the use of safe and effective recombinant clotting factors. Advanced therapies will be curative, ensuring stable and durable concentrations of the defective circulating factor. Results have so far been encouraging in terms of levels and times of expression using mainly adeno-associated vectors. However, these therapies are associated with immunogenicity and hepatotoxicity. Optimizing the vector serotypes and the transgene (variants) will boost clotting efficacy, thus increasing the viability of these protocols. It is essential that both physicians and patients be informed about the potential benefits and risks of the new therapies, and a register of gene therapy patients be kept with information of the efficacy and long-term adverse events associated with the treatments administered. In the context of hemophilia, gene therapy may result in (particularly indirect) cost savings and in a more equitable allocation of treatments. In the case of hemophilia A, further research is needed into how to effectively package the large factor VIII gene into the vector; and in the case of hemophilia B, the priority should be to optimize both the vector serotype, reducing its immunogenicity and hepatotoxicity, and the transgene, boosting its clotting efficacy so as to minimize the amount of vector administered and decrease the incidence of adverse events without compromising the efficacy of the protein expressed.


Assuntos
Terapia Genética/métodos , Hemofilia A/terapia , Hemofilia B/terapia , Animais , Fator IX/genética , Fator IX/metabolismo , Fator VIII/genética , Fator VIII/metabolismo , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/patologia , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/patologia , Humanos
4.
Int J Hematol ; 111(1): 31-41, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30083852

RESUMO

Hemophilia is a congenital hemorrhagic disease caused by genetic abnormalities in coagulation factor VIII or factor IX. Current conventional therapy to prevent bleeding requires frequent intravenous injections of coagulation factor concentrates from early childhood. Accordingly, gene therapy for hemophilia remains an exciting future prospect for patients and their families, due to its potential to cure the disease through a one-time treatment. After a series of successes in basic research, recent clinical trials have demonstrated clear efficacy of gene therapy for hemophilia using adeno-associated virus (AAV) vectors. Although this is likely to alter the paradigm of hemophilia care in the near future, it will be important to overcome immune responses against AAV. Gene therapy for hemophilia cannot be given to patients with anti-AAV capsid-neutralizing antibodies, and cellular immunity with CD8+ T cells should be controlled for sustained expression. Furthermore, long-term therapeutic effects should be closely observed because of the failure of the AAV vector genome to replicate during cell division. This review focuses on the basis of gene therapy, current successes of clinical trials, and the future direction of hemophilia gene therapy.


Assuntos
Dependovirus/genética , Fator IX/genética , Fator VIII/genética , Terapia Genética/métodos , Hemofilia A/genética , Hemofilia A/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Criança , Dependovirus/imunologia , Fator VIII/metabolismo , Edição de Genes/métodos , Expressão Gênica , Vetores Genéticos/imunologia , Vetores Genéticos/uso terapêutico , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/terapia , Hemorragia/sangue , Hemorragia/genética , Humanos
5.
Sci Rep ; 9(1): 14428, 2019 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-31594977

RESUMO

While joint damage is the primary co-morbidity of hemophilia, osteoporosis and osteopenia are also observed. Coagulation factor VIII deficient (FVIII-/-) mice develop an osteoporotic phenotype in the absence of induced hemarthrosis that is exacerbated two weeks after an induced joint injury. Here we have compared comprehensively the bone health of clotting factor VIII, factor IX, and Von Willebrand Factor knockout (FVIII-/-, FIX-/-, and VWF-/- respectively) mice both in the absence of joint hemorrhage and following induced joint injury. We found FVIII-/- and FIX-/- mice, but not VWF-/- mice, developmentally have an osteoporotic phenotype. Unilateral induced hemarthrosis causes further bone damage in both FVIII-/- and FIX-/- mice, but has little effect on VWF-/- bone health, indicating that the FVIII.VWF complex is not required for normal bone remodeling in vivo. To further investigate the bone healing following hemarthrosis in hemophilia we examined a two week time course using microCT, serum chemistry, and histological analysis. Elevated ratio of osteoprotegerin (OPG)/receptor activator of nuclear factor-kappa B ligand (RANKL), increased osterix+ osteoblastic cells, and decreased smoothness of the cortical bone surface were evident within several days of injury, indicative of acute heterotopic mineralization along the cortical surface. This was closely followed by increased interleukin-6 (IL-6) levels, increased osteoclast numbers, and significant trabecular bone loss. Uncoupled and disorganized bone formation and resorption continued for the duration of the study resulting in significant deterioration of the joint. Further elucidation of the shared mechanisms underlying abnormal bone homeostasis in the absence of FVIII or FIX is needed to guide evidence-based approaches to the screening and treatment of the prevalent bone defects in hemophilia A and B.


Assuntos
Fator IX/genética , Fator VIII/genética , Hemofilia A/metabolismo , Hemofilia B/metabolismo , Fator de von Willebrand/genética , Animais , Testes de Coagulação Sanguínea , Osso e Ossos/metabolismo , Hemofilia A/genética , Hemofilia A/patologia , Hemofilia B/genética , Hemofilia B/patologia , Humanos , Interleucina-6/genética , Masculino , Camundongos , Camundongos Knockout , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteoporose/genética , Osteoporose/patologia , Fenótipo , Ligante RANK/genética , Fator de Transcrição Sp7/genética
6.
Hum Gene Ther ; 30(10): 1190-1203, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31347416

RESUMO

In vertebrates, the liver is the central metabolic organ of the body, which carries out an estimated 500 functions that range from general detoxification to protein synthesis, bile production, metabolism of fats, carbohydrates, proteins, bilirubin, vitamin and mineral storage and it even has an immune function. Hepatocytes are considered the professional liver cells, which carry out all of these functions. With such a variety of tasks to perform, it is not surprising that more than 400 rare monogenic disorders of hepatic origin have been described. For many of these, liver transplantation remains the only curative strategy, however, this is limited by organ availability and requires lifelong immune suppression. The fact that liver transplantation is curative led to the assumption that the restoration of the expression of the defective gene would result in the resolution of the disease. Indeed, liver-directed gene therapy trials for hemophilia A and B have demonstrated the potential of gene therapy to provide long-lasting clinical benefit in the treatment of monogenic liver disorders. Thus, liver-directed gene therapy and gene editing strategies have emerged as promising alternatives to transplantation in inherited monogenic liver disorders. Herein, we review the advances and limitations of gene therapy for such disorders, covering therapeutic strategies based on gene addition and gene editing and the exciting clinical results obtained with the use of ribonucleic acid as therapeutic molecules.


Assuntos
Edição de Genes/métodos , Terapia Genética/métodos , Hemofilia A/terapia , Hemofilia B/terapia , Hepatopatias/terapia , Doenças Metabólicas/terapia , Animais , Ensaios Clínicos como Assunto , Dependovirus/genética , Dependovirus/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/patologia , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Fígado/metabolismo , Fígado/patologia , Hepatopatias/genética , Hepatopatias/metabolismo , Hepatopatias/patologia , Transplante de Fígado , Doenças Metabólicas/genética , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia
7.
Cell Rep ; 23(5): 1565-1580, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29719266

RESUMO

Hemophilia B is an ideal target for gene- and cell-based therapies because of its monogenic nature and broad therapeutic index. Here, we demonstrate the use of cell therapy as a potential long-term cure for hemophilia B in our FIX-deficient mouse model. We show that transplanted, cryopreserved, cadaveric human hepatocytes remain functional for more than a year and secrete FIX at therapeutic levels. Hepatocytes from different sources (companies and donors) perform comparably in curing the bleeding defect. We also generated induced pluripotent stem cells (iPSCs) from two hemophilia B patients and corrected the disease-causing mutations in them by two different approaches (mutation specific and universal). These corrected iPSCs were differentiated into hepatocyte-like cells (HLCs) and transplanted into hemophilic mice. We demonstrate these iPSC-HLCs to be viable and functional in mouse models for 9-12 months. This study aims to establish the use of cells from autologous and heterologous sources to treat hemophilia B.


Assuntos
Transplante de Células , Fator IX/metabolismo , Hemofilia B/terapia , Hepatócitos/transplante , Células-Tronco Pluripotentes Induzidas/transplante , Animais , Modelos Animais de Doenças , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/patologia , Hepatócitos/metabolismo , Hepatócitos/patologia , Xenoenxertos , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/patologia , Camundongos , Camundongos Knockout
8.
Haemophilia ; 24(4): 611-618, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29707861

RESUMO

INTRODUCTION: Haemophilia B is caused by a deficiency of coagulation factor IX (FIX) and characterized by bleeding in muscles and joints. In the perioperative setting, patients are treated with FIX replacement therapy to secure haemostasis. Targeting of specified FIX levels is challenging and requires frequent monitoring and adjustment of therapy. AIM: To evaluate perioperative management in haemophilia B, including monitoring of FIX infusions and observed FIX levels, whereby predictors of low and high FIX levels were assessed. METHODS: In this international multicentre study, haemophilia B patients with FIX < 0.05 IU mL-1 undergoing elective, minor or major surgical procedures between 2000 and 2015 were included. Data were collected on patient, surgical and treatment characteristics. Observed FIX levels were compared to target levels as recommended by guidelines. RESULTS: A total of 255 surgical procedures were performed in 118 patients (median age 40 years, median body weight 79 kg). Sixty percent of FIX levels within 24 hours of surgery were below target with a median difference of 0.22 IU mL-1 [IQR 0.12-0.36]; while >6 days after surgery, 59% of FIX levels were above target with a median difference of 0.19 IU mL-1 [IQR 0.10-0.39]. Clinically relevant bleeding complications (necessity of a second surgical intervention or red blood cell transfusion) occurred in 7 procedures (2.7%). CONCLUSION: This study demonstrates that targeting of FIX levels in the perioperative setting is complex and suboptimal, but although this bleeding is minimal. Alternative dosing strategies taking patient and surgical characteristics as well as pharmacokinetic principles into account may help to optimize and individualize treatment.


Assuntos
Fator IX/uso terapêutico , Hemofilia B/tratamento farmacológico , Hemofilia B/cirurgia , Período Perioperatório , Adulto , Criança , Pré-Escolar , Fator IX/metabolismo , Feminino , Hemofilia B/metabolismo , Hemorragia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Trombose/etiologia , Adulto Jovem
9.
N Engl J Med ; 377(23): 2215-2227, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29211678

RESUMO

BACKGROUND: The prevention of bleeding with adequately sustained levels of clotting factor, after a single therapeutic intervention and without the need for further medical intervention, represents an important goal in the treatment of hemophilia. METHODS: We infused a single-stranded adeno-associated viral (AAV) vector consisting of a bioengineered capsid, liver-specific promoter and factor IX Padua (factor IX-R338L) transgene at a dose of 5×1011 vector genomes per kilogram of body weight in 10 men with hemophilia B who had factor IX coagulant activity of 2% or less of the normal value. Laboratory values, bleeding frequency, and consumption of factor IX concentrate were prospectively evaluated after vector infusion and were compared with baseline values. RESULTS: No serious adverse events occurred during or after vector infusion. Vector-derived factor IX coagulant activity was sustained in all the participants, with a mean (±SD) steady-state factor IX coagulant activity of 33.7±18.5% (range, 14 to 81). On cumulative follow-up of 492 weeks among all the participants (range of follow-up in individual participants, 28 to 78 weeks), the annualized bleeding rate was significantly reduced (mean rate, 11.1 events per year [range, 0 to 48] before vector administration vs. 0.4 events per year [range, 0 to 4] after administration; P=0.02), as was factor use (mean dose, 2908 IU per kilogram [range, 0 to 8090] before vector administration vs. 49.3 IU per kilogram [range, 0 to 376] after administration; P=0.004). A total of 8 of 10 participants did not use factor, and 9 of 10 did not have bleeds after vector administration. An asymptomatic increase in liver-enzyme levels developed in 2 participants and resolved with short-term prednisone treatment. One participant, who had substantial, advanced arthropathy at baseline, administered factor for bleeding but overall used 91% less factor than before vector infusion. CONCLUSIONS: We found sustained therapeutic expression of factor IX coagulant activity after gene transfer in 10 participants with hemophilia who received the same vector dose. Transgene-derived factor IX coagulant activity enabled the termination of baseline prophylaxis and the near elimination of bleeding and factor use. (Funded by Spark Therapeutics and Pfizer; ClinicalTrials.gov number, NCT02484092 .).


Assuntos
Fator IX/genética , Terapia Genética/métodos , Vetores Genéticos , Hemofilia B/terapia , Transgenes , Adolescente , Adulto , Dependovirus/imunologia , Fator IX/metabolismo , Fator IX/uso terapêutico , Vetores Genéticos/administração & dosagem , Hemofilia B/genética , Hemofilia B/metabolismo , Hemorragia/prevenção & controle , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
11.
Mol Ther ; 25(5): 1163-1167, 2017 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-28411016

RESUMO

The X-linked bleeding disorder hemophilia causes frequent and exaggerated bleeding that can be life-threatening if untreated. Conventional therapy requires frequent intravenous infusions of the missing coagulation protein (factor VIII [FVIII] for hemophilia A and factor IX [FIX] for hemophilia B). However, a lasting cure through gene therapy has long been sought. After a series of successes in small and large animal models, this goal has finally been achieved in humans by in vivo gene transfer to the liver using adeno-associated viral (AAV) vectors. In fact, multiple recent clinical trials have shown therapeutic, and in some cases curative, expression. At the same time, cellular immune responses against the virus have emerged as an obstacle in humans, potentially resulting in loss of expression. Transient immune suppression protocols have been developed to blunt these responses. Here, we provide an overview of the clinical development of AAV gene transfer for hemophilia, as well as an outlook on future directions.


Assuntos
Fator IX/genética , Fator VIII/genética , Terapia Genética/métodos , Hemofilia A/terapia , Hemofilia B/terapia , Transfusão de Sangue , Dependovirus/genética , Dependovirus/imunologia , Fator IX/metabolismo , Fator VIII/metabolismo , Expressão Gênica , Terapia Genética/tendências , Vetores Genéticos/química , Vetores Genéticos/imunologia , Hemofilia A/genética , Hemofilia A/metabolismo , Hemofilia A/patologia , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/patologia , Humanos , Lentivirus/genética , Lentivirus/imunologia , Mutação
12.
Blood ; 129(15): 2161-2171, 2017 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-28039188

RESUMO

Wound healing requires interactions between coagulation, inflammation, angiogenesis, cellular migration, and proliferation. Healing in dermal wounds of hemophilia B mice is delayed when compared with hemostatically normal wild-type (WT) mice, with abnormal persistence of iron deposition, inflammation, and neovascularity. We observed healing following induced joint hemorrhage in WT and factor IX (FIX) knockout (FIX-/-) mice, examining also parameters previously studied in an excisional skin wound model. Hemostatically normal mice tolerated this joint bleeding challenge, cleared blood from the joint, and healed with minimal pathology, even if additional autologous blood was injected intra-articularly at the time of wounding. Following hemarthrosis, joint wound healing in hemophilia B mice was impaired and demonstrated similar abnormal histologic features as previously described in hemophilic dermal wounds. Therefore, studies of pathophysiology and therapy of hemophilic joint bleeding performed in hemostatically normal animals are not likely to accurately reflect the healing defect of hemophilia. We additionally explored the hypothesis that the use of a FIX replacement protein with extended circulating FIX activity could improve synovial and osteochondral wound healing in hemophilic mice, when compared with treatment with unmodified recombinant FIX (rFIX) in the established joint bleeding model. Significantly improved synovial wound healing and preservation of normal osteochondral architecture are achieved by extending FIX activity after hemarthrosis using glycoPEGylated FIX when compared with an equivalent dose of rFIX. These results suggest that treating joint bleeding only until hemostasis is achieved may not result in optimal joint healing, which is improved by extending factor activity.


Assuntos
Fator IX , Hemartrose , Hemofilia B , Articulações , Pele , Cicatrização , Animais , Modelos Animais de Doenças , Fator IX/genética , Fator IX/farmacologia , Hemartrose/tratamento farmacológico , Hemartrose/genética , Hemartrose/metabolismo , Hemofilia B/tratamento farmacológico , Hemofilia B/genética , Hemofilia B/metabolismo , Articulações/lesões , Articulações/metabolismo , Camundongos , Camundongos Knockout , Pele/lesões , Pele/metabolismo , Cicatrização/efeitos dos fármacos , Cicatrização/genética
13.
Biochemistry ; 54(24): 3814-21, 2015 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-26023895

RESUMO

We recently identified two hemophilia B patients who carried Gly-317 to Arg (FIX-G317R) or Gly-317 to Glu (FIX-G317E) substitutions in their FIX gene. The former mutation caused severe and the latter moderate bleeding in afflicted patients. To understand the molecular basis for the variable clinical manifestation of Gly-317 mutations, we prepared recombinant G317R and G317E derivatives of FIX and compared their kinetic properties to those of recombinant wild-type FIX in appropriate assay systems. Both physiological activators, factor XIa and extrinsic Tenase (factor VIIa-tissue factor), activated both zymogen variants with an ∼1.5-fold elevated K(m); however, extrinsic Tenase activated FIX-G317E with an ∼2-fold improved k(cat). By contrast to zymogen activation, the catalytic activities of both FIXa-G317R and FIXa-G317E enzymes toward the natural substrate, factor X, were dramatically (>4 orders of magnitude) impaired, but their apparent affinity for interaction with factor VIIIa was only slightly (<2-fold) decreased. Further studies revealed that the reactivity of FIXa-G317R and FIXa-G317E with antithrombin has been impaired 10- and 13-fold, respectively, in the absence and 166- and 500-fold, respectively, in the presence of pentasaccharide. As expected, the clotting activities of FIX variants could not be measured by the aPTT assay. These results implicate a critical role for Gly-317 in maintaining normal catalytic function for FIX/FIXa in the clotting cascade. The results further suggest that improved k(cat) of FIX-G317E activation in the extrinsic pathway together with dramatically impaired reactivity of FIXa-G317E with antithrombin may account for the less severe bleeding phenotype of a hemophilia B patient carrying the FIX-G317E mutation.


Assuntos
Precursores Enzimáticos/metabolismo , Fator IX/metabolismo , Glicina/química , Hemofilia B/genética , Hemorragia/etiologia , Proteínas Mutantes/metabolismo , Mutação , Substituição de Aminoácidos , Cisteína Endopeptidases/metabolismo , Ativação Enzimática , Precursores Enzimáticos/genética , Fator IX/genética , Fator VIIIa/metabolismo , Fator X/metabolismo , Fator XIa/metabolismo , Células HEK293 , Hemofilia B/metabolismo , Hemofilia B/fisiopatologia , Humanos , Cinética , Masculino , Mutagênese Sítio-Dirigida , Proteínas de Neoplasias/metabolismo , Proteínas Recombinantes/metabolismo , Índice de Gravidade de Doença
14.
Hum Gene Ther Clin Dev ; 26(1): 5-14, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25675273

RESUMO

Preclinical testing of new therapeutic strategies in relevant animal models is an essential part of drug development. The choice of animal models of disease that are used in these studies is driven by the strength of the translational data for informing about safety, efficacy, and success or failure of human clinical trials. Hemophilia B is a monogenic, X-linked, inherited bleeding disorder that results from absent or dysfunctional coagulation factor IX (FIX). Regarding preclinical studies of adeno-associated virus (AAV)-mediated gene therapy for hemophilia B, dogs with severe hemophilia B (<1% FIX) provide well-characterized phenotypes and genotypes in which a species-specific transgene can be expressed in a mixed genetic background. Correction of the hemophilic coagulopathy by sustained expression of FIX, reduction of bleeding events, and a comprehensive assessment of the humoral and cell-mediated immune responses to the expressed transgene and recombinant AAV vector are all feasible end points in these dogs. This review compares the preclinical studies of AAV vectors used to treat dogs with hemophilia B with the results obtained in subsequent human clinical trials using muscle- and liver-based approaches.


Assuntos
Dependovirus/genética , Terapia Genética , Hemofilia B/terapia , Animais , Modelos Animais de Doenças , Cães , Hemofilia B/metabolismo , Humanos , Fígado/metabolismo , Músculo Esquelético/metabolismo
15.
Hum Mol Genet ; 21(11): 2389-98, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22362925

RESUMO

A significant proportion of disease-causing mutations affect precursor-mRNA splicing, inducing skipping of the exon from the mature transcript. Using F9 exon 5, CFTR exon 12 and SMN2 exon 7 models, we characterized natural mutations associated to exon skipping in Haemophilia B, cystic fibrosis and spinal muscular atrophy (SMA), respectively, and the therapeutic splicing rescue by using U1 small nuclear RNA (snRNA). In minigene expression systems, loading of U1 snRNA by complementarity to the normal or mutated donor splice sites (5'ss) corrected the exon skipping caused by mutations at the polypyrimidine tract of the acceptor splice site, at the consensus 5'ss or at exonic regulatory elements. To improve specificity and reduce potential off-target effects, we developed U1 snRNA variants targeting non-conserved intronic sequences downstream of the 5'ss. For each gene system, we identified an exon-specific U1 snRNA (ExSpeU1) able to rescue splicing impaired by the different types of mutations. Through splicing-competent cDNA constructs, we demonstrated that the ExSpeU1-mediated splicing correction of several F9 mutations results in complete restoration of secreted functional factor IX levels. Furthermore, two ExSpeU1s for SMA improved SMN exon 7 splicing in the chromosomal context of normal cells. We propose ExSpeU1s as a novel therapeutic strategy to correct, in several human disorders, different types of splicing mutations associated with defective exon definition.


Assuntos
Éxons , RNA Nuclear Pequeno/química , RNA Nuclear Pequeno/metabolismo , Processamento Alternativo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Hemofilia B/genética , Hemofilia B/metabolismo , Humanos , Íntrons , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/metabolismo , Mutação , Precursores de RNA/genética , Precursores de RNA/metabolismo , Sítios de Splice de RNA , Splicing de RNA/fisiologia
16.
Blood ; 119(13): 3038-41, 2012 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-22271447

RESUMO

In previous work we transferred a human factor IX-encoding adeno-associated viral vector (AAV) into skeletal muscle of men with severe hemophilia B. Biopsy of injected muscle up to 1 year after vector injection showed evidence of gene transfer by Southern blot and of protein expression by IHC and immunofluorescent staining. Although the procedure appeared safe, circulating F.IX levels remained subtherapeutic (< 1%). Recently, we obtained muscle tissue from a subject injected 10 years earlier who died of causes unrelated to gene transfer. Using Western blot, IHC, and immunofluorescent staining, we show persistent factor IX expression in injected muscle tissue. F.IX transcripts were detected in injected skeletal muscle using RT-PCR, and isolated whole genomic DNA tested positive for the presence of the transferred AAV vector sequence. This is the longest reported transgene expression to date from a parenterally administered AAV vector, with broad implications for the future of muscle-directed gene transfer.


Assuntos
Dependovirus/genética , Fator IX/genética , Terapia Genética/métodos , Hemofilia B/terapia , Músculo Esquelético/metabolismo , Adulto , Dependovirus/fisiologia , Fator IX/metabolismo , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Hemofilia B/genética , Hemofilia B/metabolismo , Hemofilia B/patologia , Humanos , Masculino , Músculo Esquelético/patologia , Especificidade de Órgãos/genética , Índice de Gravidade de Doença , Fatores de Tempo
18.
J Gene Med ; 13(7-8): 365-72, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21710610

RESUMO

BACKGROUND: Long-term expression of the delivered target gene is critical for successful gene therapy. Recently, hepatic control region I (HCR I) originating from the apolipoprotein (apo)C-I pseudogene was shown to be a critical element for long-term gene expression in the liver of mice. HCR II is another hepatic control region of apoC-I. METHODS: HCR I, HCR II and HCR I/II-containing plasmids encoding factor IX were prepared and hydrodynamically transferred into the liver of normal and hemophilia B mice. Factor IX expression, clotting activity and formation of antibodies against the expressed gene product were compared. RESULTS: HCR I-, HCR II- and HCR I/II-containing plasmids all induced long-term gene expression in both normal and hemophilia B mice. Post-transfection factor IX expression in the hemophilia B mice remained above 500 ng/ml for 210 days. Antibodies against human factor IX were detected at a low level in the serum, although they had no effect on the levels and clotting activity of the expressed factor IX. CONCLUSIONS: We have shown in mouse models that hydrodynamic transfection of pBS-HCRII-HP-FIXA and pBS-HCRI/II-HP-FIXA was able to induce and maintain the expression and clotting activity of human factor IX for a long period of time at a potentially therapeutic level. With an appropriate delivery system, this type of plasmid vector could be clinically useful for the hepatic expression of therapeutic genes including human factor IX.


Assuntos
Fator IX/genética , Fator IX/metabolismo , Hemofilia B/genética , Hemofilia B/metabolismo , Fígado/metabolismo , Elementos Reguladores de Transcrição , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Linhagem Celular Tumoral , Dependovirus/genética , Modelos Animais de Doenças , Fator IX/imunologia , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Hemofilia B/terapia , Células Hep G2 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Especificidade de Órgãos/genética , Tempo de Tromboplastina Parcial , Regiões Promotoras Genéticas
19.
Mol Ther ; 18(7): 1318-29, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20424599

RESUMO

Muscle represents an attractive target tissue for adeno-associated virus (AAV) vector-mediated gene transfer for hemophilia B (HB). Experience with direct intramuscular (i.m.) administration of AAV vectors in humans showed that the approach is safe but fails to achieve therapeutic efficacy. Here, we present a careful evaluation of the safety profile (vector, transgene, and administration procedure) of peripheral transvenular administration of AAV-canine factor IX (cFIX) vectors to the muscle of HB dogs. Vector administration resulted in sustained therapeutic levels of cFIX expression. Although all animals developed a robust antibody response to the AAV capsid, no T-cell responses to the capsid antigen were detected by interferon (IFN)-gamma enzyme-linked immunosorbent spot (ELISpot). Interleukin (IL)-10 ELISpot screening of lymphocytes showed reactivity to cFIX-derived peptides, and restimulation of T cells in vitro in the presence of the identified cFIX epitopes resulted in the expansion of CD4(+)FoxP3(+)IL-10(+) T-cells. Vector administration was not associated with systemic inflammation, and vector spread to nontarget tissues was minimal. At the local level, limited levels of cell infiltrates were detected when the vector was administered intravascularly. In summary, this study in a large animal model of HB demonstrates that therapeutic levels of gene transfer can be safely achieved using a novel route of intravascular gene transfer to muscle.


Assuntos
Dependovirus/genética , Fator IX/genética , Vetores Genéticos/efeitos adversos , Vetores Genéticos/genética , Hemofilia B/terapia , Músculo Esquelético/metabolismo , Animais , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Cães , Fator IX/metabolismo , Citometria de Fluxo , Hemofilia B/metabolismo , Humanos , Imunoglobulina G/metabolismo , Interferon gama/metabolismo , Interleucina-10/metabolismo , Músculo Esquelético/patologia
20.
Hum Gene Ther ; 20(5): 479-85, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19196177

RESUMO

Although the desire to develop gene therapy for hemophilia B is high, safety remains a concern. Therefore, improving the therapeutic index of gene therapy vectors is an important goal. Thus, we evaluated the use of three bioengineered factor IX (FIX) variants with improved catalytic activity in the context of the helper-dependent adenoviral vector. The first vector expressed R338A-FIX, an FIX variant with the arginine at position 338 changed to an alanine, which resulted in a 2.9-fold higher specific activity (IU/mg) compared with the wild-type FIX. The second vector expressed FIX(VIIEGF1), a variant with the EGF-1 domain replaced with the EGF-1 domain from FVII, which resulted in a 3.4-fold increase in specific activity. The third expressed R338A + FIX(VIIEGF1), a novel variant containing both aforementioned modifications, which resulted in a 12.6-fold increase in specific activity. High-level, long-term, and stable expression of these three variants was observed in hemophilia B mice with no evidence of increased thrombogenicity compared with wild-type FIX. Thus, these bioengineered FIX variants can increase the therapeutic index of gene therapy vectors by permitting administration of lower doses to achieve the same therapeutic outcome. Furthermore, these variants may also be valuable for recombinant FIX protein replacement therapy.


Assuntos
Fator IX/genética , Fator IX/imunologia , Vetores Genéticos/imunologia , Hemofilia B/terapia , Animais , Fator IX/metabolismo , Terapia Genética , Vetores Genéticos/metabolismo , Hemofilia B/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Estrutura Terciária de Proteína/genética , Trombose/imunologia , Transdução Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA