Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
N Engl J Med ; 388(8): 706-718, 2023 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-36812434

RESUMO

BACKGROUND: Moderate-to-severe hemophilia B is treated with lifelong, continuous coagulation factor IX replacement to prevent bleeding. Gene therapy for hemophilia B aims to establish sustained factor IX activity, thereby protecting against bleeding without burdensome factor IX replacement. METHODS: In this open-label, phase 3 study, after a lead-in period (≥6 months) of factor IX prophylaxis, we administered one infusion of adeno-associated virus 5 (AAV5) vector expressing the Padua factor IX variant (etranacogene dezaparvovec; 2×1013 genome copies per kilogram of body weight) to 54 men with hemophilia B (factor IX activity ≤2% of the normal value) regardless of preexisting AAV5 neutralizing antibodies. The primary end point was the annualized bleeding rate, evaluated in a noninferiority analysis comparing the rate during months 7 through 18 after etranacogene dezaparvovec treatment with the rate during the lead-in period. Noninferiority of etranacogene dezaparvovec was defined as an upper limit of the two-sided 95% Wald confidence interval of the annualized bleeding rate ratio that was less than the noninferiority margin of 1.8. Superiority, additional efficacy measures, and safety were also assessed. RESULTS: The annualized bleeding rate decreased from 4.19 (95% confidence interval [CI], 3.22 to 5.45) during the lead-in period to 1.51 (95% CI, 0.81 to 2.82) during months 7 through 18 after treatment, for a rate ratio of 0.36 (95% Wald CI, 0.20 to 0.64; P<0.001), demonstrating noninferiority and superiority of etranacogene dezaparvovec as compared with factor IX prophylaxis. Factor IX activity had increased from baseline by a least-squares mean of 36.2 percentage points (95% CI, 31.4 to 41.0) at 6 months and 34.3 percentage points (95% CI, 29.5 to 39.1) at 18 months after treatment, and usage of factor IX concentrate decreased by a mean of 248,825 IU per year per participant in the post-treatment period (P<0.001 for all three comparisons). Benefits and safety were observed in participants with predose AAV5 neutralizing antibody titers of less than 700. No treatment-related serious adverse events occurred. CONCLUSIONS: Etranacogene dezaparvovec gene therapy was superior to prophylactic factor IX with respect to the annualized bleeding rate, and it had a favorable safety profile. (Funded by uniQure and CSL Behring; HOPE-B ClinicalTrials.gov number, NCT03569891.).


Assuntos
Fator IX , Terapia Genética , Hemofilia B , Humanos , Masculino , Fator IX/genética , Fator IX/uso terapêutico , Terapia Genética/métodos , Hemofilia B/complicações , Hemofilia B/genética , Hemofilia B/terapia , Hemorragia/etiologia , Hemorragia/terapia , Vetores Genéticos/administração & dosagem
2.
N Engl J Med ; 385(21): 1961-1973, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34788507

RESUMO

BACKGROUND: The goal of gene therapy for patients with hemophilia A is to safely impart long-term stable factor VIII expression that predictably ameliorates bleeding with the use of the lowest possible vector dose. METHODS: In this phase 1-2 trial, we infused an investigational adeno-associated viral (AAV) vector (SPK-8011) for hepatocyte expression of factor VIII in 18 men with hemophilia A. Four dose cohorts were enrolled; the lowest-dose cohort received a dose of 5 × 1011 vector genomes (vg) per kilogram of body weight, and the highest-dose cohort received 2 × 1012 vg per kilogram. Some participants received glucocorticoids within 52 weeks after vector administration either to prevent or to treat a presumed AAV capsid immune response. Trial objectives included evaluation of the safety and preliminary efficacy of SPK-8011 and of the expression and durability of factor VIII. RESULTS: The median safety observation period was 36.6 months (range, 5.5 to 50.3). A total of 33 treatment-related adverse events occurred in 8 participants; 17 events were vector-related, including 1 serious adverse event, and 16 were glucocorticoid-related. Two participants lost all factor VIII expression because of an anti-AAV capsid cellular immune response that was not sensitive to immune suppression. In the remaining 16 participants, factor VIII expression was maintained; 12 of these participants were followed for more than 2 years, and a one-stage factor VIII assay showed no apparent decrease in factor VIII activity over time (mean [±SD] factor VIII activity, 12.9±6.9% of the normal value at 26 to 52 weeks when the participants were not receiving glucocorticoids vs. 12.0±7.1% of the normal value at >52 weeks after vector administration; 95% confidence interval [CI], -2.4 to 0.6 for the difference between matched pairs). The participants had a 91.5% reduction (95% CI, 88.8 to 94.1) in the annualized bleeding rate (median rate, 8.5 events per year [range, 0 to 43.0] before vector administration vs. 0.3 events per year [range, 0 to 6.5] after vector administration). CONCLUSIONS: Sustained factor VIII expression in 16 of 18 participants who received SPK-8011 permitted discontinuation of prophylaxis and a reduction in bleeding episodes. No major safety concerns were reported. (Funded by Spark Therapeutics and the National Heart, Lung, and Blood Institute; ClinicalTrials.gov numbers, NCT03003533 and NCT03432520.).


Assuntos
Dependovirus , Fator VIII/genética , Fator VIII/metabolismo , Terapia Genética , Vetores Genéticos , Hemofilia A/sangue , Adolescente , Adulto , Seguimentos , Genótipo , Glucocorticoides/efeitos adversos , Glucocorticoides/uso terapêutico , Hemofilia A/genética , Hemofilia A/prevenção & controle , Hepatócitos/metabolismo , Humanos , Terapia de Imunossupressão , Masculino , Pessoa de Meia-Idade , Adulto Jovem
3.
Haemophilia ; 30(1): 75-86, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37902714

RESUMO

INTRODUCTION: Etranacogene dezaparvovec gene therapy for haemophilia B demonstrated superior efficacy at 24 months in reducing bleeds versus a ≥6-month lead-in period of prophylaxis with FIX products in the phase 3 trial, HOPE-B. In the absence of head-to-head comparisons of etranacogene dezaparvovec versus FIX products, indirect treatment comparisons (ITC) can be used. AIM: To compare the efficacy of etranacogene dezaparvovec versus rIX-FP, rFIXFc and N9-GP using ITC, and support HOPE-B results. METHODS: Data were leveraged from Phase 3 pivotal trials: HOPE-B, PROLONG-9FP, B-LONG and Paradigm 2. Annualised bleeding rates (ABR), spontaneous (AsBR) and joint (AjBR) bleeding rates, percentage of patients with no bleeds, and FIX consumption were assessed using inverse probability of treatment weighting and matching adjusted indirect comparisons. RESULTS: Etranacogene dezaparvovec demonstrated statistically significantly lower bleeding rates versus all comparators. Rate ratios for ABR, AsBR and AjBR versus rIX-FP were 0.19 (p < .0001), 0.08 (p < .0001) and 0.09 (p < .0001), respectively. Rate ratios for ABR, AsBR and AjBR versus rFIXFc were 0.14 (p < .0001), 0.13 (p = .0083) and 0.15 (p = .0111), respectively. Rate ratios for ABR and AsBR, versus N9-GP were 0.24 (p = .0231) and 0.13 (p = .0071), respectively. Etranacogene dezaparvovec demonstrated significantly higher percentage of patients with no bleeds versus rIX-FP and rFIXFc; odds ratios: 17.60 (p < .0001) and 5.65 (p = .0037), respectively. Etranacogene dezaparvovec resulted in significantly lower FIX consumption than all comparators. CONCLUSIONS: ITC suggests that etranacogene dezaparvovec offers patients with haemophilia B (≤2% of normal FIX expression) a single dose treatment that can significantly reduce bleeding rates and eliminate routine infusions associated with FIX therapies.


Assuntos
Fator IX , Hemofilia B , Humanos , Fator IX/genética , Fator IX/uso terapêutico , Hemofilia B/tratamento farmacológico , Hemofilia B/genética , Meia-Vida , Hemorragia/complicações , Terapia Genética , Proteínas Recombinantes de Fusão/uso terapêutico
4.
Haemophilia ; 30(3): 709-719, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38462823

RESUMO

INTRODUCTION: For people with haemophilia B (PwHB), bleeding may occur despite prophylaxis, negatively affecting health-related quality of life (HRQoL). The pivotal phase 3 HOPE-B trial investigating the adeno-associated virus gene transfer product, etranacogene dezaparvovec (EDZ), demonstrated sustained factor IX (FIX) activity and bleed protection in PwHB with baseline FIX levels ≤2%. AIM: Assess how EDZ affects HRQoL in HOPE-B trial participants. METHODS: HRQoL was evaluated using generic and disease-specific patient reported outcomes (PROs) including the EQ-5D-5L and the Hem-A-QoL questionnaires. Mean domain and total scores were compared 6 months pre- and the first 2 years post-EDZ administration using repeated measures linear mixed models. The percentage of participants with minimal clinically important improvements in HRQoL was also evaluated. RESULTS: Two years post-EDZ, there were nominally significant increases in the least squares (LS) mean score for the EQ-5D-5L Index Value (.04; p = .0129), reflecting better HRQoL. Nominally significant decreases in the LS mean scores, reflecting better HRQoL, were also found for the Hem-A-QoL total score (-6.0; p < .0001) and the Treatment (-13.94; p < .0001), Feelings (-9.01; p < .0001), Future (-6.45; p = .0004) and Work/School (-5.21; p = .0098) domains. The percentage of participants with ≥15-point improvement ranged from 45.83% (95% CI: 31.37%, 60.83%) for Treatment to 13.89% (95% CI: 4.67%, 29.50%) for Family Planning. Results were similar for Year 1. CONCLUSION: In conclusion, gene therapy with EDZ improved HRQoL in the first and second years in several Hem-A-QoL domains, including Treatment, Feelings, Work/School and Future domains, whereas improvement in other aspects of HRQoL were not demonstrated.


Assuntos
Terapia Genética , Hemofilia B , Qualidade de Vida , Humanos , Hemofilia B/psicologia , Hemofilia B/terapia , Terapia Genética/métodos , Masculino , Adulto , Pessoa de Meia-Idade , Adulto Jovem , Fator IX/uso terapêutico , Adolescente , Feminino , Dependovirus/genética , Inquéritos e Questionários , Índice de Gravidade de Doença
5.
Blood ; 137(6): 763-774, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33067633

RESUMO

Gene therapy has the potential to maintain therapeutic blood clotting factor IX (FIX) levels in patients with hemophilia B by delivering a functional human F9 gene into liver cells. This phase 1/2, open-label dose-escalation study investigated BAX 335 (AskBio009, AAV8.sc-TTR-FIXR338Lopt), an adeno-associated virus serotype 8 (AAV8)-based FIX Padua gene therapy, in patients with hemophilia B. This report focuses on 12-month interim analyses of safety, pharmacokinetic variables, effects on FIX activity, and immune responses for dosed participants. Eight adult male participants (aged 20-69 years; range FIX activity, 0.5% to 2.0%) received 1 of 3 BAX 335 IV doses: 2.0 × 1011; 1.0 × 1012; or 3.0 × 1012 vector genomes/kg. Three (37.5%) participants had 4 serious adverse events, all considered unrelated to BAX 335. No serious adverse event led to death. No clinical thrombosis, inhibitors, or other FIX Padua-directed immunity was reported. FIX expression was measurable in 7 of 8 participants; peak FIX activity displayed dose dependence (32.0% to 58.5% in cohort 3). One participant achieved sustained therapeutic FIX activity of ∼20%, without bleeding or replacement therapy, for 4 years; in others, FIX activity was not sustained beyond 5 to 11 weeks. In contrast to some previous studies, corticosteroid treatment did not stabilize FIX activity loss. We hypothesize that the loss of transgene expression could have been caused by stimulation of innate immune responses, including CpG oligodeoxynucleotides introduced into the BAX 335 coding sequence by codon optimization. This trial was registered at www.clinicaltrials.gov as #NCT01687608.


Assuntos
Ilhas de CpG/genética , Fator IX/uso terapêutico , Regulação da Expressão Gênica , Terapia Genética , Hemofilia B/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Adolescente , Adulto , Idoso , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Fator IX/biossíntese , Fator IX/genética , Mutação com Ganho de Função , Hemofilia B/genética , Hemofilia B/imunologia , Humanos , Imunidade Inata , Masculino , Pessoa de Meia-Idade , Moléculas com Motivos Associados a Patógenos/imunologia , Estudos Prospectivos , Rabdomiólise/etiologia , Receptor Toll-Like 9/fisiologia , Transgenes , Adulto Jovem
7.
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
8.
Blood ; 127(24): 3073-81, 2016 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-26983851

RESUMO

The availability of longitudinal data collected prospectively from 1998 to 2011 at federally funded US hemophilia treatment centers provided an opportunity to construct a descriptive analysis of how outcomes of men with severe hemophilia have been altered by the incremental advances and setbacks in hemophilia care in the last 50 years in the United States. This surveillance collaboration with the US Centers for Disease Control and Prevention assembled the largest uniformly examined population with severe hemophilia (n = 4899 men with severe factor VIII and IX deficiency). To address the heterogeneity of this population, 4 successive birth cohorts, differentially affected by eras of hemophilia care, were examined separately in regard to demographics, complications of hemophilia and its treatment, and mortality. Severely affected men in each birth cohort were compared also with the corresponding mild hemophilia birth cohorts (n = 2587 men total) to control for outcomes that might be attributable to aging and environment independent of severely defective hemostasis. The analysis demonstrates improving access to standard of care therapy, correlating the proportion of men on prophylactic factor replacement and reduced bleeding frequency for the youngest men. Frequent bleeding persisted in one third to one half of men across all ages, however, and the disability gap between severe and mild hemophilia did not narrow. The greatest cause of death was liver failure, but attempted anti-hepatitis C virus therapy and cure were low. The study suggests a continued need for national surveillance to monitor and inform hemophilia interventions and outcomes.


Assuntos
Hemofilia A/epidemiologia , Adulto , Pré-Escolar , Estudos de Coortes , Bases de Dados Factuais/estatística & dados numéricos , Acessibilidade aos Serviços de Saúde/estatística & dados numéricos , Hemofilia A/complicações , Hemofilia A/diagnóstico , Hemofilia A/terapia , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Triagem Neonatal , Índice de Gravidade de Doença , Fatores Sexuais , Estados Unidos/epidemiologia
9.
Blood ; 128(2): 286-92, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27106122

RESUMO

FIX binds tightly to collagen IV. Furthermore, a FIX mutant, FIXK5R, which binds better than wild-type FIX to collagen IV, provides better hemostasis than wild-type FIX, long after both are undetectable in the plasma. There is also credible evidence of extravascular FIX. Here, we use the saphenous vein bleeding model to compare the efficacy of recombinant FIXFc (Alprolix) and wild-type FIX (BeneFIX) in hemophilia B mice 7 days postinfusion. Although the terminal half-life of Alprolix is significantly longer than that of BeneFIX, at equal doses Alprolix is not better at controlling bleeding 7 days postinfusion, presumably because of the extravascular FIX. Both BeneFIX and Alprolix exhibit a linear response in clotting efficacy up to 150 IU/kg, where they appear to saturate an extravascular compartment, because there is no additional prophylactic benefit from higher doses. A robust pool of extravascular FIX is clearly observed surrounding blood vessels, localized to the same region as collagen IV, in 2 representative human tissues: liver and skeletal muscle. We see no increased risk for thrombosis at 250 IU/kg FIX at 6 hours postinfusion. In summary, 7 days postinfusion into hemophilia B mice, BeneFIX and Alprolix are hemostatically indistinguishable despite the latter's increased half-life. We predict that doses of FIX ∼3 times higher than the currently recommended 40 to 50 IU/kg will, because of FIX's large extravascular compartment, efficiently prolong prophylactic hemostasis without thrombotic risk.


Assuntos
Fator IX , Hemofilia B , Hemorragia , Fragmentos Fc das Imunoglobulinas , Proteínas Recombinantes de Fusão , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Fator IX/farmacocinética , Fator IX/farmacologia , Hemofilia B/sangue , Hemofilia B/tratamento farmacológico , Hemorragia/sangue , Hemorragia/prevenção & controle , Fragmentos Fc das Imunoglobulinas/farmacologia , Camundongos , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/farmacologia
10.
Mol Ther ; 22(3): 567-574, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23941813

RESUMO

Integration-deficient lentiviral vectors (IDLVs) have been shown to transduce a wide spectrum of target cells and organs in vitro and in vivo and to maintain long-term transgene expression in nondividing cells. However, epigenetic silencing of episomal vector genomes reduces IDLV transgene expression levels and renders these safe vectors less efficient. In this article, we describe for the first time a complete correction of factor IX (FIX) deficiency in hemophilia B mice by IDLVs carrying a novel, highly potent human FIX cDNA. A 50-fold increase in human FIX cDNA potency was achieved by combining two mechanistically independent yet synergistic strategies: (i) optimization of the human FIX cDNA codon usage to increase human FIX protein production per vector genome and (ii) generation of a highly catalytic mutant human FIX protein in which the arginine residue at position 338 was substituted with leucine. The enhanced human FIX activity was not associated with liver damage or with the formation of human FIX-directed inhibitory antibodies and rendered IDLV-treated FIX-knockout mice resistant to a challenging tail-clipping assay. A novel S1 nuclease-based B1-quantitative polymerase chain reaction assay showed low levels of IDLV integration in mouse liver. Overall, this study demonstrates that IDLVs carrying an improved human FIX cDNA safely and efficiently cure hemophilia B in a mouse model.


Assuntos
Fator IX/genética , Vetores Genéticos/administração & dosagem , Hemofilia B/terapia , Lentivirus/genética , Animais , Arginina/metabolismo , Códon , Modelos Animais de Doenças , Fator IX/metabolismo , Terapia Genética , Vetores Genéticos/uso terapêutico , Hemofilia B/patologia , Células Hep G2 , Humanos , Leucina/metabolismo , Fígado/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Am J Hematol ; 89(6): 621-5, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24616187

RESUMO

On March 12, 2012, the Centers for Disease Control and Prevention (CDC) held a meeting of its partners in hemophilia treatment, community-based organizations, industry, and government to review data and discuss implementation issues relevant to planned United States (U.S.) national inhibitor surveillance. Issues discussed included the current status of inhibitor surveillance in the United Kingdom (UK) and the US, the results of a US inhibitor surveillance feasibility study, proposed national surveillance schemes, laboratory testing and reporting issues and potential opportunities for future inhibitor-related research. It was concluded that implementation of a national program of inhibitor surveillance using standardized testing through an established public health registry along with patient and care provider education and targeted research provide the best opportunity to inform efforts to develop and evaluate effective prevention strategies.


Assuntos
Autoanticorpos/sangue , Hemofilia A/imunologia , Centers for Disease Control and Prevention, U.S. , Monitoramento Epidemiológico , Hemofilia A/sangue , Humanos , Estados Unidos
12.
Curr Opin Hematol ; 20(5): 410-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23852185

RESUMO

PURPOSE OF REVIEW: Consistently measurable and persistent expression of circulating clotting factor activity, associated with decreased clinical bleeding, has been achieved for the first time in a hemophilia gene therapy trial. This review examines the successes and limitations of this clinical trial for hemophilia B and approaches to advance beyond this milestone. RECENT FINDINGS: Although a self-complementary serotype 8 adeno-associated virus (scAAV8) vector approach directed factor IX expression of up to 6% in a human trial, the apparent need to suppress vector dose-dependent immune-mediated liver inflammation in some patients at the highest dose highlighted the next steps to optimize the risk-benefit of hemophilia gene therapy. The approaches being pursued include manufacturing modifications to eliminate contaminating empty vector capsids, the utilization of factor IX and factor VIII modified transgenes to improve secretion or function of the transgene product, and adjunctive pharmacologic and molecular approaches to overcome limitations imposed by naturally occurring antibodies against vectors and by the large size of the factor VIII gene. SUMMARY: Preclinical data suggest strategies in development may build upon the first gene therapy success and achieve factor IX correction sufficient to prevent bleeding without toxicity and translate success to hemophilia A gene therapy.


Assuntos
Terapia Genética/métodos , Hemofilia B/terapia , Ensaios Clínicos como Assunto , Dependovirus/genética , Fator IX/genética , Fator IX/metabolismo , Vetores Genéticos/genética , Hemofilia B/genética , Humanos
13.
Lancet Haematol ; 11(4): e265-e275, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38437857

RESUMO

BACKGROUND: Etranacogene dezaparvovec, the first gene therapy approved for haemophilia B treatment, was shown to be superior to treatment with continuous prophylactic factor IX in terms of bleeding protection 18 months after gene therapy in a phase 3 trial. We report post-hoc 24-month efficacy and safety data from this trial to evaluate the longer-term effects of etranacogene dezaparvovec in individuals with haemophilia B. METHODS: The phase 3 HOPE-B trial enrolled males aged 18 years or older with inherited haemophilia B, classified as severe (plasma factor IX activity level <1%) or moderately severe (plasma factor IX activity level ≥1% and ≤2%), with a severe bleeding phenotype and who were on stable continuous factor IX prophylaxis. Participants were treated with a single infusion of etranacogene dezaparvovec (2 × 1013 genome copies per kg of bodyweight). The primary endpoint, reported previously, was non-inferiority of the annualised bleeding rate (ABR) during the 52 weeks following stable factor IX expression (defined as months 7-18 after treatment) versus an at least 6-month lead-in period in which participants received their usual continuous factor IX prophylaxis, and is updated here up to month 24. Additional, post-hoc efficacy analyses, including adjusted ABR, factor IX activity, participants within factor IX ranges, and factor IX use, and safety analyses were performed at 24 months after gene therapy. Data were analysed in the full analysis set, which comprised the 54 patients who received at least a partial dose of gene therapy. The trial is ongoing and is registered with ClinicalTrials.gov, number NCT03569891. FINDINGS: The study began on June 27, 2018, and participants were treated between January, 2019, and March, 2020; the date of data cutoff was April 21, 2022. 54 adult males (40 White, two Asian, one Black or African American, 11 other or missing) received a single intravenous infusion of etranacogene dezaparvovec and were followed for a median of 26·51 months (IQR 24·54-27·99), after a lead-in period of 7·13 months (6·51-7·82). In the updated analysis comparing months 7-24 after gene therapy to the lead-in period, mean adjusted ABR significantly reduced from 4·18 to 1·51 (p=0·0002) for all bleeds and from 3·65 to 0·99 (p=0·0001) for factor IX-treated bleeds. During each 6-month period after gene therapy, at least 67% of participants experienced no bleeding (36 of 54 during months 0-6 and stable thereafter), compared with 14 (26%) of 54 during the lead-in period. 24 months after gene therapy, 1 (2%) participant had one-stage factor IX activity less than 5%, whereas 18 (33%) had factor IX activity more than 40% (non-haemophilia range), with mean factor IX activity stable and sustained at 36·7% (SD 19·0%). 52 (96%) of 54 participants expressed endogenous factor IX, remaining free of factor IX prophylaxis at month 24. No new safety concerns were identified and no treatment-related serious adverse events or treatment-related deaths occurred. The most common treatment-related adverse events were an increase in alanine aminotransferase (nine [17%] of 54 patients), headache (eight [15%]), influenza-like illness (seven [13%]), and an increase in aspartate aminotransferase (five [9%]). INTERPRETATION: By providing durable disease correction throughout the 24 months after gene therapy, etranacogene dezaparvovec provides a safe and effective therapeutic option for patients with severe or moderately severe haemophilia B. FUNDING: uniQure and CSL Behring.


Assuntos
Hemofilia A , Hemofilia B , Adulto , Masculino , Humanos , Hemofilia B/genética , Hemofilia B/terapia , Fator IX/efeitos adversos , Fator IX/genética , Hemorragia/prevenção & controle , Hemorragia/induzido quimicamente , Hemofilia A/tratamento farmacológico , Cefaleia/induzido quimicamente
14.
J Virol ; 86(15): 7752-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22593151

RESUMO

Adeno-associated virus (AAV) vectors have the potential to promote long-term gene expression. Unfortunately, humoral immunity restricts patient treatment and in addition provides an obstacle to the potential option of vector readministration. In this study, we describe a comprehensive characterization of the neutralizing antibody (NAb) response to AAV type 1 (AAV1) through AAV5 both in vitro and in vivo. These results demonstrated that NAbs generated from one AAV type are unable to neutralize the transduction of other types. We extended this observation by demonstrating that a rationally engineered, muscle-tropic AAV2 mutant containing 5 amino acid substitutions from AAV1 displayed a NAb profile different from those of parental AAV2 and AAV1. Here we found that a single insertion of Thr from AAV1 into AAV2 capsid at residue 265 preserved high muscle transduction, while also changing the immune profile. To better understand the role of Thr insertion at position 265, we replaced all 20 amino acids and evaluated both muscle transduction and the NAb response. Of these variants, 8 mutants induced higher muscle transduction than AAV2. Additionally, three classes of capsid NAb immune profile were defined based on the ability to inhibit transduction from AAV2 or mutants. While no relationship was found between transduction, amino acid properties, and NAb titer or its cross-reactivity, these studies map a critical capsid motif involved in all steps of AAV infectivity. Our results suggest that AAV types can be utilized not only as templates to generate mutants with enhanced transduction efficiency but also as substrates for repeat administration.


Assuntos
Substituição de Aminoácidos , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Capsídeo/imunologia , Dependovirus , Imunidade Humoral , Transdução Genética , Animais , Células COS , Linhagem Celular , Chlorocebus aethiops , Dependovirus/genética , Dependovirus/imunologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/imunologia , Músculo Esquelético/virologia
15.
Transfusion ; 53(6): 1217-25, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22998193

RESUMO

BACKGROUND: Parvovirus B19 (B19V) is a small, nonenveloped virus that typically causes a benign flu-like illness that occurs most frequently in childhood. The virus is resistant to current viral inactivation steps used in the manufacture of antihemophilic factor concentrates and B19V transmission through these products has been documented. Since 2000, B19V nucleic acid test (NAT) screening of plasma pools has been implemented to further decrease the viral burden in these products, but no study has examined populations using these products to assess the impact of the screening on B19V transmission. STUDY DESIGN AND METHODS: Blood specimens obtained from participants of a surveillance system established in federally supported specialized bleeding disorders clinics were used in a B19V seroprevalence study. RESULTS: A total of 1643 specimens from 1043 participants age 2 to 7 years born after B19V NAT screening was implemented were tested. Age-specific prevalence rates were generally higher for subjects exposed to either plasma-derived products alone or in combination with other products compared to subjects with no exposure to antihemophilic products. Overall, compared to participants unexposed to blood or blood products, those exposed to plasma-derived products alone were 1.7 times more likely to have antibodies to B19V (p = 0.002). CONCLUSION: These results are consistent with continued B19V transmission through plasma-derived factor concentrates. Effective viral inactivation and detection processes are needed to protect users of these products from infection with B19V or other new or emerging viruses.


Assuntos
Fatores de Coagulação Sanguínea/efeitos adversos , Hemofilia A , Infecções por Parvoviridae/sangue , Infecções por Parvoviridae/transmissão , Parvovirus B19 Humano/isolamento & purificação , Algoritmos , Armazenamento de Sangue/métodos , Fatores de Coagulação Sanguínea/uso terapêutico , Criança , Pré-Escolar , Doenças Transmissíveis Emergentes/sangue , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/transmissão , DNA Viral/análise , Feminino , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Hemofilia A/virologia , Hemorragia/sangue , Hemorragia/tratamento farmacológico , Hemorragia/virologia , Humanos , Controle de Infecções/métodos , Modelos Logísticos , Masculino , Infecções por Parvoviridae/epidemiologia , Parvovirus B19 Humano/genética , Prevalência , Estudos Soroepidemiológicos
16.
Curr Opin Pediatr ; 25(1): 23-30, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23274430

RESUMO

PURPOSE OF REVIEW: Disorders of hemostasis such as hemophilia, von Willebrand disease (VWD), and other clotting protein deficiencies lead to significant morbidity in the pediatric population. Because of the limitations of current treatment options, novel therapies are being developed, many of which are reviewed here. RECENT FINDINGS: Several new observations about the nature of clotting protein physiology have been made recently, creating novel perspectives on the treatment options. This review will mostly focus on the current therapy as well as new progress in hemophilia care (particularly strategies to prolong half-life of clotting factor replacements, the management of inhibitors, gene therapy, and novel therapeutic approaches), and briefly mention some progress in VWD and fibrinogen deficiency therapies. SUMMARY: New therapeutic developments have the potential to dramatically decrease morbidity and improve the quality of life of children with bleeding disorders.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/terapia , Transtornos Hemorrágicos/terapia , Criança , Coagulantes/uso terapêutico , Fator IX/uso terapêutico , Fator VIII/uso terapêutico , Terapia Genética/métodos , Hemofilia A/terapia , Humanos , Masculino
17.
Mol Ther ; 20(2): 443-55, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22068425

RESUMO

Efficient and widespread gene transfer is required for successful treatment of Duchenne muscular dystrophy (DMD). Here, we performed the first clinical trial using a chimeric adeno-associated virus (AAV) capsid variant (designated AAV2.5) derived from a rational design strategy. AAV2.5 was generated from the AAV2 capsid with five mutations from AAV1. The novel chimeric vector combines the improved muscle transduction capacity of AAV1 with reduced antigenic crossreactivity against both parental serotypes, while keeping the AAV2 receptor binding. In a randomized double-blind placebo-controlled phase I clinical study in DMD boys, AAV2.5 vector was injected into the bicep muscle in one arm, with saline control in the contralateral arm. A subset of patients received AAV empty capsid instead of saline in an effort to distinguish an immune response to vector versus minidystrophin transgene. Recombinant AAV genomes were detected in all patients with up to 2.56 vector copies per diploid genome. There was no cellular immune response to AAV2.5 capsid. This trial established that rationally designed AAV2.5 vector was safe and well tolerated, lays the foundation of customizing AAV vectors that best suit the clinical objective (e.g., limb infusion gene delivery) and should usher in the next generation of viral delivery systems for human gene transfer.


Assuntos
Dependovirus/genética , Terapia Genética , Vetores Genéticos/genética , Distrofia Muscular de Duchenne/terapia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Proteínas do Capsídeo/química , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Linhagem Celular , Criança , Pré-Escolar , Dependovirus/fisiologia , Distrofina/genética , Distrofina/metabolismo , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Modelos Moleculares , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia , Conformação Proteica , Alinhamento de Sequência , Linfócitos T/imunologia , Transdução Genética , Tropismo Viral
18.
Curr Med Res Opin ; 39(2): 227-237, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36285399

RESUMO

OBJECTIVE: Congenital hemophilia B is a rare bleeding disorder caused by defects in the gene encoding factor IX (FIX) leading to coagulation deficiency. Recurrent bleeds may cause chronic pain, disability, and reduced quality of life. Phase 2 b and 3 single-arm, open-label, single-dose trials assessing etranacogene dezaparvovec gene therapy for hemophilia B have demonstrated sustained FIX activity levels over observed periods, but long-term durability of the treatment effect has not been established. Using statistical modeling, we estimate long-term durability of FIX activity levels after receiving etranacogene dezaparvovec. METHODS: Participants from Phase 2 b (N = 3; NCT03489291) and 3 studies (N = 52; NCT03569891) were included. Two participants who did not respond to treatment were excluded. FIX activity was assessed by one-stage activated partial thromboplastin time assay. FIX activity levels at Month 6 post-treatment were considered baseline. Bayesian and Frequentist linear mixed models predicted FIX activity levels up to 25.5 years at an individual and population level with pre-treatment adeno-associated virus 5 (AAV5) neutralizing antibody (NAb) status as primary covariate. RESULTS: Bayesian and Frequentist linear mixed models predicted no more than 6/55 (10.91%) observed participants would have FIX activity levels <2% up to 25.5 years post-infusion. Bayesian model-based predictions of future participants suggest >80% would be free from prophylactic FIX replacement products 25.5 years post-infusion. Both models predicted FIX activity levels were not significantly influenced by pre-treatment AAV5 NAb status. CONCLUSIONS: People with hemophilia B receiving etranacogene dezaparvovec would likely achieve durable FIX activity levels and remain free of prophylactic FIX replacement products for up to 25.5 years following single administration. The long-term factor IX durability predictions are based on statistical methods and results in vivo may differ.


Hemophilia B is a rare bleeding condition where blood does not clot properly, causing excessive bleeding. It is caused by a change or mutation to a gene, leading to lower-than-normal levels of a clotting factor, called factor IX. Standard treatment involves replacing missing factor IX through lifelong, regular treatment with factor replacement products. Etranacogene dezaparvovec is a gene therapy developed to replace the faulty gene and increase factor IX activity levels in the blood, thereby reducing bleeding, after one treatment. In clinical trials of etranacogene dezaparvovec, people with severe or moderately severe hemophilia B had stable and long-lasting increases in factor IX activity levels that reached near to the normal range seen in people without hemophilia B.The purpose of this study was to predict whether these increases in factor IX activity levels will last over an extended period of time after receiving etranacogene dezaparvovec.Mathematical predictions showed less than 11% of clinical trial participants would have unacceptable factor IX activity levels (less than 2%) up to 25.5 years after receiving etranacogene dezaparvovec. Further predictions of potential future people with hemophilia B showed that over 80% would not need treatment with factor replacement products 25.5 years after receiving etranacogene dezaparvovec.The goal of treatment is to increase factor IX activity levels into the near-to-normal range in people with hemophilia B and therefore decrease or eliminate bleeding. These results suggest people with hemophilia B receiving etranacogene dezaparvovec would have long-lasting factor IX activity levels and would not need regular factor replacement products for up to 25.5 years following a single treatment of etranacogene dezaparvovec.


Assuntos
Fator IX , Hemofilia B , Humanos , Fator IX/genética , Fator IX/uso terapêutico , Hemofilia B/genética , Hemofilia B/terapia , Qualidade de Vida , Teorema de Bayes , Terapia Genética
19.
Blood Adv ; 7(19): 5671-5679, 2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-36490302

RESUMO

Etranacogene dezaparvovec (AMT-061) is a recombinant adeno-associated virus serotype 5 (AAV5) vector containing a codon-optimized Padua variant human factor IX (FIX) transgene with a liver-specific promoter. Here, we report 3-year outcomes from a phase 2b, open-label, single-dose, single-arm, multicenter trial conducted among adults with severe or moderately severe hemophilia B (FIX ≤2%). All participants (n = 3) received a single intravenous dose (2 × 1013 gene copies per kg) and will be followed up for 5 years. The primary end point of FIX activity ≥5% at 6 weeks was met. Secondary end points included bleed frequency, FIX concentrate use, joint health, and adverse events (AEs). All participants required routine FIX prophylaxis and had neutralizing antibodies to AAV5 before etranacogene dezaparvovec treatment. After administration, FIX activity rose to a mean of 40.8% in year 1 and was sustained in year 3 at 36.9%. All participants discontinued FIX prophylaxis. Bleeding was completely eliminated in 2 out of 3 participants. One participant required on-demand FIX replacement therapy per protocol because of elective surgical procedures, for 2 reported bleeding episodes, and twice for a single self-administered infusion because of an unreported reason. One participant experienced 2 mild, self-limiting AEs shortly after dosing. During the 3-year study period, there were no clinically significant elevations in liver enzymes, no requirement for steroids, no FIX inhibitor development, and no late-emergent safety events in any participant. Etranacogene dezaparvovec was safe and effective in adults with hemophilia B over 3 years after administration. This trial was registered at www.clinicaltrials.gov as #NCT03489291.


Assuntos
Hemofilia B , Adulto , Humanos , Dependovirus/genética , Fator IX/genética , Terapia Genética/métodos , Hemofilia B/tratamento farmacológico , Hemofilia B/genética , Hemorragia/etiologia
20.
Haemophilia ; 18 Suppl 5: 17-26, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22757680

RESUMO

Progressive joint destruction resulting from intra-articular bleeding is the major morbidity affecting patients with haemophilia (PWH), particularly those with inhibitors. Advances in understanding the detrimental processes set in motion by the exposure of joints to bleeding have shaped current management methods. However, to achieve optimal joint health in PWH, in addition to achieving haemostasis at the bleeding vessel, it may be appropriate to explore experimentally other conceptual frameworks. These include the possibilities that markers might help to identify individuals at the risk of more rapid joint deterioration, that clotting factors may have additional local action within tissues, and that outcomes might be improved with therapies that directly address wound healing and inflammation. Joint assessment tools are important. Conventional radiography is frequently used, but given the possibility of subclinical joint bleeds, accurate non-invasive imaging tools are required to detect soft tissue and cartilage changes. Magnetic resonance imaging and ultrasonography can prove valuable here. New imaging techniques should help to increase understanding of the biological basis of early events in haemophilic arthropathy. The optimal way to measure outcomes in haemophilia is to use several methods - in addition to imaging methods, a 360° approach will use physical, functional and quality-of-life instruments. In PWH, inhibitor development complicates treatment of joint bleeds and increases the risk of developing arthropathy. A new therapeutic approach for joint bleeds in inhibitor patients divides treatment into two phases: bleed control, with bypassing agent therapy until bleeding has definitely ceased, followed by regular dosing to prevent rebleeds until synovial recovery is complete.


Assuntos
Hemartrose/fisiopatologia , Hemofilia A/patologia , Atividades Cotidianas , Fatores de Coagulação Sanguínea/uso terapêutico , Avaliação da Deficiência , Hemartrose/etiologia , Hemartrose/terapia , Humanos , Qualidade de Vida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA