Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.171
Filtrer
1.
Sci Rep ; 14(1): 20534, 2024 09 04.
Article de Anglais | MEDLINE | ID: mdl-39227636

RÉSUMÉ

An important aspect of improving care for people with hemophilia B (HB) is developing optimal treatment strategies. Here we aimed to provide in-silico evidence, comparing the estimated optimal posology of factor IX (FIX) products to support the patient-physician decision-making process. A population pharmacokinetic (popPK) model-based assessment comparing the performance of FIX products (rFIX, rIX-FP, rFIXFc, N9-GP) was developed. PopPK analyses were used to determine a product's optimal posology to target predefined steady-state FIX activity trough levels in a hypothetical population of 10,000 people with severe HB. Model-derived optimal posologies were compared across several parameters including trough levels, proportion of patients per regimen and consumption, considering 64 hypothetical patient scenarios of different FIX trough level targets and ages. Results indicated a marked difference between FIX products estimated to achieve target trough levels, consumption and dosing frequencies. rIX-FP was associated with higher trough levels than rFIX and rFIXFc, at a lower weekly dose and administration frequency, across all age groups. N9-GP use in adolescents and adults was associated with lower consumption compared with rIX-FP. Insights from this study may be utilized by clinicians to inform decision-making, by considering the model-generated estimated optimal posologies alongside multiple clinical factors and patient preferences.


Sujet(s)
Facteur IX , Hémophilie B , Hémophilie B/traitement médicamenteux , Hémophilie B/sang , Humains , Facteur IX/pharmacocinétique , Facteur IX/usage thérapeutique , Facteur IX/administration et posologie , Adulte , Adolescent , Adulte d'âge moyen , Jeune adulte , Enfant , Mâle , Simulation numérique , Enfant d'âge préscolaire
2.
J Manag Care Spec Pharm ; 30(8): 868-872, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39088338

RÉSUMÉ

This viewpoint discusses cost-effectiveness estimates for EtranaDez, a gene therapy for hemophilia B, using the Institute for Clinical and Economic Review's (ICER) framework for single and short-term therapies (SSTs). EtranaDez offers long-term benefits from a single administration, in contrast to the high costs and frequent dosing required by current factor IX prophylaxis. However, the projected gains in health from EtranaDez are small relative to the cost implications of the therapy, and consequently, how the cost offsets associated with EtranaDez are counted has a substantial impact on assessing its cost-effectiveness. Strategies for assessing cost offsets used in the ICER SST framework include a 50/50 cost-sharing model between the health care system and the manufacturer and a cap of $150,000 annually on health care cost offsets. Results from the standard full cost-offset analysis as reported by ICER depicted EtranaDez as a dominant therapy with substantial cost savings compared with factor IX prophylaxis. However, while considering the ICER SST framework, particularly the $150,000 annual cap scenario, the cost-effectiveness was significantly reduced. The incremental cost-effectiveness ratio varied notably between these scenarios, challenging the conventional perception of value of gene therapy in health care. These cost-sharing scenarios highlight the potential of the ICER SST framework to help curtail inefficient health care spending. In cases in which the cost of existing treatment is exceedingly high, the application of such frameworks would improve efficiency in resource allocation, fostering a balance between incentives for innovation and economic sustainability in managed care systems.


Sujet(s)
Économies , Analyse coût-bénéfice , Facteur IX , Thérapie génétique , Hémophilie B , Hémophilie B/économie , Hémophilie B/traitement médicamenteux , Humains , Facteur IX/économie , Facteur IX/usage thérapeutique , Thérapie génétique/économie , Coûts des soins de santé
4.
Epidemiol Prev ; 48(3): 233-238, 2024.
Article de Italien | MEDLINE | ID: mdl-38995136

RÉSUMÉ

OBJECTIVES: to assess the variability in expenditure compared to 2022 assuming different rates of shifting of therapy days from current active ingredients used for the treatment of haemophilia B to nonacog beta pegolDesign: descriptive cross-sectional study. SETTING AND PARTICIPANTS: consumption in the year 2022 (data source: Medicines Utilisation Monitoring Centre, Italian Medicines Agency) of all medicinal products available in Italy containing coagulation factor IX. MAIN OUTCOMES MEASURES: for each active ingredient, the total number of therapy days and the variability in expenditure compared to 2022 were estimated on the basis of a switch of therapy days, between 5% and 20%, to nonacog beta pegol. RESULTS: on the basis of considered scenarios, the analysis shows that the total annual expenditure for clotting factors used in the treatment of haemophilia B could remain at most unchanged or reduced. Particularly, the extent of the reduction in spending could vary from 0.11% to 2.26%. This trend would be in contrast to the stable increase seen in recent years, particularly in 2022. CONCLUSIONS: this predictive spending assessment may be useful in evaluating the economic impact from new treatment options, such as etranacogene dezaparvovec gene therapy already approved by the European Medicines Agency and the Food and Drug Administration, and to improve pharmaceutical governance.


Sujet(s)
Facteur IX , Hémophilie B , Italie , Humains , Études transversales , Hémophilie B/traitement médicamenteux , Hémophilie B/économie , Facteur IX/usage thérapeutique , Facteur IX/économie , Coûts des médicaments , Protéines recombinantes/usage thérapeutique , Protéines recombinantes/économie , Polyéthylène glycols/usage thérapeutique , Polyéthylène glycols/économie , Dépenses de santé/statistiques et données numériques
6.
JBJS Case Connect ; 14(3)2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-39058793

RÉSUMÉ

CASE: A 29-year-old man with hemophilia B presented with advanced arthropathy of the right knee, resulting in poor knee functional scores and difficulties in his livelihood. The patient underwent total knee replacement while receiving nonacog beta pegol factor IX by a multidisciplinary approach. CONCLUSION: Hemophilias commonly result in end-stage hemophilic arthropathy of the joints at a young age that may warrant joint replacement surgeries. This case report illustrates the surgical protocol of total knee arthroplasty in a patient who received a long-acting factor IX preparation.


Sujet(s)
Arthroplastie prothétique de genou , Hémophilie B , Polyéthylène glycols , Humains , Mâle , Adulte , Hémophilie B/complications , Hémophilie B/traitement médicamenteux , Hémarthrose/chirurgie , Hémarthrose/étiologie , Facteur IX/administration et posologie , Facteur IX/usage thérapeutique , Articulation du genou/chirurgie , Articulation du genou/imagerie diagnostique , Protéines recombinantes
7.
J Manag Care Spec Pharm ; 30(10): 1095-1105, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38923896

RÉSUMÉ

BACKGROUND: Hemophilia B is characterized by a deficiency of clotting factor IX (FIX), leading to excessive bleeding. Hemophilia B is commonly treated using replacement FIX therapy, which may be administered prophylactically or on-demand following a bleeding episode. Previous research has found high health care resource use (HCRU) and costs among Medicare and commercially insured people with hemophilia B (PwHB), with FIX therapy being a primary driver of health care costs. OBJECTIVE: To assess HCRU, outcomes, and costs among US Medicaid beneficiaries receiving FIX prophylaxis for hemophilia B. METHODS: This study employed a retrospective comparative cohort design to assess HCRU, outcomes, and costs among adult male Medicaid beneficiaries receiving FIX prophylaxis for hemophilia B, relative to a matched comparator population of beneficiaries without bleeding disorders. Nationwide Medicaid claims and enrollment data from 2015 to 2020 were used for this analysis. Adult male PwHB who received FIX prophylaxis, defined as not having identified gaps in FIX therapy exceeding 60-days during a 1-year measurement period, and were continuously enrolled in Medicaid for at least 2 years, were matched 1:4 to comparator beneficiaries without bleeding disorders based on baseline demographic and clinical characteristics. Key measures of HCRU and outcomes included inpatient hospital admissions, outpatient hematologist visits, and bleeding events. Measures of health care costs were assessed among a subset of beneficiaries enrolled in fee-for-service Medicaid. RESULTS: PwHB receiving FIX prophylaxis were significantly more likely to have multiple inpatient hospital admissions and had a longer cumulative length of stay per person relative to comparator beneficiaries (30.2 vs 14.8 days, respectively; P = 0.0473). PwHB receiving FIX prophylaxis also had significantly higher rates of bleeding events relative to comparator beneficiaries (0.54 vs 0.02 per person, respectively; P < 0.0001) and outpatient hematologist visits (1.58 vs 0.20 per person, respectively; P < 0.0001). Annual costs among PwHB receiving FIX prophylaxis were significantly higher than costs among comparator beneficiaries ($928,370 vs $34,553 per person, respectively; P < 0.0001) and were overwhelmingly driven by costs associated with FIX therapy. CONCLUSIONS: This analysis found higher rates of HCRU and costs among Medicaid beneficiaries receiving FIX prophylaxis for hemophilia B relative to a matched comparator population of beneficiaries without bleeding disorders. Future research should examine hemophilia B costs and outcomes within the context of new treatments with innovative mechanisms of action, such as gene therapies, RNA interference therapies, and antitissue factor pathway inhibitor therapies.


Sujet(s)
Facteur IX , Coûts des soins de santé , Hémophilie B , Medicaid (USA) , Humains , États-Unis , Hémophilie B/traitement médicamenteux , Hémophilie B/économie , Medicaid (USA)/économie , Facteur IX/usage thérapeutique , Facteur IX/économie , Facteur IX/administration et posologie , Mâle , Études rétrospectives , Adulte , Adulte d'âge moyen , Jeune adulte , Acceptation des soins par les patients/statistiques et données numériques , Ressources en santé/économie , Ressources en santé/statistiques et données numériques , Études de cohortes , Adolescent
8.
Thromb Haemost ; 124(10): 912-921, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-38863155

RÉSUMÉ

BACKGROUND: Fidanacogene elaparvovec, an adeno-associated virus-based gene therapy vector expressing the high-activity factor IX (FIX) variant FIX-R338L, is in development for hemophilia B. One-stage clotting (OS) assays and chromogenic substrate (CS) assays are commonly used to measure FIX-R338L variant activity. Data from ongoing trials suggest FIX activity varies between different OS and CS assays. MATERIAL AND METHODS: To better understand FIX-R338L activity in clinical samples, an international multisite field study was conducted across a central laboratory and 18 local laboratories, using standard protocols, reagents, and instrumentation, with individual participant samples from a phase 1/2a study of fidanacogene elaparvovec. RESULTS: Unlike the wild-type FIX control, FIX-R338L activity was higher with the OS silica-based assay versus OS ellagic acid-based and CS assays. Variation in FIX activity was greater at the lowest activity levels. Activated FIX (FIXa) in plasma could result in higher OS assay activity or increased thrombin generation, which could overestimate FIX activity. However, FIXa was not detected in the participant samples, indicating that it was not contributing to the OS assay differences. Since individuals on gene therapy may receive exogenous replacement FIX products, replacement products were spiked into patient plasma samples to target a therapeutic concentration. Exogenous FIX was additive to endogenous FIX-R338L, with no interference from FIX-R338L. CONCLUSION: These results demonstrate FIX-R338L activity can be measured with OS and CS assays in clinical laboratories and provide insight into assay variability when measuring FIX with endogenously produced FIX-R338L. The findings may help establish best practices for measuring FIX-R338L activity (Clinicaltrials.gov identifier: NCT02484092).


Sujet(s)
Coagulation sanguine , Dependovirus , Facteur IX , Thérapie génétique , Hémophilie B , Humains , Facteur IX/génétique , Facteur IX/métabolisme , Facteur IX/usage thérapeutique , Hémophilie B/thérapie , Hémophilie B/génétique , Hémophilie B/sang , Thérapie génétique/méthodes , Coagulation sanguine/effets des médicaments et des substances chimiques , Dependovirus/génétique , Tests de coagulation sanguine , Vecteurs génétiques , Thrombine/métabolisme
9.
J Med Econ ; 27(1): 758-765, 2024.
Article de Anglais | MEDLINE | ID: mdl-38708771

RÉSUMÉ

INTRODUCTION: Etranacogene dezaparvovec (EDZ), Hemgenix, is a gene therapy recently approved for people with hemophilia B (PwHB). OBJECTIVE: To estimate long-term clinical impact and cost of EDZ in the United States (US). METHODS: A decision-analytic model was developed to evaluate the long-term impact of introducing EDZ for PwHB over a 20-year time horizon. Factor IX (FIX) prophylaxis comparator was a weighted average of different FIX prophylaxis regimens based on US market share data. We compared a scenario in which EDZ is introduced in the US versus a scenario without EDZ. Clinical inputs (annualized FIX-treated bleed rate; adverse event rates) were obtained from HOPE-B phase 3 trial. EDZ durability input was sourced from an analysis predicting long-term FIX activity with EDZ. EDZ one-time price was assumed at $3.5 million. Other medical costs, including FIX prophylaxis, disease monitoring, bleed management, and adverse events were from literature. The model estimated annual and cumulative costs, treated bleeds, and joint procedures over 20 years from EDZ introduction. RESULTS: Approximately 596 PwHB were eligible for EDZ. EDZ uptake was estimated to avert 11,282 bleeds and 64 joint procedures over 20 years. Although adopting EDZ resulted in an annual excess cost over years 1-5 (mean: $53 million annually, total $265 million), annual cost savings were achieved beginning in year 6 (mean: $172 million annually; total $2.58 billion in years 6-20). The total cumulative 20-year cost savings was $2.32 billion, with cumulative cost savings beginning in year 8. CONCLUSION: Introducing EDZ to treat PwHB is expected to result in cost savings and patient benefit over 20 years. Initiating PwHB on EDZ sooner can produce greater and earlier savings and additional bleeds avoided. These results may be a conservative estimate of the full value of EDZ, as PwHB would continue to accrue savings beyond 20 years.


This analysis assessed the long-term clinical and financial impact of introducing EDZ in the United States of America for people with severe or moderately severe hemophilia B. A decision-analytic model was developed comparing a scenario with EDZ and one without EDZ over 20 years. Introducing EDZ would avert 11,292 bleeds and 64 joint procedures over 20 years and would achieve cumulative cost savings in year 8, with a total cumulative 20-year cost saving of $2.32 billion.


Sujet(s)
Facteur IX , Hémophilie B , Humains , Hémophilie B/traitement médicamenteux , Hémophilie B/économie , États-Unis , Facteur IX/économie , Facteur IX/usage thérapeutique , Hémorragie/économie , Thérapie génétique/économie , Analyse coût-bénéfice , Techniques d'aide à la décision , Adulte , Mâle , Enfant , Jeune adulte , Adolescent
10.
Blood Coagul Fibrinolysis ; 35(5): 282-285, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38700550

RÉSUMÉ

Congenital hemophilia B is a rare X-linked recessive bleeding disorder caused by factor IX deficiency. Acquired hemophilia A is a rare, acquired bleeding disorder that presents with new-onset bleeding, especially in older adults, due to the development of auto-antibodies against factor VIII (FVIII). This case report presents the medical management of a patient with congenital hemophilia B and acquired hemophilia A. We highlight the limitations of maintaining factor levels with factor replacement therapy alone, particularly in hemophilia patients who have developed factor inhibitors. In addition, we draw attention to the need for dose escalation, the cost, and the need for immune-tolerance induction therapy. This case illustrates that when the current diagnosis does not explain the full clinical picture and laboratory data are inadequate, it is important to continue to seek alternative diagnoses and cost-effective treatment.


Sujet(s)
Hémophilie A , Hémophilie B , Humains , Hémophilie A/complications , Hémophilie B/complications , Mâle , Facteur VIII/usage thérapeutique , Facteur IX/usage thérapeutique
11.
Am J Manag Care ; 30(5): e157-e164, 2024 05 01.
Article de Anglais | MEDLINE | ID: mdl-38748916

RÉSUMÉ

OBJECTIVES: To describe people with hemophilia B (PWHB) in the US who experience bleeds despite factor replacement therapy and to quantify the associated burden from the third-party payer perspective. STUDY DESIGN: Observational study of adult male PWHB treated with factor IX replacement therapy identified from the PharMetrics Plus claims data from 2010 to 2019. METHODS: Patients with medically recorded bleeds (MRBs) were identified using diagnostic codes. Rates and rate ratios of inpatient admissions, emergency department (ED) visits, and outpatient visits among PWHB with and without MRBs were estimated. The presence of comorbidities was identified using diagnostic codes, and the analysis was stratified by age group. RESULTS: There were 345 PWHB with MRBs and 252 without MRBs. More than half of PWHB with MRBs (56.8%) had 1 or more comorbidity vs 39.3% of PWHB without MRBs. The prevalence of anxiety and depression was high in PWHB, regardless of bleed status and age group, whereas the prevalence of other comorbidities increased with age group. The rate of all-cause inpatient admissions for PWHB with MRBs was 14.8 per 100 person-years (95% CI, 12.8-17.1), 2.5 times higher than for PWHB without MRBs. The rate of all-cause ED visits for PWHB with MRBs was 67.6 per 100 person-years (95% CI, 63.2-72.3), 2.7 times higher than for those without MRBs. CONCLUSIONS: This study reports significant resource use and clinical burden among PWHB who seek medical care. PWHB with MRBs had considerable all-cause resource use compared with PWHB without MRBs. The prevalence of mental illness was consistently high across all age groups.


Sujet(s)
Comorbidité , Hémophilie B , Hémorragie , Humains , Mâle , Hémophilie B/épidémiologie , Hémophilie B/complications , Adulte , Études rétrospectives , Adulte d'âge moyen , Hémorragie/épidémiologie , États-Unis/épidémiologie , Jeune adulte , Facteur IX/usage thérapeutique , Sujet âgé , Adolescent
12.
J Manag Care Spec Pharm ; 30(8): 805-816, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38625717

RÉSUMÉ

BACKGROUND: Etranacogene dezaparvovec is a recently approved gene therapy for people with hemophilia B (PwHB). Current standard of care is prophylaxis with factor IX (FIX) to prevent bleeding. Etranacogene dezaparvovec increases blood FIX levels such that FIX prophylaxis could be eliminated. OBJECTIVE: To estimate the budgetary impact of etranacogene dezaparvovec adoption and utilization in a commercial health plan of the United States. METHODS: A budget impact model was developed to evaluate the introduction of etranacogene dezaparvovec to treat severe or moderately severe hemophilia B. The model considered a hypothetical 1-million-member plan over a 5-year horizon. FIX therapy prophylaxis use was estimated based on a weighted average of relevant brands using US market share data. A scenario of etranacogene dezaparvovec adoption/utilization was compared with one without etranacogene dezaparvovec utilization. Two etranacogene dezaparvovec uptake (market share growth) analyses were performed: one with gradual uptake and alternatively assuming all eligible PwHB received etranacogene dezaparvovec in year 1. The one-time cost of etranacogene dezaparvovec was assumed to be $3.5 million. Other costs (FIX prophylaxis, disease monitoring, bleed management, and adverse events) were estimated from published literature. All costs were in 2022 US dollars. Bleed and adverse event rates were sourced from the HOPE-B trial comparing etranacogene dezaparvovec to previous FIX therapy prophylaxis. The model estimated annual and per-member per-month costs over 5 years. Secondary analyses were performed considering a 10-year horizon. RESULTS: In the 1-million-member health plan, an estimated 1.8 PwHB were eligible for treatment with etranacogene dezaparvovec. Gradual uptake of etranacogene dezaparvovec resulted in cumulative 5-year budget impact of $848,509 compared with a scenario without etranacogene dezaparvovec. In years 1-5, the incremental annual and per-member per-month costs ranged from $79,824 to $271,435 and from $0.007 to $0.023, respectively. In the alternative uptake analysis, etranacogene dezaparvovec became cost saving annually beginning in year 2 and cumulatively beginning in year 5, for a 5-year savings of $754,844. Secondary analyses over 10 years found both uptake analyses cost saving. Other scenarios considered did not affect results substantially. CONCLUSIONS: Introducing etranacogene dezaparvovec as treatment for PwHB would have a modest budget increase within 5 years after treatment but may become cost saving if all eligible PwHB were treated in year 1. Initiating PwHB on etranacogene dezaparvovec sooner may produce greater overall savings and earlier annual savings. Etranacogene dezaparvovec is a treatment option that may provide overall cost savings for US commercial health plans, which would increase as the plan size increases.


Sujet(s)
Budgets , Facteur IX , Hémophilie B , États-Unis , Humains , Hémophilie B/économie , Hémophilie B/traitement médicamenteux , Hémophilie B/thérapie , Facteur IX/économie , Facteur IX/usage thérapeutique , Thérapie génétique/économie , Thérapie génétique/méthodes , Modèles économiques , Analyse coût-bénéfice
13.
Adv Ther ; 41(6): 2307-2323, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38652439

RÉSUMÉ

INTRODUCTION: Recombinant factor IX (rFIX) and recombinant FIX Fc fusion protein (rFIXFc) are standard half-life and extended half-life FIX replacement therapies, respectively, and represent established treatment options indicated for adults and children with haemophilia B. These FIX replacement therapies can be administered as prophylaxis (to prevent bleeding) or 'on-demand' (to stop bleeding). This analysis aimed to estimate the cost-effectiveness of once-weekly prophylaxis with rFIXFc versus on-demand treatment with rFIX in patients with haemophilia B without inhibitors in the Italian healthcare setting. METHODS: A Markov model was developed to assess a hypothetical cohort of adolescent or adult male patients (≥ 12 years) with haemophilia B (FIX level of ≤ 2 IU/dL) without inhibitors. Model inputs were derived from the pivotal phase 3 clinical studies for rFIXFc and rFIX, published literature and assumptions when published data were unavailable. The model employed a lifelong time horizon with 6-monthly transitions between health states, and it estimated total costs, total quality-adjusted life years (QALYs), number of bleeds, number of surgeries and incremental cost-effectiveness ratio. RESULTS: rFIXFc prophylaxis was associated with lower total costs per patient (€5,308,625 versus €6,564,510) and greater total QALYs per patient (15.936 versus 11.943) compared with rFIX on-demand; rFIXFc prophylaxis was therefore the dominant treatment strategy. The model also demonstrated that rFIXFc prophylaxis was associated with fewer incremental bleeds (- 682.29) and surgeries (- 0.39) compared with rFIX on-demand. CONCLUSIONS: rFIXFc prophylaxis provides improved health outcomes and lower costs, and represents a cost-effective treatment option compared with rFIX on-demand for adolescent and adult male patients with haemophilia B. This comparative assessment of cost-effectiveness should help to inform both clinicians and healthcare policy makers when making treatment decisions for patients with haemophilia B.


Sujet(s)
Facteur IX , Hémophilie B , Fragments Fc des immunoglobulines , Protéines de fusion recombinantes , Adolescent , Adulte , Enfant , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Analyse coût-bénéfice , Facteur IX/usage thérapeutique , Facteur IX/économie , Hémophilie B/traitement médicamenteux , Hémophilie B/économie , Hémorragie/prévention et contrôle , Fragments Fc des immunoglobulines/usage thérapeutique , Fragments Fc des immunoglobulines/économie , Italie , Chaines de Markov , Années de vie ajustées sur la qualité , Protéines de fusion recombinantes/économie , Protéines de fusion recombinantes/usage thérapeutique , Protéines recombinantes/usage thérapeutique , Protéines recombinantes/économie
14.
Blood Adv ; 8(7): 1796-1803, 2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38592711

RÉSUMÉ

ABSTRACT: The US Food and Drug Administration (FDA)'s authorization of etranacogene dezaparvovec (Hemgenix) is a significant milestone, constituting not only the first FDA approval of a gene therapy for hemophilia but also the first approval of a liver-targeted adeno-associated virus vector gene therapy. This review summarizes the nonclinical studies and clinical development that supported regulatory clearance. Similar to other gene therapies for single gene disorders, both the short-term safety and the phenotypic improvement were unequivocal, justifying the modest-sized safety and efficacy database, which included 57 participants across the phase 2b (3 participants) and phase 3 (54 participants) studies. The most common adverse reactions included liver enzyme elevation, headache, flu-like symptoms, infusion-related reactions, creatine kinase elevation, malaise, and fatigue; these were mostly transient. One participant had hepatocellular carcinoma on a study-mandated liver ultrasound conducted 1 year after vector infusion; molecular analysis of the resected tumor showed no evidence of vector-related insertional mutagenesis as the etiology. A remarkable 96% of participants in the phase 3 trial were able to stop factor IX (FIX) prophylaxis, with the study demonstrating noninferiority to FIX prophylaxis in terms of the primary end point, annualized bleeding rate. Key secondary end points such as the annualized infusion rate, which declined by 97%, and the plasma FIX activity level at 18 months after infusion, with least squares mean increase of 34.3 percentage points compared with baseline, were both clinically and statistically significant. The FDA's landmark approval of Hemgenix as a pioneering treatment for hemophilia stands on the shoulders of >20 years of gene therapy clinical research and heralds a promising future for genomic medicines.


Sujet(s)
Hémophilie A , Hémophilie B , États-Unis , Humains , Hémophilie B/génétique , Hémophilie B/thérapie , Facteur IX/génétique , Facteur IX/usage thérapeutique , Bases de données factuelles , Fatigue
15.
Haemophilia ; 30(3): 709-719, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38462823

RÉSUMÉ

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.


Sujet(s)
Thérapie génétique , Hémophilie B , Qualité de vie , Humains , Hémophilie B/psychologie , Hémophilie B/thérapie , Thérapie génétique/méthodes , Mâle , Adulte , Adulte d'âge moyen , Jeune adulte , Facteur IX/usage thérapeutique , Adolescent , Femelle , Dependovirus/génétique , Enquêtes et questionnaires , Indice de gravité de la maladie
16.
Semin Thromb Hemost ; 50(5): 714-719, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38499191

RÉSUMÉ

Hemophilia A/B are caused by deficiency or lack of coagulation factors VIII (FVIII) or factor IX (FIX), respectively, in plasma. A person with hemophilia develops bleeding in the joints and muscles at an early age, which, if left untreated, leads to early arthropathy. Preventive treatment can be achieved by regular (prophylactic) administration of FVIII/FIX. In 1958, this was implemented on a small scale in Sweden with FVIII in patients with severe hemophilia A, and in those with hemophilia B in 1972 when FIX became available. However, there were problems with human immunodeficiency virus and hepatitis infection from contaminated blood products. In the 1990s, recombinant FVIII and FIX concentrates were introduced. The major remaining problems then were the development of inhibitors, and the need for a venous route for the injections in very young children. High-titer inhibitors were treated by immune tolerance induction according to a modified model of the original Bonn high-dose protocol. A central venous line, i.e., Port-A-Cath, has enabled early prophylaxis in many children with poor venous access and has enabled the early start of home treatment with adequate injection frequency. Scoring systems for X-rays, magnetic resonance imaging, and function of joints were developed early in Sweden and have been widely disseminated worldwide, partly with modifications. Extended half-life products with half-life increased three to five times have been developed, which can provide superior bleed protection when dosed once-weekly and can maintain therapeutic trough levels when administered less frequently. The ultimate prophylaxis therapy in the future may be gene therapy.


Sujet(s)
Hémophilie A , Humains , Hémophilie A/traitement médicamenteux , Suède , Enfant , Facteur VIII/usage thérapeutique , Enfant d'âge préscolaire , Mâle , Facteur IX/usage thérapeutique , Hémophilie B/traitement médicamenteux
17.
Haemophilia ; 30 Suppl 3: 95-102, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38539060

RÉSUMÉ

INTRODUCTION: The advent of therapeutic recombinant factor VIII (FVIII) and factor IX (FIX) protein infusions revolutionized the care of persons with haemophilia in the 1990s. It kicked off an era with the increasing use of prophylactic factor infusions for patients and transformed conversations around the ideal trough activity levels as well as the ultimate goals in tailored, individualized care. Our knowledge surrounding the immunologic basis of inhibitor development and treatment derives from a time when patients were receiving frequent factor infusions and focused on immune tolerance induction following inhibitor development. DISCUSSION: More recently, care was revolutionized again in haemophilia A with the approval of emicizumab, a bispecific antibody mimicking activated FVIII function, to prevent bleeding. The use of emicizumab prophylaxis has resulted in a significantly slower accumulation of factor exposure days and continued effective prophylaxis in the case of inhibitor development. While emicizumab is effective at reducing the frequency of bleeding events in patients with haemophilia A, management of breakthrough bleeds, trauma, and surgeries still requires additional treatment. Ensuring that FVIII is a therapeutic option, particularly for life-threatening bleeding events and major surgeries is critical to optimizing the care of persons with haemophilia A. Other novel non-factor concentrate therapies, including rebalancing agents, will dramatically change the landscape for persons with haemophilia B with inhibitors. CONCLUSION: This review discusses the changing landscape regarding the timing of inhibitor development and management strategies after inhibitor development, stressing the importance of education across the community to continue to vigilantly monitor for inhibitors and be prepared to treat persons with inhibitors.


Sujet(s)
Anticorps bispécifiques , Hémophilie A , Hémophilie B , Hémostatiques , Humains , Hémophilie A/traitement médicamenteux , Facteur VIII/usage thérapeutique , Hémophilie B/traitement médicamenteux , Hémorragie/prévention et contrôle , Hémorragie/traitement médicamenteux , Anticorps bispécifiques/pharmacologie , Facteur IX/usage thérapeutique , Hémostatiques/usage thérapeutique
18.
Blood Coagul Fibrinolysis ; 35(3): 133-135, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38358900

RÉSUMÉ

Inferior vena cava thrombosis (IVCT) is rare. Thrombophilia is one of the important risk factors. It is also uncommon for gene mutations in F9 gene to cause thrombosis but not hemorrhage. A 35-year-old male patient was admitted to our department with left lower limb swelling without an obvious cause for 1 day. Through contrast-enhanced computed tomography and color Doppler ultrasound, he was found to have lower extremity deep vein thrombosis, IVCT and pulmonary embolism. Through whole-exome sequencing analysis, he was found to carry a 925.7 kb duplication (chrX:137939698-138865419, hg19) encompassing ATP11C , SRD5A1P1 , MCF2 , FGF13 and F9 genes. This duplication of F9 gene was not detected in his parents. Other thrombophilic genes defects were not found. The factor IX activities of this patient, his father and mother were 194, 70 and 148, respectively. He was treated with catheter-directed thrombolysis, AngioJet-assisted pharmaco-mechanical thromboectomy and manual aspiration thromboectomy. Complete recanalization of left femoral, iliac veins and inferior vena cava was achieved. F9 gene duplication is a rare mutation, which can induce multiple venous thrombosis through increasing the activity level of factor IX in plasma. IVCT is a serious type of venous thrombosis. Personalized intervention treatment plans should be developed based on the different clinical characteristics of each case to achieve a higher benefit-risk ratio.


Sujet(s)
Duplication de gène , Thrombose veineuse , Mâle , Humains , Adulte , Facteur IX/usage thérapeutique , Thrombose veineuse/étiologie , Thrombose veineuse/génétique , Veine cave inférieure , Traitement thrombolytique/méthodes , Cathéters/effets indésirables , Résultat thérapeutique , Protéines proto-oncogènes/usage thérapeutique , Facteurs d'échange de nucléotides guanyliques/usage thérapeutique , Adenosine triphosphatases/usage thérapeutique , Protéines de transport membranaire/usage thérapeutique
19.
Thromb Res ; 236: 242-249, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38383218

RÉSUMÉ

Early gene therapy clinical trials for the treatment of Haemophilia B have been instrumental to our global understanding of gene therapy and have significantly contributed to the rapid expansion of the field. The use of adeno-associated viruses (AAVs) as vectors for gene transfer has successfully led to therapeutic expression of coagulation factor IX (FIX) in severe haemophilia B patients. Expression of FIX has remained stable following a single administration of vector for up to 8 years at levels that are clinically relevant to reduce the incidence of spontaneous bleeds and have permitted a significant change in the disease management with reduction or elimination of the need for coagulation factor concentrates. These trials have also shed light on several concerns around AAV-mediated gene transfer such as the high prevalence of pre-existing immunity against the vector capsid as well as the elevation of liver transaminases that is associated with a loss of FIX transgene expression in some patients. However, this field is advancing very rapidly with the development of increasingly more efficient strategies to overcome some of these obstacles and importantly raise the possibility of a functional cure, which has been long sought after. This review overviews the evolution of gene therapy for haemophilia B over the last two decades.


Sujet(s)
Hémophilie B , Humains , Hémophilie B/génétique , Hémophilie B/thérapie , Vecteurs génétiques , Thérapie génétique , Facteur IX/génétique , Facteur IX/usage thérapeutique , Facteur IX/métabolisme , Hémorragie/traitement médicamenteux , Dependovirus/génétique , Dependovirus/métabolisme
20.
Blood Coagul Fibrinolysis ; 35(3): 94-100, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38358898

RÉSUMÉ

OBJECTIVE: Andexanet alfa is a targeted reversal agent for life threatening hemorrhage associated with direct acting oral anticoagulants (DOACs), but there is uncertainty regarding the benefit when compared to 4-factor prothrombin complex concentrate (4F-PCC) for this indication. We investigated the clinical outcomes and cost associated with reversal of DOACs in the setting of life-threatening intracranial hemorrhage (ICH). METHODS: A retrospective evaluation was conducted to evaluate patients with ICH in the setting of anticoagulation with DOAC from 9/1/2013 to 4/30/2020. Patients were included in the study if they received reversal with either andexanet alfa or 4F-PCC. RESULTS: Eighty-nine patients were included in the study. There was no statistically significant difference in 30-day mortality between patients who received andexanet alfa or 4F-PCC (52% vs. 35%, P  = 0.14). Radiographic stability of bleed was identified in 57% of patients receiving andexanet alfa vs. 58% of patients receiving 4F-PCC ( P  = 0.93). Median length of stay was not different between the andexanet alfa and 4F-PCC populations (7 days [IQR 6 - 12] vs. 6 days [IQR 3-12], P  = 0.66). Median cost of reversal agent was higher in patients receiving andexanet alfa compared to 4F-PCC ($15 000 [IQR 15 000-$27 000] vs. $11 650 [IQR $8567-$14 149]). CONCLUSION: Among patients with life-threatening intracranial hemorrhage in the setting of DOAC therapy, no clinical differences were observed with respect to selection of reversal agent. Prothrombin complex concentrates remain a viable alternative to reversal of DOAC therapy though multicenter, randomized, prospective studies are needed to further evaluate the role of 4F-PCC in the reversal of DOAC therapy.


Sujet(s)
Anticoagulants , Facteurs de la coagulation sanguine , Facteur Xa , Hémorragies intracrâniennes , Protéines recombinantes , Humains , Anticoagulants/usage thérapeutique , Facteur IX/usage thérapeutique , Hémorragie/traitement médicamenteux , Hémorragies intracrâniennes/traitement médicamenteux , Études rétrospectives
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE