Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Québec; INESSS; mars 2022.
Não convencional em Francês | BRISA/RedTESA | ID: biblio-1367470

RESUMO

MANDAT: À la demande du fabricant CSL Behring Canada Inc., l'Institut national d'excellence en santé et en services sociaux (INESSS) a procédé à l'évaluation du produit du système du sang AfstylaMC (lonoctocog alfa), un facteur VIII (FVIII) de coagulation humain recombinant qui s'administre par voie intraveineuse. Au Canada, lonoctocog alfa est indiqué pour la prophylaxie de routine, la maîtrise et la prévention des épisodes hémorragiques ainsi que pour la maîtrise et la prévention des saignements dans un contexte périopératoire chez les adultes et les enfants atteints d'hémophilie A (déficit congénital en facteur VIII). Les indications visées pour cette réévaluation sont identiques à celle reconnue par Santé Canada. Les neuf FVIII suivants sont présentement inscrits sur la Liste des produits du système du sang du Québec et ont servi de comparateurs. Parmi ceux-ci se trouvent six produits à action standard : AdvateMC, HelixateMC, KovaltryMC, NuwiqMC, XynthaMC (inclus Xyntha SolofuseMC) et ZonovateMC (à action standard) ainsi que trois produits à longue action : AdynovateMC, EloctateMC et EsperoctMC. DÉMARCHE D'ÉVALUATION: Une revue des données issues de la littérature et de celles fournies par le fabricant a été réalisée afin de documenter l'efficacité, l'innocuité et l'efficience de lonoctocog alfa. Des données contextuelles et expérientielles issues de la consultation d'experts et de patients sont également présentées. Élaboration par l'INESSS d'une analyse d'efficience et d'impact budgétaire. BESOIN DE SANT: L'hémophilie A, causée par une défaillance du FVIII, se manifeste par des temps de coagulation plus longs que la normale. Dans les cas sévères, le déficit en FVIII mène à des épisodes de saignement fréquents aux articulations, appelés hémarthroses, et aux tissus mous en absence de traumatisme. La prophylaxie à l'aide de FVIII recombinant constitue le traitement privilégié. Celle-ci consiste en plusieurs injections intraveineuses hebdomadaires pour remplacer le FVIII manquant. Malgré une bonne prise en charge de l'hémophilie A au Québec, certaines lacunes liées aux traitements actuels demeurent. Outre le souhait d'un traitement curatif permanent, les besoins suivants ont été identifiés par les experts rencontrés : une meilleure prévention contre le développement d'inhibiteurs (anticorps neutralisants contre le FVIII), la prévention d'arthropathies hémophiliques et des douleurs chroniques, des traitements offrant une protection hémostatique supérieure qui perdure plus longtemps et l'atténuation des contraintes liées aux injections intraveineuses répétées. RÉSULTATS: Efficacité: Lonoctocog alfa est considéré comme un FVIII à action standard. Lonoctocog alfa apparait au moins aussi efficace que ses comparateurs pour prévenir les saignements, lorsqu'utilisé en prophylaxie. Lonoctocog alfa apparait aussi efficace que ses comparateurs pour traiter les saignements perthérapeutiques. Dans les études répertoriées, lonoctocog alfa démontre une efficacité hémostatique bonne ou excellente lors de chirurgies. Innocuité: Le profil d'innocuité de lonoctocog alfa est jugé acceptable. Qualité de vie: Aucune donnée sur l'impact de lonoctocog alfa sur la qualité de vie n'a été présentée. Perspective de l'expert: À la lumière des données disponibles, l'expert consulté est d'avis que l'efficacité de la prophylaxie de lonoctocog alfa est comparable à celle des comparateurs, soient tous les FVIII inscrits à la Liste. Selon l'expert, le profil d'innocuité de lonoctocog alfa est comparable aux autres options disponibles pour la population ciblée.


Assuntos
Humanos , Fator VIII/administração & dosagem , Hemofilia A/prevenção & controle , Avaliação em Saúde , Análise Custo-Benefício
2.
Brasília; CONITEC; set. 2021.
Não convencional em Português | BRISA/RedTESA | ID: biblio-1370304

RESUMO

INTRODUÇÃO: A hemofilia A é uma doença hereditária ligada ao cromossomo X, caracterizada pela deficiência ou anormalidade da atividade coagulante do fator VIII e representa a maioria dos casos de coagulopatias hereditárias, com aproximadamente 10.123 casos no país em 2016. A manifestação clínica mais frequente da doença é a hemorragia musculoesquelética, principalmente os sangramentos intra articulares (hemartroses) que afetam especialmente as articulações do joelho, tornozelo, cotovelo, ombro e coxofemoral. Hemartroses de repetição em uma mesma articulação podem levar à degeneração articular progressiva com perda funcional e, nos casos graves, os sangramentos ocorrem frequentemente sem causa aparente. O tratamento dos pacientes com hemofilia A requer a infusão intravenosa do fator de coagulação deficiente (FVIII), sendo feito sob demanda (tratamento do episódio hemorrágico) ou de forma profilática para manter os seus níveis séricos adequados, prevenindo os episódios hemorrágicos. O Programa de Coagulopatias do Ministério da Saúde disponibiliza o FVIII recombinante com meia-vida padrão para o uso em profilaxia. A tecnologia proposta consiste em um FVIII recombinante com meia-vida estendida. Os produtos com meia-vida estendida foram desenvolvidos mais recentemente com o objetivo de disponibilizar um tempo maior de FVIII na circulação sanguínea, proporcionando um intervalo maior entre as infusões e melhor proteção contra sangramentos. TECNOLOGIA: Alfadamoctocogue pegol. PERGUNTA: O uso de alfadamoctocogue pegol para profilaxia secundária em pacientes com hemofilia A, a partir de 12 anos, previamente tratados (e sem inibidores) é mais seguro, eficaz e custo-efetivo do que o tratamento padrão no SUS? EVIDÊNCIAS CLÍNICAS: O único estudo comparativo apresentado, ainda que com qualidade de evidência baixa, não demonstrou diferença estatisticamente significativa no principal desfecho, taxa anualizada de sangramento, entre a tecnologia proposta e a tecnologia disponibilizada no SUS. Dentre outros potenciais benefícios relacionados a menor frequência de infusão do alfadamoctocogue pegol, apenas um estudo avaliou isoladamente a satisfação dos pacientes com a tecnologia proposta, sem comparação à tecnologia padrão. Na ausência de evidências científicas demonstrando superioridade da tecnologia proposta, o demandante realizou um painel Delphi, com especialistas na área de hemofilia A que atuam no SUS em diferentes regiões do Brasil, que sugeriu que os pacientes mais beneficiados com o uso do alfadamoctocogue pegol seriam aqueles com perfil sangrador, farmacocinética desfavorável, baixa adesão ao tratamento e com alta atividade diária. AVALIAÇÃO ECONÔMICA: Um estudo de custo-minimização foi construído baseado na premissa do estudo de Batt, 2019 de que ambas as tecnologias possuem a mesma eficácia. Foram construídos cenários para início do tratamento em diferentes faixas etárias (12 e 30 anos) e cenários para diferentes utilizações de UIs dos medicamentos (cenário base e proposto). No horizonte da vida toda, os resultados para os pacientes iniciando com 12 anos de idade foi uma economia por paciente de R$ 1.342.233,18 e R$ 3.625.885,71 para os cenários base e proposto respectivamente. ANÁLISE DE IMPACTO ORÇAMENTÁRIO: Os resultados da análise de custo-efetividade foram utilizados para a construção da análise de impacto orçamentário. Dados do Perfil de Coagulopatias de 2016, do IBGE e do Painel Delphi foram utilizados para estimar o número de pacientes elegíveis ao tratamento. Estimou-se que de 30% a 40% dos pacientes teriam um perfil sangrador e seriam elegíveis a substituição pela formulação de liberação estendida (alfadamoctocogue pegol). Esses valores foram utilizados no market share proposto. Ao final de 5 anos, estimou-se uma economia de R$323.024.411,22. MONITORAMENTO DO HORIZONTE TECNOLÓGICO: Foram detectadas quatro tecnologias potenciais para o tratamento de pacientes com hemofilia a partir de 12 anos de idade. O Mim-8, um anticorpo IgG4 mimético do fator 8 de coagulação, que tem como alvos os fatores IX e X de coagulação, o fitusiran, um RNA silenciador (siRNA), direcionado ao RNA mensageiro (RNAm) codificador de antitrombina, e os anticorpos monoclonais IgG4 concizumabe e marstacimab, inibidores do inibidor da via do fator tissular (TFPI). Além dos potenciais medicamentos descritos, com mecanismos de ação diferentes dos fatores de coagulação, estão em fase 3 clínica os fatores VIII recombinantes FRSW-107, SCT-800 e efanesoctocog alfa (BIVV001). Além desses, foram registrados, em outros países, os fatores VIII recombinantes damoctocog alfa pegol (Japão, 2019); lonoctocog alfa (EUA, 2016) e turoctocog alfa pegol (Alemanha e Suíça, 2019). O alfadamoctocogue é protegido pela patente PI 0517795-2, depositada no Instituto Nacional da Propriedade Intelectual (INPI) em 14/11/2005, com validade até 31/03/30 (45). CONSIDERAÇÕES FINAIS: Não houve estudos de comparação direta que mostrasse que a intervenção é superior ou possui a mesma efetividade do comparador. Apenas um estudo de comparação indireta demonstrou não haver significância estatística entre as alternativas. A avaliação de custo-minimização e o impacto orçamentário mostraram resultados que representaram economia para o SUS com a incorporação da nova tecnologia. RECOMENDAÇÃO PRELIMINAR DA CONITEC: Diante do exposto, a Conitec, em sua 5ª reunião extraordinária, realizada no dia 12 de maio de 2021, deliberou que a matéria fosse disponibilizada em consulta pública com recomendação preliminar de todos os membros do plenário desfavorável à incorporação do alfadamoctocogue pegol para o tratamento de hemofilia A em profilaxia secundária para pacientes a partir de 12 anos de idade no SUS. Os membros do plenário concordaram que, apesar da comodidade posológica, diante da impossibilidade de desconto nos impostos, o impacto orçamentário que antes produziria uma economia, se torna um gasto próximo a 200 milhões de reais ao final de 5 anos. A matéria foi disponibilizada em consulta pública. CONSULTA PÚBLICA: Foram recebidas 3.387 contribuições, sendo 434 pelo formulário para contribuições técnico-científicas e 2.953 pelo formulário para contribuições sobre experiência ou opinião de pacientes, familiares, amigos ou cuidadores de pacientes, profissionais de saúde ou pessoas interessadas no tema. Das 434 contribuições de cunho técnico-científico recebidas, 66 foram analisadas, já que as demais não apresentaram informação alguma (em branco) ou argumentação técnica sobre as evidências. No total, 117 concordaram com a recomendação inicial da Conitec, 9 não concordaram e não discordaram e 308 discordaram. Das 117 que concordaram, 44 apresentaram algum comentário sobre essa opinião e todos eles discordavam da decisão da Conitec. Das 2.952 contribuições recebidas sobre experiência com a tecnologia ou opinião sobre o tema, 339 foram analisadas, já que as demais não apresentaram informação alguma (em branco). No total, 559 concordaram com a recomendação inicial da Conitec, 62 não concordaram e não discordaram e 2.331 discordaram. Os assuntos abordados pelos participantes que discordaram da recomendação preliminar foram majoritariamente relacionados ao acesso e a possibilidade de mais uma opção terapêutica, os ganhos em qualidade de vida e a comodidade posológica que a tecnologia traz aos pacientes. RECOMENDAÇÃO FINAL DA CONITEC: Pelo exposto, o Plenário da Conitec, em sua 101ª Reunião Ordinária, no dia primeiro de setembro de 2021, recomendou, por maioria simples, a incorporação de alfadamoctocogue pegol para a profilaxia secundária em pacientes com Hemofilia A, a partir de 12 anos, previamente tratados e sem inibidor no SUS, conforme Protocolo estabelecido pelo Ministério da Saúde. Considerou-se como justificativa para a decisão a maior vantagem posológica e a economia demonstrada ao SUS. Por fim, foi assinado o Registro de Deliberação nº 664/2021. DECISÃO: Incorporar o alfadamoctocogue pegol para profilaxia secundária em pacientes com Hemofilia A, no âmbito do Sistema Único de Saúde ­ SUS, conforme a Portaria nº 11, publicada no Diário Oficial da União nº 31, seção 1, página 71, em 14 de fevereiro de 2022.


Assuntos
Humanos , Fator VIII/administração & dosagem , Prevenção Secundária/instrumentação , Hemofilia A/prevenção & controle , Sistema Único de Saúde , Brasil , Análise Custo-Benefício/economia
3.
J Manag Care Spec Pharm ; 27(5): 674-681, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33908277

RESUMO

At an upfront price of $2.125 million, the one-time gene therapy onasemnogene abeparvovec for spinal muscular atrophy, a rare neuromuscular disorder that is usually fatal by 2 years of age if untreated, has been called the "most expensive drug ever." This flawed characterization raises important methodological and policy issues regarding valuation of high-cost treatments. We reviewed several other high-cost therapies-with a particular focus on hemophilia A treatment-studied by the nonprofit Institute for Clinical and Economic Review (ICER). In ICER's summary report of 2 treatments for managing hemophilia A, published in this month's JMCP issue, the estimated $15-$18 million lifetime cost of factor VIII is characterized as "far too high," representing "a failure of competition [that] … builds a platform for pricing of treatments … that will only exacerbate these problems." Current literature indicates several factors underlying high factor VIII treatment cost (eg, historical pattern of innovation and lack of market competition) that may also drive the pricing dynamics of advanced therapies for other rare diseases. When a treatment's price is driven high (or "distorted"), an economic principle known as "theory of the second best" suggests that market price becomes a poor estimate of social opportunity cost, and adjustments should be made for such distortions. In any case, a high-cost standard of care creates an opportunity for new technology to generate cost savings, providing an inducement for market entry. Recognizing that this potentially creates a tendency to produce price distortions for new treatments, ICER has attempted to apply some ad hoc adjustments. However, challenges remain in creating a "level playing field" across different disease-modifying or potentially curative innovations (eg, one-time therapy vs ongoing or lifelong treatment with repeated doses). While additional policy work is needed to address this dilemma, it would clearly be misleading to assume that gene therapies are inherently expensive. Rigorous economic evaluation of novel therapies requires careful comparison of lifetime cost and benefits vs standard of care, including adjustments for pricing distortions. Fortunately, economic theory suggests that we could adjust to this circumstance by using the social opportunity costs of interventions based on an appropriate variable cost-effectiveness threshold that would be higher for rare severe diseases. DISCLOSURES: The research reported in this Viewpoints article was funded by Novartis Gene Therapies, Inc. Garrison and Jiao were paid by Novartis Gene Therapies, Inc., to conduct this research. Garrison has also received consulting fees from BioMarin, Inc, and UniQure. Dabbous is a full-time employee of Novartis Gene Therapies, Inc., and holds Novartis stock and stock options.


Assuntos
Análise Custo-Benefício , Terapia Genética/economia , Terapia Genética/métodos , Custos de Medicamentos/estatística & dados numéricos , Fator VIII/administração & dosagem , Fator VIII/economia , Humanos
4.
J Manag Care Spec Pharm ; 27(3): 316-326, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33645244

RESUMO

BACKGROUND: Standard of care for bleed prevention in patients with severe congenital hemophilia A is continuous prophylaxis with factor VIII (FVIII), typically administered intravenously 2-3 times per week in the home setting. Nonfactor prophylaxis and gene therapy are emerging novel prophylaxis strategies for hemophilia A, and it is important to compare their health economics with that of FVIII prophylaxis. Current data on resource utilization and costs in the adult hemophilia A prophylaxis population are limited, and a structured approach to analyze annual costs in these patients using administrative claims data has not been previously reported. OBJECTIVE: To assess health care resource utilization and costs of continuous FVIII prophylaxis in commercially insured adults with hemophilia A without inhibitors. METHODS: Administrative claims records from beneficiaries covered by major selfinsured companies in the United States from January 1999 through March 2017 (OptumHealth Care Solutions) were queried, and records for adult patients (aged 18-64 years) diagnosed with hemophilia A who received FVIII were extracted. Three criteria were defined to distinguish patients most likely to be managed with continuous FVIII prophylaxis from those on episodic treatment based on the frequency and timing of FVIII claims over a 12-month period of continuous enrollment: (1) having ≥ 4 FVIII claims, (2) having ≥ 6 FVIII claims, or (3) having no gaps > 60 days between FVIII claims. Patients with evidence of bypassing agent use were excluded. Health care resource utilization and costs were assessed for all patients with any FVIII use and for patients defined as being managed with continuous FVIII prophylaxis based on each criterion. RESULTS: The analysis included 189 patients with a diagnosis code for hemophilia A (ICD 9-CM code 286.0; ICD-10-CM code D66) from January 1999 through March 2017 who had at least 12 months of continuous enrollment and at least 1 noninpatient/nonemergency department claim for FVIII concentrate (any type) during their last 12 months of continuous enrollment (overall cohort). Within the overall cohort, 118, 94, and 61 patients met the criteria for FVIII prophylaxis based on the first, second, and third definitions, respectively. Per patient mean (SD) total health care costs for the overall cohort was $287,055 (306,933). For patients meeting criteria 1 through 3, per patient costs ranged from $407,752 (321,036) to $551,645 (302,841). FVIII concentrate accounted for over 90% of costs, with mean (SD) annual FVIII costs of $264,777 (292,423) in the overall cohort and $384,197 (303,826), $433,029 (313,711), and $531,098 (297,142) among patients meeting the respective definitions for prophylaxis. CONCLUSIONS: This analysis highlights the substantial economic burden associated with managing adults with hemophilia A on FVIII prophylaxis, where per patient mean total annual health care costs ranged from $407,752 to $551,645. Over 90% of such costs were attributable to FVIII concentrate dispensed. DISCLOSURES: This study was funded by BioMarin Pharmaceutical, which was involved in protocol development, analysis plan development, data interpretation, manuscript preparation, and publication decisions. All authors contributed to protocol development, analysis plan development, data interpretation, and manuscript development. All authors maintained control over the final content. Sammon, Solari, Kim, and Hinds are employees and shareholders of BioMarin Pharmaceutical. Cook, Sheikh, and Chawla are employees of Analysis Group, a consulting company that was contracted by BioMarin Pharmaceutical to conduct this study and develop the manuscript. Croteau has received professional fees from BioMarin Pharmaceutical, Bayer, CSL Behring, Genentech, and Pfizer. Thornberg has received professional fees from BioMarin Pharmaceutical, Genentech, Novo Nordisk, Sanofi, and Spark Therapeutics, as well as research funding from Novo Nordisk and Sanofi.


Assuntos
Fator VIII/uso terapêutico , Custos de Cuidados de Saúde , Hemofilia A/tratamento farmacológico , Aceitação pelo Paciente de Cuidados de Saúde/estatística & dados numéricos , Adolescente , Adulto , Esquema de Medicação , Fator VIII/administração & dosagem , Feminino , Humanos , Infusões Intravenosas , Revisão da Utilização de Seguros , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Estados Unidos , Adulto Jovem
5.
Thromb Haemost ; 121(6): 731-740, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33506481

RESUMO

BACKGROUND: The pharmacokinetic (PK) properties of extended half-life (EHL) factor VIII (FVIII) concentrates differ, leading to variation in the optimal dosing regimen for the individual patient. The aim of this study was to establish these PK differences for various EHL FVIII concentrates by in silico simulations. METHODS: FVIII level over time profiles of rFVIII-SC, BAY 81-8973, rFVIII-Fc, BAX 855, BAY 94-9027, and standard half-life (SHL) rFVIII concentrates were simulated for 1,000 severe hemophilia A patients during steady-state dosing of 40 IU/kg every 72 hours or dosing as advised in the summary of product characteristics (SmPC). RESULTS: Although the elimination half-life values were comparable for rFVIII-FC, BAX 855, and BAY 94-9027, a higher area under the curve (AUC; 2,779 IU/h/dL) for BAY 94-9027 was obtained. During steady-state dosing of 40 IU/kg every 72 hours, 58.5% (rFVIII-SC), 69.3% (BAY 81-8972), 89.0% (rFVIII-Fc), 83.9% (BAX 855), and 93.7% (BAY 94-9027) of the patients maintained a trough level of 1 IU/dL, compared with 56.0% for SHL rFVIII. Following dosing schemes described in the SmPC, between 51.0 and 65.4% or 23.2 and 31.1% of the patients maintained a target trough level of 1 IU/dL or 3 IU/dL, respectively. CONCLUSION: BAY 94-9027 showed the largest increase of AUC and best target attainment compared with SHL rFVIII, followed closely by BAX 855 and rFVIII-Fc. BAY 81-8973 and rFVIII-SC showed smaller PK improvements. Although our analyses increase insight into the PK of these FVIII concentrates, more studies evaluating the relation between factor levels and bleeding risk are needed.


Assuntos
Coagulantes/farmacocinética , Fator VIII/farmacocinética , Hemofilia A/tratamento farmacológico , Modelos Biológicos , Polietilenoglicóis/farmacocinética , Proteínas Recombinantes de Fusão/farmacocinética , Coagulantes/administração & dosagem , Simulação por Computador , Cálculos da Dosagem de Medicamento , Fator VIII/administração & dosagem , Meia-Vida , Hemofilia A/sangue , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Método de Monte Carlo , Polietilenoglicóis/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem
6.
Haemophilia ; 26 Suppl 5: 3-10, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32935397

RESUMO

INTRODUCTION: Few studies, both in Australia and overseas, have examined the social impacts of living with haemophilia A (HA) or the economic costs associated with the disorder. The purpose of this paper is to examine the epidemiology and societal burden of people with HA (PwHA) in Australia, with a particular focus on men with this disorder. METHODS: The epidemiology and societal burden of HA in Australia, with a particular focus on men with this disorder, were assessed, using data available in the Australian and international literature and publicly available data. RESULTS: The mean annual prevalence of HA is approximately 1-2 per 10 000 males. Prophylactic treatment is used in one-quarter (25.1%) of people with moderate HA, and 82.2% of people with severe HA. Within the latter group, 16.1% have inhibitors for Factor VIII, predisposing them to worse morbidity, mortality and quality of life when compared to the non-inhibitor population. Joint pain and joint disease occur commonly in PwHA, with up to 70% of adults with HA experiencing joint problems. HA is associated with poor physical health, and PwHA miss school and work due to bleeding-related events. CONCLUSION: HA is associated with substantial economic burden; with large differences in costs reported between countries. Overall, HA imposes a significant burden of disease on PwHA, their families and the community at large.


Assuntos
Efeitos Psicossociais da Doença , Hemofilia A/epidemiologia , Adolescente , Adulto , Fatores Etários , Idoso , Austrália/epidemiologia , Criança , Pré-Escolar , Fator VIII/administração & dosagem , Fator VIII/efeitos adversos , Fator VIII/uso terapêutico , Saúde Global/estatística & dados numéricos , Hemofilia A/complicações , Hemofilia A/diagnóstico , Hemofilia A/terapia , Hospitalização , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Prevalência , Vigilância em Saúde Pública , Qualidade de Vida , Sistema de Registros , Índice de Gravidade de Doença , Fatores Sexuais , Adulto Jovem
7.
Blood ; 136(6): 740-748, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32369559

RESUMO

The bispecific antibody emicizumab is increasingly used for hemophilia A treatment. However, its specificity for human factors IX and X (FIX and FX) has limited its in vivo functional analysis to primate models of acquired hemophilia. Here, we describe a novel mouse model that allows emicizumab function to be examined. Briefly, FVIII-deficient mice received IV emicizumab 24 hours before tail-clip bleeding was performed. A second infusion with human FIX and FX, administered 5 minutes before bleeding, generated consistent levels of emicizumab (0.7-19 mg/dL for 0.5-10 mg/kg doses) and of both FIX and FX (85 and 101 U/dL, respectively, after dosing at 100 U/kg). Plasma from these mice display FVIII-like activity in assays (diluted activated partial thromboplastin time and thrombin generation), similar to human samples containing emicizumab. Emicizumab doses of 1.5 mg/kg and higher significantly reduced blood loss in a tail-clip-bleeding model using FVIII-deficient mice. However, reduction was incomplete compared with mice treated with human FVIII concentrate, and no difference in efficacy between doses was observed. From this model, we deducted FVIII-like activity from emicizumab that corresponded to a dose of 4.5 U of FVIII per kilogram (ie, 9.0 U/dL). Interestingly, combined with a low FVIII dose (5 U/kg), emicizumab provided enough additive activity to allow complete bleeding arrest. This model could be useful for further in vivo analysis of emicizumab.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Fator IX/administração & dosagem , Fator X/administração & dosagem , Hemofilia A/tratamento farmacológico , Hemorragia/tratamento farmacológico , Modelos Animais , Animais , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/imunologia , Quimioterapia Combinada , Fator IX/análise , Fator IX/imunologia , Fator VIII/administração & dosagem , Fator VIII/análise , Fator VIII/uso terapêutico , Fator X/análise , Fator X/imunologia , Fator XIa/farmacologia , Feminino , Hemofilia A/sangue , Hemofilia A/complicações , Hemofilia A/imunologia , Hemorragia/etiologia , Infusões Intravenosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Tempo de Tromboplastina Parcial , Cauda/lesões , Trombina/biossíntese
8.
Haemophilia ; 26(4): 584-590, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32432832

RESUMO

INTRODUCTION: The management of haemophilia is critical to minimize the risk of disability and reduce the burden on China's healthcare system. AIM: This study was based on a single centre in China and was conducted to understand the evolution of real-world haemophilia care over the past 15 years. METHODS: We retrospectively analysed clinical characteristics, diagnosis, treatment and medical expenditures of 428 patients with haemophilia from January 2004 to December 2018 from the Institute of Hematology & Blood Diseases Hospital in Tianjin, China. RESULTS: The delayed diagnosis time significantly decreased from 13.3 ± 5.1 years before 2004 to 0.4 ± 0.4 year in 2014-2018 (P < .05). Among children and adults receiving prophylactic treatment, the annual factor consumption increased from 2004-2008 (168.8 IU/kg in children and 120.7 IU/kg in adults) to 2009-2013 (389.2 IU/kg in children and 316.2 IU/kg in adults) and 2014-2018 (1328.0 IU/kg in children and 878.8 IU/kg in adults, P < .001). The annual medical insurance expenditure for haemophilia had increased steadily over the past 10 years. The number of patients tested regularly for inhibitors increased from 2004 (1.9% [2/105]) to 2018 (21.5% [59/275]). The seroprevalence of hepatitis C virus (HCV) was 33.8% during the years examined, while the incidence rates of HCV among patients significantly decreased (7.3% in 2008 to 0.4% in 2018). CONCLUSION: Significant improvements in the management of haemophilia were observed from 2004 to 2018. These results highlight the joint effort of the reimbursement policy and drug regulatory management paving the way for a better future for patients with haemophilia in China.


Assuntos
Atenção à Saúde/economia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia A/prevenção & controle , Hepatite C/epidemiologia , Adolescente , Adulto , Criança , China/epidemiologia , Efeitos Psicossociais da Doença , Diagnóstico Tardio/estatística & dados numéricos , Fator VIII/administração & dosagem , Gastos em Saúde/estatística & dados numéricos , Hemofilia A/diagnóstico , Hemofilia A/economia , Hepacivirus/isolamento & purificação , Hepatite C/sangue , Humanos , Incidência , Masculino , Estudos Retrospectivos , Estudos Soroepidemiológicos , Adulto Jovem
9.
J Manag Care Spec Pharm ; 26(9): 1109-1120, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32452276

RESUMO

BACKGROUND: Hemophilia A (HA) can result in bleeding events because of low or absent clotting factor VIII (FVIII). Prophylactic treatment for severe HA includes replacement FVIII infusions and emicizumab, a bispecific factor IXa- and factor X-directed antibody. OBJECTIVE: To develop an economic model to predict the short- and long-term clinical and economic outcomes of prophylaxis with emicizumab versus short-acting recombinant FVIII among persons with HA in the United States. METHODS: A Markov model was developed to compare clinical outcomes and costs of emicizumab versus FVIII prophylaxis among persons with severe HA from U.S. payer and societal perspectives. Patients started prophylaxis at age 1 year in the base case. Mutually exclusive health states considered were "no arthropathy," "arthropathy," "surgery," and "death." Serious adverse events, breakthrough bleeds, and inhibitor development were simulated throughout the modeled time horizon. In addition to the prophylaxis drug costs, patients could incur other direct costs related to breakthrough bleeds treatment, serious adverse events, development of inhibitors, arthropathy, and orthopedic surgery. Indirect costs associated with productivity loss (i.e., missed work or disabilities) were applied for adults. Model inputs were obtained from the HAVEN 3 trial, published literature, and expert opinion. The model used a lifetime horizon, and results for 1 year and 5 years were also reported. Deterministic sensitivity analyses and scenario analyses were conducted to assess robustness of the model. RESULTS: Over a lifetime horizon, the cumulative number of all treated bleeds and joint bleeds avoided on emicizumab versus FVIII prophylaxis were 278.2 and 151.7, respectively. Correspondingly, arthropathy (mean age at onset: 12.9 vs. 5.4 years) and FVIII inhibitor development (mean age at development: 13.9 vs. 1.1 years) were delayed. Total direct and indirect costs were lower for emicizumab versus FVIII prophylaxis for all modeled time horizons ($97,159 vs. $331,610 at 1 year; $603,146 vs. $1,459,496 at 5 years; and $15,238,072 vs. $22,820,281 over a lifetime horizon). The sensitivity analyses indicated that clinical outcomes were sensitive to efficacy inputs, while economic outcomes were driven by the discount rate, dosing schedules, and treatments after inhibitor development. Results for moderate to severe patients were consistent with findings in the severe HA population. CONCLUSIONS: The model suggests that emicizumab prophylaxis confers additional clinical benefits, resulting in a lower number of bleeding events and delayed onset of arthropathy and inhibitor development across all time assessment horizons. Compared with short-acting recombinant FVIII, emicizumab prophylaxis leads to superior patient outcomes and cost savings from U.S. payer and societal perspectives. DISCLOSURES: Funding for this study was provided by Genentech. Raimundo and Patel are employees of Genentech and own stock or stock options. Zhou, Han, Ji, Fang, Zhong, and Betts are employees of Analysis Group, which received consultancy fees from Genentech for conducting this study. Mahajerin received consultancy fees from Genentech for work on this study. Portions of this research were presented as a poster at the 2018 American Society of Hematology Conference; December 1-4, 2018; San Diego, CA.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Fator VIII/administração & dosagem , Hemofilia A/tratamento farmacológico , Modelos Econômicos , Adolescente , Anticorpos Biespecíficos/economia , Anticorpos Monoclonais Humanizados/economia , Criança , Pré-Escolar , Coagulantes/administração & dosagem , Coagulantes/economia , Fator VIII/economia , Hemofilia A/economia , Humanos , Lactente , Artropatias/epidemiologia , Masculino , Cadeias de Markov , Fatores de Tempo , Estados Unidos
10.
J Manag Care Spec Pharm ; 26(4): 504-512, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32020842

RESUMO

BACKGROUND: Recombinant factor VIII (rFVIII) products have been developed with improved pharmacokinetics, offering some patients the potential to extend dosing intervals, thereby reducing their dosing frequency while minimizing the occurrence of bleeding events. No clinical trials have been conducted to compare the bleeding rates and use of these long-acting products. OBJECTIVES: To (a) assess real-world use of prophylaxis regimens in patients using 1 of 3 different long-acting products-rVIII-SingleChain, rFVIIIFc, or PEG-rFVIII; and (b) compare bleeding rates, dosing frequency, and factor consumption in 3 cohorts of patients. For rVIII-SingleChain patients, these measures were also compared with the prior products these patients used. METHODS: De-identified patient chart data were collected from 11 hemophilia treatment centers in the United States. Patients were included if they had been treated with rVIII-SingleChain, rFVIIIFc, or PEG-rFVIII prophylaxis for ≥ 8 weeks at the time of data collection. Matching for age and disease severity was attempted between the 3 patient groups. Data were also collected for patients who switched from their prior FVIII product to prophylaxis with rVIII-SingleChain. RESULTS: Data were obtained for 120 male patients. The majority of patients were dosing 2 times per week or less frequently (rVIII-SingleChain 65.0%, rFVIIIFc 70.0%, and PEG-rFVIII 72.5%). Annualized bleeding rates were comparable among the 3 cohorts, with median (mean) values of 2.0 (2.6) with rVIII-SingleChain and rFVIIIFc, and 3.0 (3.7) with PEG-rFVIII. The overall median (mean) FVIII consumption in IU per kg per week (IU/kg/week) was 91.9 (91.1) with rVIII-SingleChain, 108.5 (103.6) with rFVIIIFc, and 97.6 (111.0) with PEG-rFVIII, resulting in expected mean annual consumption of 322,140 IU, 361,816 IU, and 373,100 IU, respectively, for a 70 kg patient aged ≥12 years. The mean consumption was significantly different among the 3 products for all patients (P = 0.0164) and for those dosed 2 times per week (P < 0.0001). Among patients infusing 2 times per week, median (mean) consumption with rVIII-SingleChain was 83.8 (81.2) IU/kg/week, compared with 109.6 (104.4) IU/kg/week for rFVIIIFc and 92.1 (91.5) IU/kg/week for PEG-rFVIII. Additionally, switching from prophylaxis with prior FVIII products to rVIII-SingleChain increased the proportion of patients dosing ≤ 2 times per week (20% to 65%), decreased mean consumption (103.3 to 91.9 IU/kg/week; P = 0.0164), and maintained the mean annualized bleeding rates (2.9 to 2.6; P = 0.5665). CONCLUSIONS: Results for rVIII-SingleChain confirm the findings from its pivotal trial. Analyses of annualized bleeding rates demonstrate comparable clinical outcomes of rVIII-SingleChain to the other 2 long-acting products assessed. In patients aged ≥ 12 years, rVIII-SingleChain prophylaxis may result in an 11.0% and 13.7% lower mean factor consumption than rFVIIIFc and PEG-rFVIII, respectively, representing a potential cost-saving opportunity of 34% in both cases-at the current wholesale acquisition cost of the corresponding products. In addition, in patients using rVIII-SingleChain prophylactically, consumption was reduced compared with their prior products, while bleeding control was well maintained. DISCLOSURES: This study was funded by CSL Behring. Analyses were conducted by Adivo Associates. Maro is an employee of Adivo Associates. Desai and Yan are employees of CSL Behring. Simpson has received consulting honoraria for participation in advisory boards for CSL Behring, Genentech, Octapharma, and Bioverativ and speakers bureau for Bayer and Novo Nordisk. Data were presented in part at the Hemostasis and Thrombosis Research Society; May 9-11, 2019; New Orleans, LA, and at the International Society on Thrombosis and Haemostasis; July 6-10, 2019; Melbourne, Australia.


Assuntos
Fator VIII/administração & dosagem , Hemofilia A/tratamento farmacológico , Hemorragia/prevenção & controle , Adolescente , Criança , Redução de Custos , Esquema de Medicação , Custos de Medicamentos , Fator VIII/economia , Fator VIII/farmacocinética , Meia-Vida , Hemofilia A/complicações , Hemofilia A/economia , Hemorragia/epidemiologia , Hemorragia/etiologia , Humanos , Masculino , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/economia , Proteínas Recombinantes/farmacocinética , Estudos Retrospectivos , Fatores de Tempo , Resultado do Tratamento , Estados Unidos/epidemiologia
11.
Blood Transfus ; 18(5): 374-385, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-31855153

RESUMO

BACKGROUND: The recombinant factor VIII (rFVIII)-IgG1 Fc fusion protein (rFVIII-Fc) was the first available extended half-life rFVIII, shown to prolong dosing intervals of individualised prophylaxis in patients with severe haemophilia A, maintaining low bleeding rates and unchanged or lower FVIII dose versus standard half-life (SHL) rFVIII. Few data are available about real-world experience with rFVIII-Fc, including criteria for patient switching from SHL products, follow up and prophylaxis optimisation. MATERIALS AND METHODS: A single-centre retrospective study was designed to review patients switched to rFVIII-Fc, based on individual needs, after pharmacokinetic (PK) assessment, according to routine clinical practice. In patients with adequate post-switch follow up, data about rFVIII-Fc prophylaxis were compared with those from the last 18-months SHL rFVIII prophylaxis. RESULTS: Of 25 candidates, 18 patients (15 severe, 3 moderate; aged 9-62 years; 3 with inhibitor history) started rFVIII-Fc regimens, with comparable FVIII weekly dose and reduced infusion frequency (mean -30%) in all 17 patients previously on SHL rFVIII prophylaxis thrice weekly or every other day. Over a mean 18-month follow up in 13 patients, compared with SHL products, further reduced infusion frequency (mean -40%; p<0.001; interval ≥4 days in 9 patients), improved treatment satisfaction (Hemo-sat questionnaires), significantly lower FVIII weekly dose and annual consumption (mean -12%; p=0.019), comparable bleeding rates and FVIII trough levels, and improved management of breakthrough bleeding were observed. von Willebrand Factor Antigen (VWF:Ag) correlated to PK variables and both had relationships with rFVIII-Fc weekly dose, increasing statistical significance over the follow-up period. No inhibitors or drug-related adverse events were recorded. DISCUSSION: In this real-world series of patients, a switch to rFVIII-Fc, based on careful assessment of clinical needs, PK testing and treatment monitoring, was able to optimise individual convenience, efficacy and costs of prophylaxis.


Assuntos
Fator VIII , Hemofilia A , Hemorragia , Fragmentos Fc das Imunoglobulinas , Proteínas Recombinantes de Fusão , Adolescente , Adulto , Criança , Custos e Análise de Custo , Fator VIII/administração & dosagem , Fator VIII/economia , Fator VIII/farmacocinética , Seguimentos , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Hemofilia A/economia , Hemorragia/sangue , Hemorragia/economia , Hemorragia/prevenção & controle , Humanos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/economia , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/economia , Proteínas Recombinantes de Fusão/farmacocinética , Estudos Retrospectivos
12.
J Med Econ ; 22(12): 1328-1337, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31530050

RESUMO

Aims: Cumulative exogenous factor VIII (FVIII) exposure is an important predictor of developing neutralizing antibodies (inhibitors) to FVIII in patients with persons with hemophilia A (PwHA). The aim of this study was to model the costs of emicizumab versus FVIII prophylaxis and total treatment costs for patients with severe HA.Materials and Methods: An Excel-based decision model was developed to calculate cumulative costs in PwHA over a 20-year time horizon from the US payer perspective. The model considered persons with severe HA beginning at age 12 months with no prior FVIII exposure and initiating prophylaxis with emicizumab or FVIII. PwHA could develop inhibitors on accumulation of 20 FVIII exposure days. PwHA with inhibitors replaced FVIII with bypassing agents until inhibitors resolved spontaneously, following immune tolerance induction (ITI), or at the end of the time horizon. The primary model outcome was the difference in emicizumab versus FVIII treatment costs in 2019 USD. Sensitivity analyses were performed to test the robustness of results.Results: Total incremental cost over 20 years was -$1,945,480 (emicizumab arm, $4,919,058; FVIII arm, $6,864,538). Prophylaxis costs (emicizumab arm, $4,096,105; FVIII arm, $6,290,919) comprised the majority of costs in both groups, followed by breakthrough bleed treatment for the FVIII arm ($342,652) and ITI costs for the emicizumab arm ($733,671). Higher costs in the FVIII group reflected earlier inhibitor development (FVIII, 4 months; emicizumab, 162 months) and switch to bypassing agents.Limitations: The model design reflects a simplified treatment pathway for patients with severe HA who initiate FVIII or emicizumab prophylaxis. In the absence of clinical data, a key conservative assumption of the model is that patients receiving emicizumab and FVIII prophylaxis have the same risk of developing inhibitors.Conclusions: This study suggests that prophylaxis with emicizumab results in cost savings compared to FVIII prophylaxis in HA.


Assuntos
Anticorpos Biespecíficos/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Coagulantes/uso terapêutico , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemorragia/prevenção & controle , Anticorpos Biespecíficos/economia , Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais Humanizados/economia , Anticorpos Monoclonais Humanizados/imunologia , Coagulantes/administração & dosagem , Coagulantes/imunologia , Fator VIII/administração & dosagem , Fator VIII/imunologia , Humanos , Modelos Econômicos , Índice de Gravidade de Doença
13.
Brasília; CONITEC; fev. 2019. ilus, tab.
Não convencional em Português | BRISA/RedTESA | ID: biblio-997358

RESUMO

INTRODUÇÃO: A hemofilia A é caracterizada pela deficiência ou anormalidade do fator VIII da coagulação e seu tratamento consiste na reposição do fator deficiente, por meio dos concentrados de fator plasmático ou recombinante, uso de medicamentos adjuvantes e na profilaxia dos sangramentos. Segundo dados de 2015 do Perfil das Coagulopatias Hereditárias no Brasil, o número de pacientes com Hemofilia A era de 9.908 no Brasil. Uma das complicações de grande relevância para as pessoas com hemofilia refere-se ao desenvolvimento de inibidores, que são anticorpos direcionados contra os fatores VIII infundidos no tratamento. A indução de imunotolerância (ITI) corresponde a um tratamento de dessensibilização, que consiste na infusão diária ou em dias alternados do concentrado de fator deficiente, na tentativa de dessensibilizar o paciente. Os concentrados de fator VIII de origem plasmática e recombinante fazem parte do rol de tecnologias ofertada pelo SUS para o tratamento de pacientes com Hemofilia A e inibidores. TECNOLOGIA: Alfaefmoroctocogue (Eloctate®). PERGUNTA: O uso de Alfaefmoroctocogue (rFVIIIFc) é mais eficaz, seguro ou custo-efetivo na indução de imunotolerância em indivíduos com hemofilia A e inibidores quando comparado às opções disponíveis atualmente no SUS? EVIDÊNCIAS CIENTÍFICAS: Todos os estudos apresentados são considerados por baixo nível de evidências devido às limitações metodológicas e vieses relacionados ao tipo de estudo. Os resultados mostram que ITI com rFVIIIFc é eficaz e pode resultar em erradicação de inibidores e ITIs bem-sucedidas em muitos pacientes com alto risco de intolerância a ITI em primeira tentativa e em alguns pacientes submetidos à ITI de resgate. Além disso, ITI com rFVIIIFc demonstrou uma rápida diminuição no título de Bethesda e a indução de tolerância em um tempo mais curto na maioria dos pacientes em ITI pela primeira vez apesar de seus perfis de risco. Apesar do benefício demonstrado pelos estudos que avaliaram ao todo apenas 25 pacientes, estudos mais robustos são necessários para avaliar melhor a eficácia do rFVIIIFc no tratamento de ITI em pacientes com hemofilia A. AVALIAÇÃO ECONÔMICA: A análise de custo-minimização apresentada pelo demandante demonstrou o potencial de redução de custos proporcionado pela incorporação de Alfaefmoroctocogue ao SUS para tratamento de ITI. O modelo possui grandes limitações, principalmente relacionadas às premissas adotadas e as incertezas entre os parâmetros, sendo o próprio tipo de análise considerado inadequado. AVALIAÇÃO DE IMPACTO ORÇAMENTÁRIO: A análise de impacto orçamentário demonstrou um potencial de redução de custo de aproximadamente R$ 13,5 milhões acumulados no período de 5 anos. Apesar das limitações da análise esta economia pode ser maximizada caso a taxa de difusão de Alfaefmoroctocogue seja ainda maior do que a proposta pelo demandante (50% do mercado no quinto ano após a incorporação). MONITORAMENTO DO HORIZONTE TECNOLÓGICO: No horizonte foram detectadas duas tecnologias para indução de imunotolerância em pacientes com hemofilia A e inibidores, Fitusiran® (sem registro na ANVISA) e Emicizumabe (registrado na ANVISA em 2018). CONSIDERAÇÕES: São necessárias novas evidências para melhor compreensão dos benéficos clínicos do Alfaefmoroctocogue para indução de imunotolerância em pacientes com hemofilia A quando comparado às alternativas disponíveis atualmente no SUS. RECOMENDAÇÃO PRELIMINAR DA CONITEC: O plenário, em reunião da CONITEC realizada no dia 07 de novembro de 2018, recomendou que o tema fosse submetido à consulta pública com recomendação preliminar não favorável à incorporação do Alfaefmoroctocogue (Fator VIII de coagulação recombinante Fc) para tratamento de pacientes com hemofilia A e inibidores. Considerou-se que há grande incerteza a respeito da eficácia e segurança do medicamento quando comparado as opções de tratamento já disponíveis no SUS, devido aos poucos relatos encontrados na literatura e inexistência de estudos mais robustos e com maior número de pacientes, além disso a análise econômica apresentada e a análise de impacto orçamentário apresentaram limitações importantes que atribuíram elevada incerteza quanto as estimativas reais de custos e de impacto orçamentário. CONSULTA PÚBLICA: Foram recebidas 79 contribuições de experiência ou opinião e 11 contribuições de cunho técnico-científico, onde a maioria foi discordando com a recomendação preliminar da CONITEC. Todas as contribuições foram avaliadas quantitativamente e qualitativamente. As contribuições destacaram a importância da incorporação de mais uma opção terapêutica para o tratamento de imunotolerância de pacientes com hemofilia A. As contribuições científicas enviadas não apresentaram evidências adicionais sobre a eficácia, efetividade e segurança do Alfaefmoroctocogue. DELIBERAÇÃO FINAL: Os membros da CONITEC presentes na 74ª reunião ordinária, nos dias 06 e 07 de fevereiro de 2019, deliberaram, por unanimidade, não recomendar a incorporação ao SUS do medicamento Alfaefmoroctocogue (Fator VIII de coagulação recombinante Fc) para tratamento de pacientes com hemofilia A e inibidores. Foi assinado em 07 de fevereiro o registro de deliberação nº 421/2019. DECISÃO: Não incorporar o alfaefmoroctocogue (fator VIII de coagulação recombinante Fc) para indução de imunotolerância em pacientes com hemofilia A e inibidores, no âmbito do Sistema Único de Saúde - SUS. Dada pela Portaria nº 8, de 21 de fevereiro de 2019, publicada no Diário Oficial da União nº 38, de 22/02/2019, seção 1, página 54.


Assuntos
Humanos , Fator VIII/administração & dosagem , Hemofilia A , Avaliação da Tecnologia Biomédica , Avaliação em Saúde/economia , Sistema Único de Saúde , Brasil , Análise Custo-Benefício/economia
14.
Expert Rev Hematol ; 11(12): 937-943, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30449223

RESUMO

Introduction: rFVIIIFC was the first extended half-life product to complete the phase 3 development program and be registered. It was developed to reduce the high treatment burden imposed by prophylaxis. It is now one of four extended half-life products available for a variety of indications in hemophilia A. This article focus on the efficacy use of rFVIIIFC in the prevention of bleeds in hemophilia A. Areas covered: This article provides an update on efficacy data from three clinical studies describing the use of rFVIIIFC in the treatment and prevention of bleeds in hemophilia A. The update includes the efficacy use of rFVIII in all age groups, in the perisurgical setting, in immune tolerance induction, and in improving the quality of life of patients. The role of rFVIIIFC prophylaxis in the face of rapidly evolving non-replacement therapy and gene therapy is summarized. Expert commentary: The role of rFVIIIFC in hemophilia A prophylaxis is uncertain in the light of development of newer prophylaxis agents with better route of administration, improved pharmacokinetic and superior efficacy profiles. While rFVIIIFC was primarily developed for prophylaxis in hemophilia A, this role may change in the face of competitive extended half-life products and non-replacement therapies.


Assuntos
Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Fragmentos Fc das Imunoglobulinas/uso terapêutico , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Fator VIII/administração & dosagem , Fator VIII/economia , Fator VIII/farmacologia , Hemofilia A/prevenção & controle , Hemostasia/efeitos dos fármacos , Humanos , Tolerância Imunológica/efeitos dos fármacos , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/economia , Fragmentos Fc das Imunoglobulinas/farmacologia , Qualidade de Vida , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/economia , Proteínas Recombinantes de Fusão/farmacologia , Resultado do Tratamento
15.
Manag Care ; 27(10): 39-50, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30309448

RESUMO

PURPOSE: To identify international units (IUs) dispensed and consequent expenditures for standard half-life (SHL) versus extended half-life (EHL) recombinant factor VIII (rFVIII) replacement products in hemophilia A patients in a real-world setting. DESIGN: Two U.S. claims databases were analyzed. METHODOLOGY: Number of IUs dispensed and quarterly expenditures for rFVIII products were collected from the Optum Clinformatics Data Mart and Truven Health MarketScan Databases. Truven claims were also analyzed for factor IUs dispensed and expenditures for patients with data for ≥3 months before and after switching to an EHL product. RESULTS: The Optum and Truven databases, respectively, included 276 (SHL, n=243; EHL, n=33) and 500 (SHL, n=409; EHL, n=91) hemophilia A patients. Median quarterly factor IUs dispensed in Optum were 10% higher with EHL versus SHL products over nine quarters, and 45% higher with EHL versus SHL products in Truven over 10 quarters. Median quarterly expenditures in the EHL cohort were 51% (individual quarterly medians range, 1%-101%) higher than in the SHL cohort in Optum and 122% higher (individual quarterly medians range, 1%-189%) in Truven. Twenty-nine Truven patients switched to an EHL product; median factor IUs dispensed varied quarterly. The lowest SHL and highest EHL values occurred in the quarter immediately before switching and the first quarter post-switch, respectively. Overall median quarterly expenditures were higher post-switch; this was consistent over seven quarters. CONCLUSION: We found higher expenditures over two years for hemophilia A patients using EHL versus SHL products. Switching to an EHL rFVIII product was associated with variable factor IUs dispensed and consistently higher expenditures.


Assuntos
Fator VIII/administração & dosagem , Fator VIII/economia , Gastos em Saúde , Hemofilia A/tratamento farmacológico , Custos e Análise de Custo , Estudos Transversais , Bases de Dados Factuais , Substituição de Medicamentos/economia , Meia-Vida , Humanos , Revisão da Utilização de Seguros , Masculino , Estudos Retrospectivos
17.
J Thromb Haemost ; 15(11): 2115-2124, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28836341

RESUMO

Essentials High-quality data are lacking on use of prophylaxis in adults with hemophilia and arthropathy. SPINART was a 3-year randomized clinical trial of late/tertiary prophylaxis vs on-demand therapy. Prophylaxis improved function, quality of life, activity and pain but not joint structure by MRI. Prophylaxis improves function but must start before joint bleeding onset to prevent arthropathy. SUMMARY: Background Limited data exist on the impact of prophylaxis on adults with severe hemophilia A and pre-existing joint disease. Objectives To describe 3-year bleeding, joint health and structure, health-related quality-of-life (HRQoL) and other outcomes from the open-label, randomized, multinational SPINART study. Patients/Methods Males aged 12-50 years with severe hemophilia A, ≥ 150 factor VIII exposure days, no inhibitors and no prophylaxis for > 12 consecutive months in the past 5 years were randomized to sucrose-formulated recombinant FVIII prophylaxis or on-demand therapy (OD). Data collected included total and joint bleeding events (BEs), joint structure (magnetic resonance imaging [MRI]), joint health (Colorado Adult Joint Assessment Scale [CAJAS]), HRQoL, pain, healthcare resource utilization (HRU), activity, and treatment satisfaction. Results Following 3 years of prophylaxis, adults maintained excellent adherence, with a 94% reduction in BEs despite severe pre-existing arthropathy; 35.7% and 76.2% of prophylaxis participants were bleed-free or had fewer than two BEs per year, respectively. As compared with OD, prophylaxis was associated with improved CAJAS scores (least squares [LS] mean, - 0.31 [n = 42] versus + 0.63 [n = 42]) and HAEMO-QoL-A scores (LS mean, + 3.98 [n = 41] versus - 6.00 [n = 42]), less chronic pain (50% decrease), and approximately two-fold less HRU; activity, Euro QoL-5D-3L (EQ-5D-3L) scores and satisfaction scores also favored prophylaxis. However, MRI score changes were not different for prophylaxis versus OD (LS mean, + 0.79 [n = 41] versus + 0.96 [n = 38]). Conclusions Over a period of 3 years, prophylaxis versus OD in adults with severe hemophilia A and arthropathy led to decreased bleeding, pain, and HRU, better joint health, activity, satisfaction, and HRQoL, but no reduction in structural arthropathy progression, suggesting that pre-existing joint arthropathy may be irreversible.


Assuntos
Fator VIII/administração & dosagem , Hemartrose/prevenção & controle , Hemofilia A/tratamento farmacológico , Hemostasia/efeitos dos fármacos , Hemostáticos/administração & dosagem , Articulações/efeitos dos fármacos , Adolescente , Adulto , Artralgia/diagnóstico , Artralgia/etiologia , Artralgia/prevenção & controle , Criança , Efeitos Psicossociais da Doença , Esquema de Medicação , Fator VIII/efeitos adversos , Hemartrose/diagnóstico por imagem , Hemartrose/etiologia , Hemofilia A/sangue , Hemofilia A/complicações , Hemofilia A/diagnóstico , Hemostáticos/efeitos adversos , Humanos , Articulações/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Adesão à Medicação , Pessoa de Meia-Idade , Medição da Dor , Satisfação do Paciente , Estudos Prospectivos , Qualidade de Vida , Índice de Gravidade de Doença , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
18.
Haemophilia ; 23(5): e394-e402, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28641362

RESUMO

INTRODUCTION: The International Immune Tolerance Study (I-ITI) demonstrated comparable success rates between low (FVIII 50 IU/kg/TIW) and high dose (FVIII 200 IU/kg/day) regimens. While costlier, the high dose ITI regimen achieved shorter time-to-treatment success with fewer bleeding episodes compared to the low dose ITI regimen. Adding bypassing agent prophylaxis (BAP) to a low dose ITI regimen may reduce bleeding while still being less costly than high dose ITI. AIM AND METHODS: An economic model was developed to compare high dose ITI to low dose ITI with BAP. All model inputs were derived from clinical trials. The I-ITI study indicated a median time to negative inhibitor titre of 4.6 and 9.2 months and average number of bleeds/patient of 4.2 and 9.9 for the high and low dose regimens respectively. Based on the BAP trials, aPCC (85 U/kg/TIW) and rFVIIa (90 µg/kg/day) achieved a 62% and 45% reduction in bleeding frequency respectively. Cost analysis was from a US third party payer perspective and limited to drug costs. One-way, two-way and probabilistic sensitivity analyses were performed. RESULTS: Costs of low dose ITI with aPCC prophylaxis until negative inhibitor titre is achieved was 24.0% less compared to high dose ITI. Low dose ITI with rFVIIa prophylaxis cost 46.5% more compared to high dose ITI. Model results were robust in the majority of the sensitivity analyses. CONCLUSION: A low dose ITI regimen with aPCC prophylaxis may be cost saving compared to a high dose ITI regimen with the potential to reduce morbidity by lowering the risk for breakthrough bleeds.


Assuntos
Análise Custo-Benefício , Custos de Medicamentos , Fator VIII/administração & dosagem , Hemofilia A/tratamento farmacológico , Hemorragia/prevenção & controle , Tolerância Imunológica , Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Tomada de Decisão Clínica , Gerenciamento Clínico , Fator VIII/efeitos adversos , Fator VIII/imunologia , Hemofilia A/complicações , Hemofilia A/imunologia , Humanos , Isoanticorpos/imunologia , Modelos Econômicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/efeitos adversos , Proteínas Recombinantes/imunologia
19.
Blood Coagul Fibrinolysis ; 28(6): 425-430, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27898515

RESUMO

: The objective of this study was to assess the cost-effectiveness of pharmacokinetic-driven prophylaxis in severe haemophilia A patients. A microsimulation model was developed to evaluate the cost-effectiveness of pharmacokinetic-driven prophylaxis vs. standard prophylaxis and estimate cost, annual joint bleed rate (AJBR), and incremental cost-effectiveness ratio over a 1-year time horizon for a hypothetical population of 10 000 severe haemophilia A patients. A dose of 30 IU/kg per 48 h was assumed for standard prophylaxis. Pharmacokinetic prophylaxis was individually adjusted to maintain trough levels at least 1 and 5 IU/dl or less. AJBR was estimated on the relationship between factor VIII (FVIII) levels and bleeding rate reported in the literature. Sensitivity analyses were performed to assess the stability of the model and the reliability of results. The FVIII dose was reduced in the 27.8% of patients with a trough level more than 5 IU/dl on standard prophylaxis, with a negligible impact on AJBR (+0.1 bleed/year). The FVIII dose was increased in the 10.6% of patients with trough levels less than 1 IU/dl on standard prophylaxis, with a significant reduction of AJBR (-1.9 bleeds/year). On average, overall, pharmacokinetic-driven prophylaxis was shown to decrease the AJBR from 1.012 to 0.845 with a slight reduction of the infusion dose of 0.36 IU/kg, with total saving of 5 197&OV0556; per patient-year. Pharmacokinetic-driven prophylaxis was preferable (i.e. more effective and less costly) compared with standard prophylaxis, with savings of 31 205&OV0556; per bleed avoided. Pharmacokinetic-driven prophylaxis, accounting for patients' individual pharmacokinetic variability, appears to be a promising strategy to improve outcomes with efficient use of available resources in severe haemophilia A patients.


Assuntos
Análise Custo-Benefício , Hemofilia A/economia , Farmacocinética , Pré-Medicação/métodos , Fator VIII/administração & dosagem , Fator VIII/economia , Hemartrose/economia , Hemartrose/prevenção & controle , Hemofilia A/tratamento farmacológico , Humanos , Pré-Medicação/economia
20.
Expert Rev Hematol ; 9(12): 1203-1208, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27809624

RESUMO

INTRODUCTION: Prophylactic factor replacement is the standard of care for all people with severe hemophilia to prevent bleeding and associated complications. Current weight-based fixed dose prophylaxis regimens are effective; however, they lack flexibility and usually fail to meet the individual needs and expectations of the patients. Recent developments in hemophilia treatment provide new opportunities for a more personalized prophylaxis. Areas covered: Rationale and methods of individualizing prophylaxis in hemophilia A on the basis of current evidence are discussed in this review. For this relevant literature in English and German was searched using PubMed database. Expert commentary: Major determinants of personalized prophylaxis include age, bleeding pattern, personal pharmacokinetics, joint health, co-morbidities, venous access and adherence. An ideal prophylaxis programme should take into account all of the aforementioned items and also be able to meet the needs. Extended half-life factor concentrates, new hemostatic molecules and tools using population pharmacokinetics to estimate personal factor requirements will serve individualizing prophylaxis in a more precise manner.


Assuntos
Hemofilia A/complicações , Hemorragia/etiologia , Hemorragia/prevenção & controle , Medicina de Precisão , Pré-Medicação , Análise Custo-Benefício , Fator VIII/administração & dosagem , Fator VIII/uso terapêutico , Hemartrose/etiologia , Hemartrose/prevenção & controle , Humanos , Variantes Farmacogenômicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA