Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 508
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Osteoporos Int ; 35(7): 1173-1183, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38565690

RESUMO

This study evaluated the cost-effectiveness of sequential treatment with romosozumab-to-alendronate compared to alendronate monotherapy and teriparatide-to-alendronate, in postmenopausal osteoporotic women from a Belgian healthcare perspective. Romosozumab-to-alendronate was found to be cost-effective compared to alendronate monotherapy and dominant compared to teriparatide-to-alendronate for osteoporotic women at high risk of fracture in Belgium. PURPOSE: This study aimed to evaluate the cost-effectiveness of sequential treatment with romosozumab followed by alendronate compared to alendronate monotherapy and teriparatide followed by alendronate, in postmenopausal osteoporotic women at high risk of fracture, from a Belgian healthcare perspective. Romosozumab is reimbursed in Belgium since December 2021. METHODS: A Markov microsimulation model was used to evaluate the cost-effectiveness of romosozumab-to-alendronate compared to alendronate monotherapy and to teriparatide-to-alendronate over a lifetime horizon. Patients transition between five different health states every 6 months based on fracture risks or death. The model was populated with Belgium-specific epidemiological and cost data, where available. The fracture risk reduction of romosozumab treatment was collated from the ARCH study, and from a published network meta-analysis. Costs were included from a healthcare perspective (NIHDI). Cost-effectiveness was reported in terms of costs per quality-adjusted life year (QALY), reported in Euro (€) 2022. Deterministic (DSA) and probabilistic sensitivity analyses (PSA) were performed. RESULTS: Romosozumab-to-alendronate was associated with 0.12 additional QALYs at an additional cost of €2314 compared to alendronate monotherapy, resulting in an ICER of €19,978. Compared to teriparatide-to-alendronate, romosozumab-to-alendronate was found to be dominant, with higher QALYs and lower costs. The base-case results were robust to uncertainty in the input parameters when conducting the sensitivity analysis. CONCLUSION: Sequential treatment with romosozumab followed by alendronate was found to be cost-effective compared to alendronate monotherapy and dominant compared to teriparatide followed by alendronate for postmenopausal women with osteoporosis at high risk of fracture in Belgium.


Assuntos
Alendronato , Anticorpos Monoclonais , Conservadores da Densidade Óssea , Análise Custo-Benefício , Custos de Medicamentos , Cadeias de Markov , Osteoporose Pós-Menopausa , Fraturas por Osteoporose , Anos de Vida Ajustados por Qualidade de Vida , Teriparatida , Humanos , Feminino , Fraturas por Osteoporose/prevenção & controle , Fraturas por Osteoporose/economia , Fraturas por Osteoporose/epidemiologia , Conservadores da Densidade Óssea/uso terapêutico , Conservadores da Densidade Óssea/economia , Bélgica/epidemiologia , Osteoporose Pós-Menopausa/tratamento farmacológico , Osteoporose Pós-Menopausa/economia , Osteoporose Pós-Menopausa/complicações , Alendronato/uso terapêutico , Alendronato/economia , Alendronato/administração & dosagem , Teriparatida/uso terapêutico , Teriparatida/economia , Teriparatida/administração & dosagem , Idoso , Custos de Medicamentos/estatística & dados numéricos , Anticorpos Monoclonais/economia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Quimioterapia Combinada , Pessoa de Meia-Idade , Esquema de Medicação , Substituição de Medicamentos/economia , Substituição de Medicamentos/estatística & dados numéricos
3.
Nature ; 603(7899): 25-27, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35233098

Assuntos
Antivirais/uso terapêutico , Tratamento Farmacológico da COVID-19 , Ensaios Clínicos como Assunto , Reposicionamento de Medicamentos , Interações Hospedeiro-Patógeno/efeitos dos fármacos , SARS-CoV-2/efeitos dos fármacos , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/uso terapêutico , Administração Oral , Alanina/administração & dosagem , Alanina/análogos & derivados , Alanina/uso terapêutico , Animais , Anti-Inflamatórios/administração & dosagem , Anti-Inflamatórios/uso terapêutico , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/economia , Anticorpos Neutralizantes/uso terapêutico , Antivirais/administração & dosagem , Antivirais/farmacologia , COVID-19/economia , COVID-19/imunologia , COVID-19/mortalidade , COVID-19/virologia , Vacinas contra COVID-19 , Citidina/análogos & derivados , Citidina/uso terapêutico , Depsipeptídeos/farmacologia , Depsipeptídeos/uso terapêutico , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Combinação de Medicamentos , Sinergismo Farmacológico , Ésteres/farmacologia , Ésteres/uso terapêutico , Guanidinas/farmacologia , Guanidinas/uso terapêutico , Hospitalização , Humanos , Hidroxilaminas/uso terapêutico , Internacionalidade , Lactamas/uso terapêutico , Leucina/uso terapêutico , Camundongos , National Institutes of Health (U.S.)/organização & administração , Nitrilas/uso terapêutico , Fator 1 de Elongação de Peptídeos/antagonistas & inibidores , Peptídeos Cíclicos/farmacologia , Peptídeos Cíclicos/uso terapêutico , Prolina/uso terapêutico , Inibidores de Proteases/farmacologia , Inibidores de Proteases/uso terapêutico , RNA Polimerase Dependente de RNA/antagonistas & inibidores
4.
Pharm Res ; 39(2): 239-250, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35118567

RESUMO

PURPOSE: We have hypothesized that a high concentration of circulating monocytes and macrophages may contribute to the fast weight-based clearance of monoclonal antibodies (mAbs) in young children. Exploring this hypothesis, this work uses modeling to clarify the role of monocytes and macrophages in the elimination of mAbs. METHODS: Leveraging pre-clinical data from mice, a minimal physiologically-based pharmacokinetic model was developed to characterize mAb uptake and FcRn-mediated recycling in circulating monocytes, macrophages, and endothelial cells. The model characterized IgG disposition in complex scenarios of site-specific FcRn deletion and variable endogenous IgG levels. Evaluation was performed for predicting IgG disposition with co-administration of high dose IVIG. A one-at-a-time sensitivity analysis quantified the role of relevant cellular parameters on IgG elimination in various scenarios. RESULTS: The plasma AUC of mAbs was highly sensitive to endothelial cell parameters, but had near-nil sensitivity to monocyte and macrophage parameters, even in scenarios with 90% loss of FcRn expression/activity. In mice with normal FcRn expression, simulations suggest that less than 2% of an IV dose is eliminated in macrophages, while endothelial cells are predicted to dominate mAb elimination. CONCLUSIONS: The model suggests that the role of monocytes and macrophages in IgG homeostasis includes extensive uptake and highly efficient FcRn-mediated protection, but not appreciable degradation when FcRn is present. Therefore, it is very unlikely that a high concentration of circulating monocytes can contribute to explaining the fast weight-based clearance of mAbs in very young children, even if FcRn expression/activity was 90% lower in children than in adults.


Assuntos
Anticorpos Monoclonais/farmacocinética , Antígenos de Histocompatibilidade Classe I/metabolismo , Imunoglobulina G/metabolismo , Macrófagos/metabolismo , Modelos Biológicos , Monócitos/metabolismo , Receptores Fc/metabolismo , Animais , Anticorpos Monoclonais/administração & dosagem , Vias de Eliminação de Fármacos , Células Endoteliais/metabolismo , Antígenos de Histocompatibilidade Classe I/genética , Imunoglobulina G/administração & dosagem , Imunoglobulinas Intravenosas/administração & dosagem , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Fc/genética
5.
Future Oncol ; 18(3): 301-309, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34709061

RESUMO

Background: The introduction of daratumumab into the treatment of multiple myeloma has improved outcomes in patients; however, community oncologists often dose more frequently than the US FDA-approved label. Materials and methods: Integra analyzed its database to elucidate daratumumab treatment patterns and the impact of increased utilization on the cost of care for multiple myeloma. Results: Following week 24, 671 (65%) of 1037 patients remained on daratumumab-containing regimens, with 330 patients continuing more frequent treatments than the expected once-every-4-weeks dosing described in the standard dosing schedule. Patients received an average of 14% more daratumumab doses than the FDA-approved label indicates, increasing the 1-year daratumumab costs by an estimated US$31,353. Conclusion: Daratumumab is utilized more frequently than the FDA-recommended dosing, leading to higher multiple myeloma treatment costs.


Lay abstract Since its first approval in 2015, daratumumab has become the backbone of many multiple myeloma treatment regimens. While its approval has improved outcomes in many patients who undergo treatment, it is expensive and has largely contributed to the increasing costs of care in multiple myeloma. In its most common treatment schedule, patients should transition from weekly and biweekly dosing to treatment once every 4 weeks. However, many providers maintain their patients on a more frequent dosing schedule, which increases Medicare 1-year costs by an estimated US$31,353 and may have unforeseen impacts on adverse events and patient outcomes.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Custos e Análise de Custo/estatística & dados numéricos , Uso de Medicamentos/estatística & dados numéricos , Mieloma Múltiplo/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Anticorpos Monoclonais/economia , Esquema de Medicação , Custos de Medicamentos/estatística & dados numéricos , Uso de Medicamentos/economia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Intervalo Livre de Progressão , Estudos Retrospectivos , Estados Unidos
6.
J Manag Care Spec Pharm ; 28(1): 108-114, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34949111

RESUMO

DISCLOSURES: Funding for this summary was contributed by Arnold Ventures, The Donaghue Foundation, Harvard Pilgrim Health Care, and Kaiser Foundation Health Plan to the Institute for Clinical and Economic Review (ICER), an independent organization that evaluates the evidence on the value of health care interventions. ICER's annual policy summit is supported by dues from AbbVie, America's Health Insurance Plans, Anthem, Alnylam, AstraZeneca, Biogen, Blue Shield of CA, Boehringer-Ingelheim, Cambia Health Services, CVS, Editas, Evolve Pharmacy, Express Scripts, Genentech/Roche, GlaxoSmithKline, Harvard Pilgrim, Health Care Service Corporation, HealthFirst, Health Partners, Humana, Johnson & Johnson (Janssen), Kaiser Permanente, LEO Pharma, Mallinckrodt, Merck, Novartis, National Pharmaceutical Council, Pfizer, Premera, Prime Therapeutics, Regeneron, Sanofi, Sun Life Financial, uniQure, and United Healthcare. Agboola, Herron-Smith, Nhan, Rind, and Pearson are employed by ICER. Through their affiliated institutions, Atlas, Brouwer, Carlson, and Hansen received funding from ICER for the work described in this summary.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/economia , Dermatite Atópica/tratamento farmacológico , Inibidores de Janus Quinases/administração & dosagem , Inibidores de Janus Quinases/economia , Antineoplásicos Imunológicos , Análise Custo-Benefício , Política de Saúde , Humanos , Anos de Vida Ajustados por Qualidade de Vida , Resultado do Tratamento
9.
Lima; Instituto Nacional de Salud; sept. 2021.
Não convencional em Espanhol | LILACS, BRISA/RedTESA | ID: biblio-1354733

RESUMO

INTRODUCCIÓN: La enfermedad por coronavirus 2019 (COVID-19), causada por el coronavirus 2 del Síndrome respiratorio agudo grave (SARS-CoV-2) fue inicialmente reportada en Wuhan, China en diciembre de 2019 (2). El espectro de la enfermedad es amplio e incluye desde cuadros leves y autolimitados hasta neumonía atípica severa y progresiva, falla multiorgánica y muerte (3,4). La Organización Mundial de la Salud (OMS) al 29 de junio de 2021 reportó 2.6 millones de casos nuevos en la última semana, con más de 57 mil nuevas muertes reportadas (5). Desde la identificación inicial del SARS-CoV-2, hasta el momento de este reporte, la OMS ha identificado cuatro como variantes cuya patogenicidad es preocupante (Alpha: B.1.1.7, Beta: B.1.351, Gamma: P.1 y Delta: B.1.617.2) y que podrían modificar el curso de la pandemia (6). Casirivimab e Imdevimab son anticuerpos monoclonales tipo inmunoglobulina G1 (IgG1) humana, ambos se administran en conjunto y que se dirigen contra el virus SARS-CoV-2 que causa la COVID-19, ya que se unen a los epítopos no competitivos del dominio de unión al receptor (RBD) de la proteína espiga (S) del SARS-CoV-2. La combinación de estos anticuerpos monoclonales, denominada REGEN COV, ha recibido la Autorización de uso de emergencia (EUA) por la Administración de Alimentos y Medicamentos de los Estados Unidos (FDA) (7). La presente nota técnica tiene como objetivo describir la evidencia científica publicada respecto a la eficacia y efectividad de la combinación de los anticuerpos monoclonales Casirivimab e Imdevimab (REGEN COV) en la prevención de COVID-19. MÉTODOS: Se realizó una búsqueda sistemática hasta el 10 de setiembre de 2021 en las bases de datos científicas MEDLINE/PubMed, LILACS/Biblioteca virtual en salud (BVS) y L.OVE/Epistemonikos, incluyendo términos en lenguaje natural y lenguaje estructurado (Tesauros) según cada base de datos para el fármaco de interés: "REGEN-COV", "casirivimab", "imdevimab", "REGN10933" y "REGN10987"; "SARS-CoV-2", "COVID-19". Se incluyeron publicaciones de ensayos clínicos o en etapa de pre-impresión (manuscritos no certificados por una revisión por pares) que reporten resultados para al menos uno de los desenlaces. Se consideraron publicaciones en idioma: inglés, español o portugués. Adicionalmente, se incluyeron estudios de ensayos de neutralización que trataban de la efectividad del fármaco de interés frente a las variantes del SARS-CoV-2. Se excluyeron estudios realizados en animales. La selección de los estudios y extracción de los datos no fue pareada. No se hizo evaluación de riesgo de sesgo de las publicaciones incluidas. RESULTADOS: En la Revisión Rápida N° 04-2021 de UNAGESP, se realizó una búsqueda sistematizada hasta el 28 de junio de 2021 (1); en esta búsqueda se recuperaron 02 reportes correspondientes a las 02 partes de un ensayo clínico aleatorizado del fármaco de interés (Casirivimab e Imdevimab) para la prevención de infección sintomática por SARS-CoV-2. Adicionalmente, se realizó una búsqueda sistematizada hasta el 10 de setiembre en la que se recuperaron 04 documentos sobre la efectividad de este fármaco sobre las variantes de preocupación (VOC) del SARS-CoV-2. CONCLUSIONES: El objetivo de esta nota técnica fue describir la evidencia científica publicada respecto a la eficacia y efectividad de la combinación de los anticuerpos monoclonales Casirivimab e Imdevimab (REGEN COV) en la prevención de COVID-19. La intervención evaluada para la prevención de COVID-19 fue la combinación de anticuerpos monoclonales REGEN-COV (Casirivimab (REGN 10933) 600mg e Indevimab (REGN 10987) 60mg) que es administrada por vía subcutánea. Se identificó un ensayos clínicos aleatorizados (ECA) realizado en partes (parte A y B) que evaluaron la eficacia de la combinación REGEN COV (Casirivimab/Imdevimab) para prevenir infección sintomática por SARS-CoV-2 (COVID-19); no se identificó ningún estudio que evaluara la efectividad de este fármaco para la prevención de COVID-19. Se identificaron 4 estudios que evaluaron la capacidad de neutralización de estos anticuerpos frente a las variantes Alpha, Beta, Gamma y Delta del SARS-CoV-2 en estudios in vitro. La combinación REGEN COV (Casirivimab 600mg/Imdevimab 600mg) administrada por vía subcutánea, en pacientes con PCR negativo y exposición a un contacto intradomiciliario con infección por SARS-CoV-2, redujo el riesgo de desarrollar COVID-19 en un 81.4% en relación al riesgo de los que recibieron placebo (1.5% vs 7.8%, RR: 0.186, IC 95% 0.09 ­ 0.35). En relación a los eventos adversos, en la parte A del ECA revisado se reportaron 265/1311 (20.2%) eventos adversos en el grupo de intervención y 379/1306 (29%) en el grupo placebo. Los eventos adversos más frecuentes fueron cefalea y reacción en el lugar de inyección. La frecuencia de eventos adversos serios (EAS) fue de 10/1311 (0.8%) en el grupo intervención y de 15/1306 (1.1%) en el grupo placebo. Ninguno de los EAS fueron atribuidos a la vacuna. En el grupo de intervención no se reportó ningún evento de especial interés. Se reportaron 4 fallecimiento después del término del seguimiento, 2 en cada brazo, de los cuales ninguno fue atribuido a la vacunación. En cuanto a la capacidad de neutralización frente a las variantes de preocupación (VOC) del SARS-CoV-2, los estudios revisados informaron que la combinación REGEN COV (Casirivimab/Imdevimab) mantuvo la capacidad de neutralización contra la variante Alpha (B.1.1.7) y Delta (B.1.617.1 y B.1.617.2), y mantuvo parcialmente la capacidad de neutralización contra la variante Beta (B.1.351) y Gamma (P.1), probablemente debido a mutaciones K417N y E484K resistentes a Casirivimab.


Assuntos
Humanos , SARS-CoV-2/efeitos dos fármacos , COVID-19/prevenção & controle , Anticorpos Monoclonais/administração & dosagem , Eficácia , Análise Custo-Benefício , Combinação de Medicamentos
10.
Lima; Instituto Nacional de Salud; ago. 2021.
Não convencional em Espanhol | LILACS, BRISA/RedTESA | ID: biblio-1354107

RESUMO

ANTECEDENTES: La enfermedad por coronavirus 2019 (COVID-19), causada por el virus SARS-CoV-2 sigue produciendo millones de casos y muertes a nivel global hasta la fecha. Si bien ya existen vacunas eficaces para prevenir dicha enfermedad, algunos medicamentos han mostrado tener un rol terapéutico, como son los anticuerpos monoclonales específicos para proteínas del virus SARS-CoV-2. Por esta razón, se elaboró la presente Revisión Rápida con el fin de identificar y evaluar la evidencia disponible sobre este tema. OBJETIVO: El objetivo de esta revisión es identificar y sistematizar la evidencia disponible sobre la eficacia de los anticuerpos monoclonales específicos para SARS-CoV-2 previniendo el desarrollo de COVID-19, o mejorando los desenlaces clínicos de los pacientes con dicha enfermedad. METODOLOGÍA: Se realizó una revisión rápida basada en dos preguntas: a) En pacientes sin COVID-19 sintomático, ¿el uso de anticuerpos monoclonales específicos para SARS-CoV-2 comparado con no uso o placebo, es eficaz previniendo el desarrollo de COVID-19 sintomático? b) En pacientes con infección confirmada por SARS-CoV-2, ¿el uso de anticuerpos monoclonales específicos para SARS-CoV-2 comparado con no uso o placebo, es eficaz mejorando los desenlaces clínicos de los pacientes? Sólo se incluyeron ensayos clínicos aleatorizados o no. Para ello, se realizó una búsqueda sistemática en las bases de datos MEDLINE/PubMed, EMBASE/Ovid, LILACS, la Biblioteca Cochrane, MedRxiv, y BioRxiv. Luego de eliminar duplicados, fueron seleccionaron los ítems que cumplieran con la pregunta de investigación. No se evaluó riesgo de sesgo ni la calidad de la evidencia identificada. RESULTADOS: Se identificaron 14 estudios, 4 sobre prevención post exposición COVID-19, y 10 sobre eficacia mejorando desenlaces clínicos en pacientes con la enfermedad. De estos, 2 fueron en pacientes hospitalizados sin enfermedad crítica, y 8 en ambulatorios. Solo 5 de los 14 estudios fueron publicaciones científicas revisadas por pares. Los estudios encontrados evaluaron los anticuerpos monoclonales: Bamlanivimab (LY-CoV555), Bamlanivimab/Etesevimab (LY-CoV555+LY-CoV016), la combinación Casirivimab/Imdevimab (REGN10933+REGN10987), Sotrovimab (GSK4182136), Regdanvimab (CT-P59), y la combinación Cilgavimab/Tixagevimab (AZD7442, AZD8895+AZD1061). Respecto al desenlace de Prevención de infección sintomática post-exposición, Bamlanivimab 4200mg IV tendría una eficacia a las 8 semanas del 44%. Casirivimab/Imdevimab 600mg/600mg SC tendría una eficacia de 81.4% a los 28 días en pacientes PCR negativos al momento de recibir la medicación, y 31.5% en pacientes PCR positivos. Cilgavimab/Tixagevimab 300mg IM no tendría efecto significativo 30 a 180 días post-exposición, excepto en aquellos con PCR negativo basal, con una eficacia de 73%. Respecto a los desenlaces clínicos en pacientes hospitalizados con COVID-19, ni el estudio ACTIV-3 evaluando Bamlanivimab 7000mg IV, ni el estudio RECOVERY evaluando Casirivimab/Imdevimab 4000 mg/4000 mg IV, encontraron eficacia; ya sea mejorando la recuperación clínica, o disminuyendo muertes/ventilación mecánica, respectivamente. Sobre eficacia del tratamiento en pacientes ambulatorios, los tres reportes del estudio BLAZE-1 concluyen que Bamlanivimab IV combinado con Etesevimab (2800mg/2800mg) reduce la proporción de hospitalizaciones o visita a emergencia o muerte a los 29 días entre un 70% y 84%. El estudio de Regeneron® con Casirivimab/Imdevimab IV 1200mg/1200mg, y 4000mg/4000mg, muestra reducción significativa en atenciones médicas hasta el día 29 de 57% en general, 65% si tenían anticuerpos negativos basales, y 71% con factores de progresión. Otro reporte muestra reducción en hospitalizaciones o muerte de 71.3% (1200/1200mg) y 70.4% (600mg/600mg). Sotrovimab 500mg IV redujo hospitalización o muerte hasta el día 29 en 85%; y Regdanvimab IV redujo hospitalización o necesidad de oxígeno a los 28 días en 54% (40mg/Kg), y en 44% (80mg/Kg). Los tres reportes del estudio BLAZE-1 concluyen que Bamlanivimab IV no reduce la carga viral a los 11 días; en cambio, Bamlanivimab/Etesevimab 2800mg/2800mg IV si reduce la carga a los 7 y 11 días. Los reportes de Regeneron® concluyen que Casirivimab/Imdevimab IV reduce la carga viral al día 7. Regdanvimab no muestra diferencias significativas en la velocidad de negativización del PCR. Ningún estudio mostró diferencias en la proporción de eventos adversos o eventos adversos serios. CONCLUSIONES: Existe evidencia basada en ensayos clínicos, en su mayoría no revisados por pares, que los anticuerpos monoclonales serian medicamentos seguros y eficaces para prevenir enfermedad sintomática luego de exposición, sobre todo en aquellos con PCR negativo para SARS-CoV-2; y también mejorando los desenlaces clínicos (incluyendo hospitalización o muerte) en pacientes ambulatorios con COVID-19. No tendrían eficacia en el tratamiento de pacientes con COVID-19 ya hospitalizados.


Assuntos
Humanos , SARS-CoV-2/imunologia , COVID-19/prevenção & controle , COVID-19/tratamento farmacológico , Anticorpos Monoclonais/administração & dosagem , Eficácia , Análise Custo-Benefício
11.
Int J Toxicol ; 40(4): 322-336, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34255983

RESUMO

Myostatin is a negative regulator of skeletal muscle and has become a therapeutic target for muscle atrophying disorders. Although previous inhibitors of myostatin offered promising preclinical data, these therapies demonstrated a lack of specificity toward myostatin signaling and have shown limited success in the clinic. Apitegromab is a fully human, monoclonal antibody that binds to human promyostatin and latent myostatin with a high degree of specificity, without binding mature myostatin and other closely related growth factors. To support the clinical development of apitegromab, we present data from a comprehensive preclinical assessment of its pharmacology, pharmacokinetics, and safety across multiple species. In vitro studies confirmed the ability of apitegromab to inhibit the activation of promyostatin. Toxicology studies in monkeys for 4 weeks and in adult rats for up to 26 weeks showed that weekly intravenous administration of apitegromab achieved sustained serum exposure and target engagement and was well-tolerated, with no treatment-related adverse findings at the highest doses tested of up to 100 mg/kg and 300 mg/kg in monkeys and rats, respectively. Additionally, results from an 8-week juvenile rat study showed no adverse effects on any endpoint, including neurodevelopmental, motor, and reproductive outcomes at 300 mg/kg administered weekly IV. In summary, the nonclinical pharmacology, pharmacokinetic, and toxicology data demonstrate that apitegromab is a selective inhibitor of proforms of myostatin that does not exhibit toxicities observed with other myostatin pathway inhibitors. These data support the conduct of ongoing clinical studies of apitegromab in adult and pediatric patients with spinal muscular atrophy (SMA).


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/uso terapêutico , Distrofias Musculares/terapia , Miostatina/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Humanos , Macaca fascicularis , Masculino , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Testes de Toxicidade
13.
MAbs ; 13(1): 1868078, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33557682

RESUMO

The biosimilar concept is now well established. Clinical data accumulated pre- and post-approval have supported biosimilar uptake, in turn stimulating competition in the biologics market and increasing patient access to biologics. Following technological advances, other innovative biologics, such as "biobetters" or "value-added medicines," are now reaching the market. These innovative biologics differ from the reference product by offering additional clinical or non-clinical benefits. We discuss these innovative biologics with reference to CT-P13, initially available as an intravenous (IV) biosimilar of reference infliximab. A subcutaneous (SC) formulation, CT-P13 SC, has now been developed. Relative to CT-P13 IV, CT-P13 SC offers clinical benefits in terms of pharmacokinetics, with comparable efficacy, safety, and immunogenicity, as well as increased convenience for patients and reduced demands on healthcare system resources. As was once the case for biosimilars, nomenclature and regulatory pathways for innovative biologics require clarification to support their uptake and ultimately benefit patients.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Medicamentos Biossimilares/administração & dosagem , Desenvolvimento de Medicamentos , Infliximab/administração & dosagem , Administração Intravenosa , Animais , Anticorpos Monoclonais/efeitos adversos , Anticorpos Monoclonais/economia , Anticorpos Monoclonais/farmacocinética , Medicamentos Biossimilares/efeitos adversos , Medicamentos Biossimilares/economia , Medicamentos Biossimilares/farmacocinética , Análise Custo-Benefício , Difusão de Inovações , Composição de Medicamentos , Custos de Medicamentos , Desenvolvimento de Medicamentos/economia , Humanos , Infliximab/efeitos adversos , Infliximab/economia , Infliximab/farmacocinética , Injeções Subcutâneas
14.
J Clin Oncol ; 39(10): 1119-1128, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33411586

RESUMO

PURPOSE: The MAIA trial found that addition of daratumumab to lenalidomide and dexamethasone (DRd) significantly prolonged progression-free survival in transplant-ineligible patients with newly diagnosed multiple myeloma, compared with lenalidomide and dexamethasone alone (Rd). However, daratumumab is a costly treatment and is administered indefinitely until disease progression. Therefore, it is unclear whether it is cost-effective to use daratumumab in the first-line setting compared with reserving its use until later lines of therapy. METHODS: We created a Markov model to compare healthcare costs and clinical outcomes of transplant-ineligible patients treated with daratumumab in the first-line setting compared with a strategy of reserving daratumumab until the second-line. We estimated transition probabilities from randomized trials using parametric survival modeling. Lifetime direct healthcare costs, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratios (ICERs) were calculated for first-line daratumumab versus second-line daratumumab from a US payer perspective. RESULTS: First-line daratumumab was associated with an improvement of 0.52 QALYs and 0.66 discounted life-years compared with second-line daratumumab. While both treatment strategies were associated with considerable lifetime expenditures ($1,434,937 v $1,112,101 in US dollars), an incremental cost of $322,836 for first-line daratumumab led to an ICER of $618,018 per QALY. The cost of daratumumab would need to be decreased by 67% for first-line daratumumab to be cost-effective at a willingness-to-pay threshold of $150,000 per QALY. CONCLUSION: Using daratumumab in the first-line setting for transplant-ineligible patients may not be cost-effective under current pricing. Delaying daratumumab until subsequent lines of therapy may be a reasonable strategy to limit healthcare costs without significantly compromising clinical outcomes. Mature overall survival data are necessary to more fully evaluate cost-effectiveness in this setting.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Custos de Medicamentos/estatística & dados numéricos , Custos de Cuidados de Saúde/estatística & dados numéricos , Mieloma Múltiplo/tratamento farmacológico , Idoso , Anticorpos Monoclonais/administração & dosagem , Estudos de Coortes , Análise Custo-Benefício , Dexametasona/administração & dosagem , Humanos , Lenalidomida/administração & dosagem , Cadeias de Markov , Avaliação de Resultados em Cuidados de Saúde/métodos , Avaliação de Resultados em Cuidados de Saúde/estatística & dados numéricos , Intervalo Livre de Progressão , Anos de Vida Ajustados por Qualidade de Vida
15.
J Manag Care Spec Pharm ; 27(1): 112-117, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33377437

RESUMO

BACKGROUND: List prices of tumor necrosis factor (TNF) inhibitors drastically increased during the last decade, but previous research has shown that half of these increases were offset by rising manufacturer discounts. It remains unclear to what extent manufacturers' discounts have offset increases in list prices of each self-administered injectable TNF inhibitor. Evaluating trends in net prices and discounts at the product level will be paramount in understanding the role of competition in the biologic market. OBJECTIVES: To (a) describe product-level changes in net prices of each self-administered injectable TNF inhibitor available in 2007-2019 and (b) quantify to what extent manufacturer discounts have offset increases in list prices. METHODS: We obtained 2007-2019 pricing data for etanercept, adalimumab, certolizumab, and golimumab from the investment firm SSR Health, which uses company-reported sales to estimate net prices and discounts for brand products manufactured by publicly traded companies. For each drug and year, we calculated annual costs of treatment for patients with rheumatoid arthritis based on list and net prices and discounts in Medicaid and other payers. RESULTS: From 2007-2019, list prices of etanercept and adalimumab increased by 293% and 295%, respectively; however, discounts offset 47% and 45% of these increases, leading to net price increases of 171% and 203%. List prices of golimumab and certolizumab increased by 183% and 182%, respectively, but with discounts offsetting 58% and 59% of these increases, net prices increased by 103% and 109%. Net prices of golimumab started to decrease after 2016, while net prices of adalimumab and certolizumab experienced their first drop in 2019. Across the study period, discounts in Medicaid and in other payers increased, respectively, from 21% to 85% and 6% to 32% for etanercept; from 26% to 88% and 19% to 35% for adalimumab; from 28% to 63% and 22% to 46% for golimumab; and from 29% to 83% and 27% to 47% for certolizumab. CONCLUSIONS: Despite growing manufacturer discounts, net prices of self-administered injectable TNF inhibitors still increased at a mean annual rate of 9.6% in 2007-2019. This led to net prices tripling for adalimumab and more than doubling for etanercept, golimumab, and certolizumab. DISCLOSURES: This study was funded by the Myers Family Foundation. Hernandez is funded by the National Heart, Lung and Blood Institute (grant number K01HL142847). Funding sources had no role in design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; or decision to submit the manuscript for publication. Hernandez has served on Pfizer's scientific advisory board. The other authors have nothing to disclose.


Assuntos
Antirreumáticos/uso terapêutico , Custos de Medicamentos/tendências , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Adalimumab/administração & dosagem , Adalimumab/economia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/economia , Antirreumáticos/administração & dosagem , Etanercepte/administração & dosagem , Etanercepte/economia , Humanos , Injeções , Autoadministração , Estados Unidos
16.
Expert Rev Pharmacoecon Outcomes Res ; 21(2): 285-297, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32484365

RESUMO

Objective: To compare the cost of adverse events (AEs) associated with preventive treatment of migraine with fremanezumab, versus erenumab, galcanezumab, and onabotulinumtoxinA.Methods: A probabilistic modeling analysis was performed, using second-order Monte Carlo simulations, with 1,000 simulations, in patients with at least 4 days of migraine per month, from the perspective of the National Health System and a time horizon of 12 weeks. The frequency of AEs described in the clinical trials was analyzed with 12 weeks of treatment. Unit costs (€) of management of AEs were obtained from public health prices, expert panels, and published Spanish studies.Results: Fremanezumab would generate average savings of -€469 (95% CI -€303; -€674) versus erenumab, -€268 (95% CI -€171; -€391) versus galcanezumab, -€1,100 (95% CI -€704; -€1,608) or -€1,295 (95% CI -€835; -€1,893) versus onabotulinumtoxinA using real-life or clinical trial data, respectively.Conclusions: The different safety profile of treatment with fremanezumab, compared to erenumab, galcanezumab, and onabotulinumtoxinA, would generate savings in health-care resources in all the scenarios considered.


Assuntos
Anticorpos Monoclonais/efeitos adversos , Toxinas Botulínicas Tipo A/efeitos adversos , Custos de Cuidados de Saúde/estatística & dados numéricos , Transtornos de Enxaqueca/prevenção & controle , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/economia , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/economia , Toxinas Botulínicas Tipo A/administração & dosagem , Toxinas Botulínicas Tipo A/economia , Redução de Custos , Custos e Análise de Custo , Humanos , Transtornos de Enxaqueca/economia , Método de Monte Carlo , Probabilidade , Espanha
17.
Int J Clin Pharm ; 43(1): 181-190, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32909221

RESUMO

Background There is a strong rationale for fixed-dosing of monoclonal antibodies in oncology. Although fixed-dosing of recently introduced monoclonal antibodies is well accepted, the rationale is also applicable for other monoclonal antibodies that already have been used for years, but are still body-size-based dosed in many hospitals. In the Netherlands Cancer Institute, Antoni van Leeuwenhoek (NKI-AVL), fixed-dosing has been implemented now for all monoclonal antibodies and, therefore, this site offers an ideal opportunity for a cost analysis study. Objective To investigate the financial impact of switching to fixed-dosing in the NKI-AVL. Setting The NKI-AVL. Method Information on the preparations of monoclonal antibodies was collected from August 2017 to February 2020. We compared the number of vials needed during preparation for fixed-dosing and body-size -based dosing strategies. The economic impact was calculated for 2 scenarios: scenario 1 assumed clustering of all preparations per day and scenario 2 assumed no clustering of preparations. Main outcome measure Number of saved vials and the correlating savings in health care costs. Results The implementation of fixed-dosing resulted in a substantial reduction in vials used for almost all monoclonal antibodies. The economic savings were calculated to be €0,8 and €3,1 million per year for scenario 1 and 2, respectively. Conclusion Fixed-dosing resulted in substantial savings in health care costs.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Antineoplásicos Imunológicos/administração & dosagem , Neoplasias , Anticorpos Monoclonais/economia , Antineoplásicos Imunológicos/economia , Análise Custo-Benefício , Custos de Cuidados de Saúde , Humanos , Neoplasias/tratamento farmacológico , Países Baixos
18.
J Natl Cancer Inst ; 113(3): 282-291, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33227816

RESUMO

BACKGROUND: Whether social determinants of health are associated with survival in the context of pediatric oncology-targeted immunotherapy trials is not known. We examined the association between poverty and event-free survival (EFS) and overall survival (OS) for children with high-risk neuroblastoma treated in targeted immunotherapy trials. METHODS: We conducted a retrospective cohort study of 371 children with high-risk neuroblastoma treated with GD2-targeted immunotherapy in the Children's Oncology Group trial ANBL0032 or ANBL0931 at a Pediatric Health Information System center from 2005 to 2014. Neighborhood poverty exposure was characterized a priori as living in a zip code with a median household income within the lowest quartile for the cohort. Household poverty exposure was characterized a priori as sole coverage by public insurance. Post hoc analyses examined the joint effect of neighborhood and household poverty using a common reference. All statistical tests were 2-sided. RESULTS: In multivariable Cox regressions adjusted for disease and treatment factors, household poverty-exposed children experienced statistically significantly inferior EFS (hazard ratio [HR] = 1.90, 95% confidence interval [CI] = 1.28 to 2.82, P = .001) and OS (HR = 2.79, 95% CI = 1.63 to 4.79, P < .001) compared with unexposed children. Neighborhood poverty was not independently associated with EFS or OS. In post hoc analyses exploring the joint effect of neighborhood and household poverty, children with dual-poverty exposure (neighborhood poverty and household poverty) experienced statistically significantly inferior EFS (HR = 2.21, 95% CI = 1.48 to 3.30, P < .001) and OS (HR = 3.70, 95% CI = 2.08 to 6.59, P < .001) compared with the unexposed group. CONCLUSIONS: Poverty is independently associated with increased risk of relapse and death among neuroblastoma patients treated with targeted immunotherapy. Incorporation of social and environmental factors in future trials as health-care delivery intervention targets may increase the benefit of targeted therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Imunoterapia/economia , Neuroblastoma/tratamento farmacológico , Neuroblastoma/economia , Pobreza/estatística & dados numéricos , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/economia , Protocolos de Quimioterapia Combinada Antineoplásica/economia , Ensaios Clínicos Fase III como Assunto , Estudos de Coortes , Feminino , Humanos , Imunoterapia/métodos , Imunoterapia/estatística & dados numéricos , Lactente , Isotretinoína/administração & dosagem , Isotretinoína/uso terapêutico , Masculino , Estudos Multicêntricos como Assunto , Neuroblastoma/mortalidade , Modelos de Riscos Proporcionais , Ensaios Clínicos Controlados Aleatórios como Assunto , Características de Residência/estatística & dados numéricos , Estudos Retrospectivos , Estados Unidos/epidemiologia
19.
Expert Rev Pharmacoecon Outcomes Res ; 21(5): 1011-1016, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33086882

RESUMO

BACKGROUND: Spending on drugs provided by the Brazilian Public Health System (BPHS) for the treatment of rheumatoid arthritis (RA) increased substantially with the beginning of the supply of biological disease-modifying anti-rheumatic drugs (bDMARD). This study aims to perform a cost-utility analysis of the most used biological drugs for the treatment of RA in Brazil. METHODS: a Markov model was used to carry out the cost-utility analysis. The data were obtained from a prospective cohort of RA patients using adalimumab, etanercept, and golimumab in Brazil. The BPHS perspective was adopted and the time horizon was five years. Deterministic and probabilistic sensitivity analyses were performed to evaluate the uncertainty. RESULTS: golimumab was the most cost-effective drug. Etanercept was dominated by golimumab. Adalimumab presented an incremental cost-utility ratio (ICUR) of $95,095.37 compared to golimumab in five years of follow-up. These results were confirmed by sensitivity analyses. CONCLUSION: the utility among adalimumab, etanercept, and golimumab was similar and the cost was the component that most impacted the economic model. Therefore, depending on the agreed price with the drug manufacturers, the incremental cost-utility ratio may vary among them.


Assuntos
Antirreumáticos/administração & dosagem , Artrite Reumatoide/tratamento farmacológico , Modelos Econômicos , Inibidores do Fator de Necrose Tumoral/administração & dosagem , Adalimumab/administração & dosagem , Adalimumab/economia , Adulto , Idoso , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/economia , Antirreumáticos/economia , Artrite Reumatoide/economia , Brasil , Estudos de Coortes , Análise Custo-Benefício , Etanercepte/administração & dosagem , Etanercepte/economia , Feminino , Seguimentos , Humanos , Masculino , Cadeias de Markov , Pessoa de Meia-Idade , Estudos Prospectivos , Inibidores do Fator de Necrose Tumoral/economia
20.
J Crohns Colitis ; 15(5): 709-718, 2021 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-33125060

RESUMO

BACKGROUND AND AIMS: Anti-tumour necrosis factor alpha [anti-TNF] treatment accounts for 31% of health care expenditures associated with ulcerative colitis [UC]. Withdrawal of anti-TNF in patients with UC in remission may decrease side effects and infections, while promoting cost containment. Approximately 36% of patients relapse within 12-24 months of anti-TNF withdrawal, but reintroduction of treatment is successful in 80% of patients. We aimed to evaluate the cost-effectiveness of continuation versus withdrawal of anti-TNF in patients with UC in remission. METHODS: We developed a Markov model comparing cost-effectiveness of anti-TNF continuation versus withdrawal, from a health care provider perspective. Transition probabilities were calculated from literature, or estimated by an expert panel of 11 gastroenterologists. Deterministic and probabilistic sensitivity analyses were performed to account for assumptions and uncertainty. The cost-effectiveness threshold was set at an incremental cost-effectiveness ratio of €80,000 per quality-adjusted life-year [QALY]. RESULTS: At 5 years, anti-TNF withdrawal was less costly [-€10,781 per patient], but also slightly less effective [-0.04 QALY per patient] than continued treatment. Continuation of anti-TNF compared with withdrawal costs €300,390/QALY, exceeding the cost-effectiveness threshold. Continued therapy would become cost-effective if the relapse rate following anti-TNF withdrawal was ≥43% higher, or if adalimumab or infliximab [biosimilar] prices fell below €87/40 mg and €66/100 mg, respectively. CONCLUSIONS: Continuation of anti-TNF in UC patients in remission is not cost-effective compared with withdrawal. A stop-and-reintroduction strategy is cost-saving but is slightly less effective than continued therapy. This strategy could be improved by identifying patients at increased risk of relapse.


Assuntos
Anticorpos Monoclonais/economia , Medicamentos Biossimilares/economia , Colite Ulcerativa/tratamento farmacológico , Análise Custo-Benefício , Fármacos Gastrointestinais/economia , Infliximab/economia , Inibidores de Proteínas Quinases/economia , Adalimumab/administração & dosagem , Adalimumab/economia , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/economia , Medicamentos Biossimilares/administração & dosagem , Fármacos Gastrointestinais/administração & dosagem , Humanos , Infliximab/administração & dosagem , Cadeias de Markov , Piperidinas/administração & dosagem , Piperidinas/economia , Inibidores de Proteínas Quinases/administração & dosagem , Pirimidinas/administração & dosagem , Pirimidinas/economia , Anos de Vida Ajustados por Qualidade de Vida , Recidiva , Indução de Remissão , Ustekinumab/administração & dosagem , Ustekinumab/economia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA