Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Diagnostics (Basel) ; 13(22)2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37998531

RESUMEN

A previous study investigated robustness of manual flash (MF) and robust optimized (RO) volumetric modulated arc therapy plans for breast radiotherapy based on five patients in 2020 and indicated that the RO was more robust than the MF, although the MF is still current standard practice. The purpose of this study was to compare their plan robustness in terms of dose variation to clinical target volume (CTV) and organs at risk (OARs) based on a larger sample size. This was a retrospective study involving 34 female patients. Their plan robustness was evaluated based on measured volume/dose difference between nominal and worst scenarios (ΔV/ΔD) for each CTV and OARs parameter, with a smaller difference representing greater robustness. Paired sample t-test was used to compare their robustness values. All parameters (except CTV ΔD98%) of the RO approach had smaller ΔV/ΔD values than those of the MF. Also, the RO approach had statistically significantly smaller ΔV/ΔD values (p < 0.001-0.012) for all CTV parameters except the CTV ΔV95% and ΔD98% and heart ΔDmean. This study's results confirm that the RO approach was more robust than the MF in general. Although both techniques were able to generate clinically acceptable plans for breast radiotherapy, the RO could potentially improve workflow efficiency due to its simpler planning process.

2.
Haemophilia ; 29(2): 488-497, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36528890

RESUMEN

INTRODUCTION: EHL FVIII products and emicizumab provide clinicians with other prophylactic options for treating hemophilia A, however, it is unclear if emicizumab is a cost-saving option. The objective of this study is to estimate the health and economic effects of using prophylactic EHL FVIII, SHL FVIII, and emicizumab in severe haemophilia A patients. MATERIALS AND METHODS: A state-transition Markov model evaluated the cost-effectiveness of prophylactic SHL FVIII, EHL FVIII, and emicizumab in a cohort of 2-year-old male patients over a lifetime horizon in the form of a cost-utility analysis using a Canadian provincial ministry of health payer perspective. The transition probabilities, costs, and utilities were obtained from literature and the Canadian Bleeding Disorders Registry. Probabilistic sensitivity and scenario analyses were performed to test the robustness of the model. RESULTS: The base-case analysis, over a lifetime horizon, resulted in a total cost and utilities per person for SHL FVIII, EHL FVIII, and emicizumab of $27.2 million (M), $36.7 M, and $26.2 M, respectively, and 31.30, 31.16, and 31.61 quality-adjusted life years, respectively. Emicizumab treatment resulted in 29 and 16 less bleeds in a lifetime compared to SHL FVIII and EHL FVIII, respectively. Probabilistic sensitivity analysis showed that emicizumab was cost-saving 100% of the time compared to SHL FVIII and EHL FVIII. CONCLUSION: The cost-utility analysis showed that emicizumab is more effective and may be less costly than FVIII for Canadian haemophilia A patients, conditional on drug cost assumptions. Our model indicates that emicizumab may be a potentially favourable treatment option for minimising healthcare costs and providing higher effectiveness.


Asunto(s)
Anticuerpos Biespecíficos , Hemofilia A , Masculino , Humanos , Preescolar , Hemofilia A/tratamiento farmacológico , Análisis Costo-Beneficio , Canadá , Anticuerpos Biespecíficos/uso terapéutico , Hemorragia/prevención & control , Factor VIII/uso terapéutico , Factor VIII/farmacología
3.
Haemophilia ; 27(3): 358-365, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33650745

RESUMEN

INTRODUCTION: Emicizumab is dosed as mg/kg and, according to the label, any unused drug left in the vial(s) must be discarded, thereby wasting expensive resources. The aim of this study was to use population pharmacokinetics to illustrate the implications of changing the dosing interval to avoid wastage. METHODS: We used a previously published emicizumab PopPK model after extending its validation to children. We simulated PK parameters for labelled dosing regimens and for regimens using full vials with infusion frequency varied to keep the steady-state drug concentration unchanged. Cost and drug savings were calculated. RESULTS: The model evaluation was successful. When rounding up, the average individual below 53, 47 and 39 has a time-to-trough increase of up to 5.7, 7.9 and 5.8 days for the QW, Q2 W and Q4 W regimen, respectively. This resulted in an annual cost reduction of up to $173,136, $75,747 and $61,319 USD per patient. At higher body weights, rounding down the dose to the nearest vial resulted in negligible changes in the steady state concentration and cost savings of up to $93,781, $46,891 and $23,446 USD per patient, respectively. CONCLUSION: Individuals with a lower body weight may benefit from increasing dose intervals and rounding up dose up to the nearest vial, and individuals with a higher body weight from maintaining the injection frequency and rounding dose down to the nearest vial without significant change in emicizumab levels. Administering the entire vial may result in a reduction of vials used annually and potential cost savings.


Asunto(s)
Anticuerpos Biespecíficos , Anticuerpos Monoclonales Humanizados , Costos de los Medicamentos , Anticuerpos Biespecíficos/farmacocinética , Anticuerpos Monoclonales Humanizados/farmacocinética , Niño , Ahorro de Costo , Humanos
4.
Thromb Res ; 196: 550-558, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33157394

RESUMEN

INTRODUCTION: Hemophilia A is a genetic bleeding disorder resulting from a lack of clotting factor VIII. Where extended half-life products are available, people with hemophilia may stop their current drug regimen and switch to a EHL product providing a more convenient dosing regimen. While most factor VIII concentrate regimens are started prophylactically based on international units per weight, this "one-size-fits-all" approach does not account for the large pharmacokinetic variability between individuals. AIMS: We explored methods to predict individual PK of an EHL product by using population pharmacokinetic models and eta-values (η), a value that quantifies how individuals deviate from a population for any PK parameter, derived from a prior product. In addition, we wanted to investigate which individuals would benefit from this method compared to using a PopPK model alone. METHODS: PK data from subjects (n = 39) who have taken both Adynovate and Eloctate was collected from clinical trial data and from the Web-Accessible Population Pharmacokinetic Service - Hemophilia (WAPPS-Hemo) database. In addition, PK data from subjects (n = 200) who switched from a standard half-life product to Eloctate was also extracted from the WAPPS-Hemo database. Two methods to estimate individual PK outcomes of the second product were compared. The PopPK method used the Eloctate PopPK model published from WAPPS-Hemo, while the η-method incorporated individually scaled η from the prior product's PopPK model. Both methods were assessed for its performance in predicting PK outcomes. Absolute percent differences were calculated between the predicted and observed PK outcomes. Infusions were parsed into subgroups based on number of samples and individual η-percentiles for analysis. RESULTS: For the three switching protocols (Adynovate to Eloctate, Eloctate to Adynovate, and SHL FVIII to Eloctate), the η-method resulted in a relative difference reduction in mean absolute percent difference of 27.8% (range 1-59%), 4.9% (range 0-129%), and 18.0% (0-79%) in half-life compared to the PopPK method respectively. With some exceptions (in particular central volume), the η-method produced relative difference reduction in mean absolute percent differences up to 33% lower compared to the PopPK method. When individuals were parsed based on their η-values (either CL or V1), the two methods differentiate up to 64% in terms of half-life and time to 0.02 IU/mL predictions for individuals with a low (0th to 20th percentile) ηCL or ηV1 on the first product. Individuals with higher number of observations per infusion on the first product resulted in better predictions in PK parameter estimates when using the η-method. CONCLUSION: The use of prior knowledge by implementing η-values into PopPK models may provide clinicians with a safer and more effective method to choose a dosing regimen for patients with hemophilia A switching from one factor concentrate to another. However, the η-method was unable to better predict an increase or decrease in half-life of a future product compared to the PopPK method, and thus supports the conclusion that most individuals would still benefit from a trial on the EHL and subsequent estimation of their individual PK profile from sparse measurements on the EHL.


Asunto(s)
Hemofilia A , Hemostáticos , Bases de Datos Factuales , Factor VIII/uso terapéutico , Semivida , Hemofilia A/tratamiento farmacológico , Hemorragia , Humanos
5.
Haemophilia ; 26(3): 384-400, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32281726

RESUMEN

BACKGROUND: The Web-Accessible Population Pharmacokinetic Service (WAPPS) project generates individually predicted pharmacokinetic (PK) profiles and tailored prophylactic treatment regimens for haemophilic patients, which rely on a set of population PK (PopPK) models providing concentrate-specific priors for the Bayesian forecasting methodology. AIM: To describe the predictive performance of the WAPPS PopPK models in use on the WAPPS-Hemo platform. METHODS: Data for modelling include dense PK data obtained from industry sponsored and independent PK studies, and dense and sparse data accumulated through WAPPS-Hemo. WAPPS PopPK models were developed via non-linear mixed-effect modelling taking into account the effects of covariates and between-individual-and sometimes between-occasion-variability. Model evaluation consisted of (a) prediction-corrected Visual Predictive Check (pcVPC), (b) Limited Sampling Analysis (LSA) and (c) repeated hold-out cross-validation. RESULTS: Thirty-three WAPPS PopPK models built on data from 3188 patients (ages 1-78 years) under treatment by factor VIII or IX products (FVIII, FIX) were evaluated. Overall, models exhibit excellent performance characteristics. The pcVPC shows that the observed PK data fall within acceptable 90% interpercentile predictive bands. A slight overprediction beyond the expected half-life, an anticipated result of using sparse data, occurs for some models. The LSA results in lower than 3% of relative error for FVIII and FIX products and 16% for engineered FIX products. Cross-Validation analysis yields relative errors lower than 1.5% and 1.4% in estimates of half-life and time to 0.02 IU/mL, respectively. CONCLUSION: The WAPPS-Hemo models consistently showed excellent performance characteristics for the intended use for Bayesian forecasting of individual PK profiles.


Asunto(s)
Factor IX/farmacocinética , Factor IX/uso terapéutico , Factor VIII/farmacocinética , Factor VIII/uso terapéutico , Hemofilia A/genética , Femenino , Humanos , Masculino
6.
Thromb Res ; 184: 31-37, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31689604

RESUMEN

INTRODUCTION: This study proposes a method to predict individual pharmacokinetics of a future product by using the individual pharmacokinetic profile on the current product and the PopPK models of the current and future product. METHODS: Individual dense data was collected from two PK crossover studies, one enrolling 29 patients switching from Advate to Eloctate and one enrolling 15 patients switching from Advate to Novoeight. Three methods were designed to predict the second product's individual PK parameters (CL, V1, Q, and V2). Method 1 used the second product's typical population value of PK parameters from its PopPK model. Method 2 used the second product's calculated PK parameters based on individual covariates and its PopPK model. Method 3 used method 2, along with the predicted η-values of CL and V1 from the first product and its PopPK model. Each method was used to assess PK prediction during switching from Advate to Novoeight, Novoeight to Advate, and Advate to Eloctate. RESULTS: The three methods produced different outcomes. The mean absolute relative errors for half-life were lowest for method 3 for each study (11.6%, 13.1%, 13.6%). The regression line between predicted and observed half-life for method 3 was closest to the line of identity for each study (0.84, 0.67, 0.66). CONCLUSION: Taking into account individual PK from a previous clotting factor product was shown to provide better means of estimating individual PK for a new product. This may improve regimen design across switches and reduce the time to tailor optimal dose of FVIII products.


Asunto(s)
Teorema de Bayes , Factor VIII/farmacocinética , Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/uso terapéutico , Adulto , Femenino , Humanos , Masculino , Adulto Joven
7.
Res Pract Thromb Haemost ; 3(3): 528-541, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31294337

RESUMEN

The objective of this scoping review is to summarize the current use of pharmacokinetics for tailoring prophylaxis in hemophilia patients switching between clotting factor products. Patients with hemophilia may require switching of clotting factor concentrates due to a variety of factors, but there have been perceived risks associated with switching, such as inhibitor development or suboptimal protection due to inadequate dosing while titrating treatment. Studies that look at patients switching from one clotting factor concentrate to another are categorized in terms of their primary and/or secondary objectives, notably biosimilarity and comparative pharmacokinetic studies and inhibitor development studies. Research on how best to switch concentrates with respect to dosing regimen are lacking, and currently a trial-and-error approach is used for dosing the new factor concentrate. In the future, studies looking at the predictability of pharmacokinetics (PK) of a new factor concentrate based on individual PK knowledge of the original factor concentrate may offer clinical benefit by providing a safer switching approach and protocol.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...