Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Nutrients ; 15(20)2023 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-37892445

RESUMO

The global prevalence of type 2 diabetes mellitus (T2DM) has surged in recent decades, and the identification of differential glycemic responders can aid tailored treatment for the prevention of prediabetes and T2DM. A mixed meal tolerance test (MMTT) based on regular foods offers the potential to uncover differential responders in dynamical postprandial events. We aimed to fit a simple mathematical model on dynamic postprandial glucose data from repeated MMTTs among participants with elevated T2DM risk to identify response clusters and investigate their association with T2DM risk factors and gut microbiota. Data were used from a 12-week multi-center dietary intervention trial involving high-risk T2DM adults, comparing high- versus low-glycemic index foods within a Mediterranean diet context (MEDGICarb). Model-based analysis of MMTTs from 155 participants (81 females and 74 males) revealed two distinct plasma glucose response clusters that were associated with baseline gut microbiota. Cluster A, inversely associated with HbA1c and waist circumference and directly with insulin sensitivity, exhibited a contrasting profile to cluster B. Findings imply that a standardized breakfast MMTT using regular foods could effectively distinguish non-diabetic individuals at varying risk levels for T2DM using a simple mechanistic model.


Assuntos
Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Masculino , Adulto , Feminino , Humanos , Diabetes Mellitus Tipo 2/epidemiologia , Diabetes Mellitus Tipo 2/etiologia , Diabetes Mellitus Tipo 2/prevenção & controle , Glicemia/análise , Refeições , Fatores de Risco , Insulina
2.
CPT Pharmacometrics Syst Pharmacol ; 12(9): 1227-1237, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37300376

RESUMO

Progression-free survival (PFS) is an important clinical metric for comparing and evaluating similar treatments for the same disease within oncology. After the completion of a clinical trial, a descriptive analysis of the patients' PFS is often performed post hoc using the Kaplan-Meier estimator. However, to perform predictions, more sophisticated quantitative methods are needed. Tumor growth inhibition models are commonly used to describe and predict the dynamics of preclinical and clinical tumor size data. Moreover, frameworks also exist for describing the probability of different types of events, such as tumor metastasis or patient dropout. Combining these two types of models into a so-called joint model enables model-based prediction of PFS. In this paper, we have constructed a joint model from clinical data comparing the efficacy of FOLFOX against FOLFOX + panitumumab in patients with metastatic colorectal cancer. The nonlinear mixed effects framework was used to quantify interindividual variability (IIV). The model describes tumor size and PFS data well, and showed good predictive capabilities using truncated as well as external data. A machine-learning guided analysis was performed to reduce unexplained IIV by incorporating patient covariates. The model-based approach illustrated in this paper could be useful to help design clinical trials or to determine new promising drug candidates for combination therapy trials.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica , Humanos , Intervalo Livre de Progressão , Terapia Combinada , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
3.
BMC Cancer ; 23(1): 409, 2023 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-37149596

RESUMO

BACKGROUND: To increase the chances of finding efficacious anticancer drugs, improve development times and reduce costs, it is of interest to rank test compounds based on their potential for human use as early as possible in the drug development process. In this paper, we present a method for ranking radiosensitizers using preclinical data. METHODS: We used data from three xenograft mice studies to calibrate a model that accounts for radiation treatment combined with radiosensitizers. A nonlinear mixed effects approach was utilized where between-subject variability and inter-study variability were considered. Using the calibrated model, we ranked three different Ataxia telangiectasia-mutated inhibitors in terms of anticancer activity. The ranking was based on the Tumor Static Exposure (TSE) concept and primarily illustrated through TSE-curves. RESULTS: The model described data well and the predicted number of eradicated tumors was in good agreement with experimental data. The efficacy of the radiosensitizers was evaluated for the median individual and the 95% population percentile. Simulations predicted that a total dose of 220 Gy (5 radiation sessions a week for 6 weeks) was required for 95% of tumors to be eradicated when radiation was given alone. When radiation was combined with doses that achieved at least 8 [Formula: see text] of each radiosensitizer in mouse blood, it was predicted that the radiation dose could be decreased to 50, 65, and 100 Gy, respectively, while maintaining 95% eradication. CONCLUSIONS: A simulation-based method for calculating TSE-curves was developed, which provides more accurate predictions of tumor eradication than earlier, analytically derived, TSE-curves. The tool we present can potentially be used for radiosensitizer selection before proceeding to subsequent phases of the drug discovery and development process.


Assuntos
Antineoplásicos , Neoplasias , Radiossensibilizantes , Humanos , Animais , Camundongos , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Antineoplásicos/uso terapêutico , Terapia Combinada
4.
Front Nutr ; 10: 1304540, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38357465

RESUMO

Motivation: In the field of precision nutrition, predicting metabolic response to diet and identifying groups of differential responders are two highly desirable steps toward developing tailored dietary strategies. However, data analysis tools are currently lacking, especially for complex settings such as crossover studies with repeated measures.Current methods of analysis often rely on matrix or tensor decompositions, which are well suited for identifying differential responders but lacking in predictive power, or on dynamical systems modeling, which may be used for prediction but typically requires detailed mechanistic knowledge of the system under study. To remedy these shortcomings, we explored dynamic mode decomposition (DMD), which is a recent, data-driven method for deriving low-rank linear dynamical systems from high dimensional data.Combining the two recent developments "parametric DMD" (pDMD) and "DMD with control" (DMDc) enabled us to (i) integrate multiple dietary challenges, (ii) predict the dynamic response in all measured metabolites to new diets from only the metabolite baseline and dietary input, and (iii) identify inter-individual metabolic differences, i.e., metabotypes. To our knowledge, this is the first time DMD has been applied to analyze time-resolved metabolomics data. Results: We demonstrate the potential of pDMDc in a crossover study setting. We could predict the metabolite response to unseen dietary exposures on both measured (R2 = 0.40) and simulated data of increasing size (Rmax2= 0.65), as well as recover clusters of dynamic metabolite responses. We conclude that this method has potential for applications in personalized nutrition and could be useful in guiding metabolite response to target levels. Availability and implementation: The measured data analyzed in this study can be provided upon reasonable request. The simulated data along with a MATLAB implementation of pDMDc is available at https://github.com/FraunhoferChalmersCentre/pDMDc.

5.
Cancer Chemother Pharmacol ; 90(3): 239-250, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35922568

RESUMO

PURPOSE: Tumor growth inhibition (TGI) models are regularly used to quantify the PK-PD relationship between drug concentration and in vivo efficacy in oncology. These models are typically calibrated with data from xenograft mice and before being used for clinical predictions, translational methods have to be applied. Currently, such methods are commonly based on replacing model components or scaling of model parameters. However, difficulties remain in how to accurately account for inter-species differences. Therefore, more research must be done before xenograft data can fully be utilized to predict clinical response. METHOD: To contribute to this research, we have calibrated TGI models to xenograft data for three drug combinations using the nonlinear mixed effects framework. The models were translated by replacing mice exposure with human exposure and used to make predictions of clinical response. Furthermore, in search of a better way of translating these models, we estimated an optimal way of scaling model parameters given the available clinical data. RESULTS: The predictions were compared with clinical data and we found that clinical efficacy was overestimated. The estimated optimal scaling factors were similar to a standard allometric scaling exponent of - 0.25. CONCLUSIONS: We believe that given more data, our methodology could contribute to increasing the translational capabilities of TGI models. More specifically, an appropriate translational method could be developed for drugs with the same mechanism of action, which would allow for all preclinical data to be leveraged for new drugs of the same class. This would ensure that fewer clinically inefficacious drugs are tested in clinical trials.


Assuntos
Neoplasias , Animais , Xenoenxertos , Humanos , Camundongos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Critérios de Avaliação de Resposta em Tumores Sólidos , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Eur J Pharm Sci ; 176: 106256, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35820630

RESUMO

In this work we evaluate the study design of LPS challenge experiments used for quantification of drug induced inhibition of TNFα response and provide general guidelines of how to improve the study design. Analysis of model simulated data, using a recently published TNFα turnover model, as well as the optimal design tool PopED have been used to find the optimal values of three key study design variables - time delay between drug and LPS administration, LPS dose, and sampling time points - that in turn could make the resulting TNFα response data more informative. Our findings suggest that the current rule of thumb for choosing the time delay should be reconsidered, and that the placement of the measurements after maximal TNFα response are crucial for the quality of the experiment. Furthermore, a literature study summarizing a wide range of published LPS challenge studies is provided, giving a broader perspective of how LPS challenge studies are usually conducted both in a preclinical and clinical setting.


Assuntos
Lipopolissacarídeos , Fator de Necrose Tumoral alfa , Lipopolissacarídeos/farmacologia , Projetos de Pesquisa
7.
J Pharmacol Toxicol Methods ; 115: 107171, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35398273

RESUMO

Cardiovascular (CV) effects represent a major safety issue during drug development. Typically, this risk is mitigated by preclinical in vivo CV studies, based on which measured CV readouts are analyzed independently. Here, we apply a regression approach to simultaneously integrate CV readouts, i.e., heart rate (HR), mean arterial pressure (MAP) and QT from five dog telemetry studies. These CV studies comprise data on verapamil, captopril, dofetilide, pimobendan, and formoterol, and are combined with the respective dog pharmacokinetic (PK) profiles. A published PK/CV model structure for rats is extended by a semi-mechanistic parameterization of the interaction between HR and QT specific to dogs. This semi-mechanistic modelling approach allows differentiation between compound-independent system-specific parameters (e.g., HR baseline) and compound-specific parameters (e.g., EC50). Compared to previous results in rodents, estimated parameters for dogs indicate stronger dependency of stroke volume on HR, slower HR response, faster QT response and steeper concentration-response relationships. In addition, we illustrate how to practically apply the PK/CV model to derive concentration-response relationships for CV readouts. This approach allows a more detailed quantitative evaluation based on the maximum effect on CV effects (Emax), the EC50, and the steepness of this relation (Hill coefficient) especially for HR-independent effects on QT interval duration (QTc) while taking the systemic feedback into account. This approach also allows to derive plasma concentrations associated with relevant CV effects ("threshold concentration"; CTHRESH). The presented modelling analysis highlights the potential of an integrative evaluation of CV data and provides a framework for obtaining quantitative insights from safety pharmacology evaluations.


Assuntos
Sistema Cardiovascular , Síndrome do QT Longo , Animais , Cães , Desenvolvimento de Medicamentos , Eletrocardiografia , Frequência Cardíaca , Síndrome do QT Longo/induzido quimicamente , Ratos , Telemetria/métodos , Verapamil/farmacologia
8.
J Pharmacokinet Pharmacodyn ; 49(2): 167-178, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34623558

RESUMO

A central question in drug discovery is how to select drug candidates from a large number of available compounds. This analysis presents a model-based approach for comparing and ranking combinations of radiation and radiosensitizers. The approach is quantitative and based on the previously-derived Tumor Static Exposure (TSE) concept. Combinations of radiation and radiosensitizers are evaluated based on their ability to induce tumor regression relative to toxicity and other potential costs. The approach is presented in the form of a case study where the objective is to find the most promising candidate out of three radiosensitizing agents. Data from a xenograft study is described using a nonlinear mixed-effects modeling approach and a previously-published tumor model for radiation and radiosensitizing agents. First, the most promising candidate is chosen under the assumption that all compounds are equally toxic. The impact of toxicity in compound selection is then illustrated by assuming that one compound is more toxic than the others, leading to a different choice of candidate.


Assuntos
Neoplasias , Radiossensibilizantes , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/radioterapia , Radiossensibilizantes/farmacologia , Radiossensibilizantes/uso terapêutico
9.
CPT Pharmacometrics Syst Pharmacol ; 11(2): 212-224, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34797036

RESUMO

Home-based measures of lung function, inflammation, symptoms, and medication use are frequently collected in respiratory clinical trials. However, new statistical approaches are needed to make better use of the information contained in these data-rich variables. In this work, we use data from two phase III asthma clinical trials demonstrating the benefit of benralizumab treatment to develop a novel longitudinal mixed effects model of peak expiratory flow (PEF), a lung function measure easily captured at home using a hand-held device. The model is based on an extension of the mixed effects modeling framework to incorporate stochastic differential equations and allows for quantification of several statistical properties of a patient's PEF data: the longitudinal trend, long-term fluctuations, and day-to-day variability. These properties are compared between treatment groups and related to a patient's exacerbation risk using a repeated time-to-event model. The mixed effects model adequately described the observed data from the two clinical trials, and model parameters were accurately estimated. Benralizumab treatment was shown to improve a patient's average PEF level and reduce long-term fluctuations. Both of these effects were shown to be associated with a lower exacerbation risk. The day-to-day variability was neither significantly affected by treatment nor associated with exacerbation risk. Our work shows the potential of a stochastic model-based analysis of home-based lung function measures to support better estimation and understanding of treatment effects and disease stability. The proposed analysis can serve as a complement to descriptive statistics of home-based measures in the reporting of respiratory clinical trials.


Assuntos
Asma , Asma/tratamento farmacológico , Humanos
10.
Healthcare (Basel) ; 9(8)2021 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-34442098

RESUMO

The development and implementation of artificial intelligence (AI) applications in health care contexts is a concurrent research and management question. Especially for hospitals, the expectations regarding improved efficiency and effectiveness by the introduction of novel AI applications are huge. However, experiences with real-life AI use cases are still scarce. As a first step towards structuring and comparing such experiences, this paper is presenting a comparative approach from nine European hospitals and eleven different use cases with possible application areas and benefits of hospital AI technologies. This is structured as a current review and opinion article from a diverse range of researchers and health care professionals. This contributes to important improvement options also for pandemic crises challenges, e.g., the current COVID-19 situation. The expected advantages as well as challenges regarding data protection, privacy, or human acceptance are reported. Altogether, the diversity of application cases is a core characteristic of AI applications in hospitals, and this requires a specific approach for successful implementation in the health care sector. This can include specialized solutions for hospitals regarding human-computer interaction, data management, and communication in AI implementation projects.

11.
Eur J Pharm Sci ; 165: 105937, 2021 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-34260892

RESUMO

This study presents a non-linear mixed effects model describing tumour necrosis factor alpha (TNFα) release after lipopolysaccharide (LPS) provocations in absence or presence of anti-inflammatory test compounds. Inter-occasion variability and the pharmacokinetics of two test compounds have been added to this second-generation model, and the goal is to produce a framework of how to model TNFα response in LPS challenge studies in vivo and demonstrate its general applicability regardless of occasion or type of test compound. Model improvements based on experimental data were successfully implemented and provided a robust model for TNFα response after LPS provocation, as well as reliable estimates of the median pharmacodynamic parameters. The two test compounds, Test Compound A and roflumilast, showed 81.1% and 74.9% partial reduction of TNFα response, respectively, and the potency of Test Compound A was estimated to 0.166 µmol/L. Comparing this study with previously published work reveals that our model leads to biologically reasonable output, handles complex data pooled from different studies, and highlights the importance of accurately distinguishing the stimulatory effect of LPS from the inhibitory effect of the test compound.


Assuntos
Lipopolissacarídeos , Fator de Necrose Tumoral alfa , Anti-Inflamatórios/farmacologia , Humanos
12.
Math Biosci ; 338: 108595, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33831415

RESUMO

Proliferation of an in vitro population of cancer cells is described by a linear cell cycle model with n states, subject to provocation with m chemotherapeutic compounds. Minimization of a linear combination of constant drug exposures is considered, with stability of the system used as a constraint to ensure a stable or shrinking cell population. The main result concerns the identification of redundant compounds, and an explicit solution formula for the case where all exposures are nonzero. The orthogonal case, where each drug acts on a single and different stage of the cell cycle, leads to a version of the classic inequality between the arithmetic and geometric means. Moreover, it is shown how the general case can be solved by converting it to the orthogonal case using a linear invertible transformation. The results are illustrated with two examples corresponding to combination treatment with two and three compounds, respectively.


Assuntos
Antineoplásicos , Ciclo Celular , Modelos Biológicos , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Tratamento Farmacológico , Quimioterapia Combinada , Humanos
13.
J Pharmacol Exp Ther ; 377(2): 218-231, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33648939

RESUMO

Cardiovascular adverse effects in drug development are a major source of compound attrition. Characterization of blood pressure (BP), heart rate (HR), stroke volume (SV), and QT-interval prolongation are therefore necessary in early discovery. It is, however, common practice to analyze these effects independently of each other. High-resolution time courses are collected via telemetric techniques, but only low-resolution data are analyzed and reported. This ignores codependencies among responses (HR, BP, SV, and QT-interval) and separation of system (turnover properties) and drug-specific properties (potencies, efficacies). An analysis of drug exposure-time and high-resolution response-time data of HR and mean arterial blood pressure was performed after acute oral dosing of ivabradine, sildenafil, dofetilide, and pimobendan in Han-Wistar rats. All data were modeled jointly, including different compounds and exposure and response time courses, using a nonlinear mixed-effects approach. Estimated fractional turnover rates [h-1, relative standard error (%RSE) within parentheses] were 9.45 (15), 30.7 (7.8), 3.8 (13), and 0.115 (1.7) for QT, HR, total peripheral resistance, and SV, respectively. Potencies (nM, %RSE within parentheses) were IC 50 = 475 (11), IC 50 = 4.01 (5.4), EC 50 = 50.6 (93), and IC 50 = 47.8 (16), and efficacies (%RSE within parentheses) were I max = 0.944 (1.7), Imax = 1.00 (1.3), E max = 0.195 (9.9), and Imax = 0.745 (4.6) for ivabradine, sildenafil, dofetilide, and pimobendan. Hill parameters were estimated with good precision and below unity, indicating a shallow concentration-response relationship. An equilibrium concentration-biomarker response relationship was predicted and displayed graphically. This analysis demonstrates the utility of a model-based approach integrating data from different studies and compounds for refined preclinical safety margin assessment. SIGNIFICANCE STATEMENT: A model-based approach was proposed utilizing biomarker data on heart rate, blood pressure, and QT-interval. A pharmacodynamic model was developed to improve assessment of high-resolution telemetric cardiovascular safety data driven by different drugs (ivabradine, sildenafil, dofetilide, and pimobondan), wherein system- (turnover rates) and drug-specific parameters (e.g., potencies and efficacies) were sought. The model-predicted equilibrium concentration-biomarker response relationships and was used for safety assessment (predictions of 20% effective concentration, for example) of heart rate, blood pressure, and QT-interval.


Assuntos
Biomarcadores Farmacológicos/sangue , Pressão Sanguínea , Fármacos Cardiovasculares/toxicidade , Frequência Cardíaca , Animais , Cardiotoxicidade/sangue , Cardiotoxicidade/etiologia , Cardiotoxicidade/fisiopatologia , Fármacos Cardiovasculares/administração & dosagem , Fármacos Cardiovasculares/farmacocinética , Ivabradina/administração & dosagem , Ivabradina/farmacocinética , Ivabradina/toxicidade , Masculino , Fenetilaminas/administração & dosagem , Fenetilaminas/farmacocinética , Fenetilaminas/toxicidade , Piridazinas/administração & dosagem , Piridazinas/farmacocinética , Piridazinas/toxicidade , Ratos , Ratos Wistar , Citrato de Sildenafila/administração & dosagem , Citrato de Sildenafila/farmacocinética , Citrato de Sildenafila/toxicidade , Sulfonamidas/administração & dosagem , Sulfonamidas/farmacocinética , Sulfonamidas/toxicidade
14.
Cancer Chemother Pharmacol ; 83(6): 1159-1173, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30976845

RESUMO

PURPOSE: Radiation therapy, whether given alone or in combination with chemical agents, is one of the cornerstones of oncology. We develop a quantitative model that describes tumor growth during and after treatment with radiation and radiosensitizing agents. The model also describes long-term treatment effects including tumor regrowth and eradication. METHODS: We challenge the model with data from a xenograft study using a clinically relevant administration schedule and use a mixed-effects approach for model-fitting. We use the calibrated model to predict exposure combinations that result in tumor eradication using Tumor Static Exposure (TSE). RESULTS: The model is able to adequately describe data from all treatment groups, with the parameter estimates taking biologically reasonable values. Using TSE, we predict the total radiation dose necessary for tumor eradication to be 110 Gy, which is reduced to 80 or 30 Gy with co-administration of 25 or 100 mg kg-1 of a radiosensitizer. TSE is also explored via a heat map of different growth and shrinkage rates. Finally, we discuss the translational potential of the model and TSE concept to humans. CONCLUSIONS: The new model is capable of describing different tumor dynamics including tumor eradication and tumor regrowth with different rates, and can be calibrated using data from standard xenograft experiments. TSE and related concepts can be used to predict tumor shrinkage and eradication, and have the potential to guide new experiments and support translations from animals to humans.


Assuntos
Modelos Biológicos , Neoplasias/radioterapia , Radiossensibilizantes/administração & dosagem , Animais , Relação Dose-Resposta a Droga , Feminino , Humanos , Camundongos , Camundongos Nus , Dosagem Radioterapêutica , Especificidade da Espécie , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Pharmacokinet Pharmacodyn ; 46(3): 223-240, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30778719

RESUMO

A mechanism-based biomarker model of TNFα-response, including different external provocations of LPS challenge and test compound intervention, was developed. The model contained system properties (such as kt, kout), challenge characteristics (such as ks, kLPS, Km, LPS, Smax, SC50) and test-compound-related parameters (Imax, IC50). The exposure to test compound was modelled by means of first-order input and Michaelis-Menten type of nonlinear elimination. Test compound potency was estimated to 20 nM with a 70% partial reduction in TNFα-response at the highest dose of 30 mg·kg-1. Future selection of drug candidates may focus the estimation on potency and efficacy by applying the selected structure consisting of TNFα system and LPS challenge characteristics. A related aim was to demonstrate how an exploratory (graphical) analysis may guide us to a tentative model structure, which enables us to better understand target biology. The analysis demonstrated how to tackle a biomarker with a baseline below the limit of detection. Repeated LPS-challenges may also reveal how the rate and extent of replenishment of TNFα pools occur. Lack of LPS exposure-time courses was solved by including a biophase model, with the underlying assumption that TNFα-response time courses, as such, contain kinetic information. A transduction type of model with non-linear stimulation of TNFα release was finally selected. Typical features of a challenge experiment were shown by means of model simulations. Experimental shortcomings of present and published designs are identified and discussed. The final model coupled to suggested guidance rules may serve as a general basis for the collection and analysis of pharmacological challenge data of future studies.


Assuntos
Fator de Necrose Tumoral alfa/metabolismo , Animais , Biomarcadores/metabolismo , Lipopolissacarídeos/farmacologia , Masculino , Modelos Biológicos , Ratos , Ratos Sprague-Dawley
16.
J Pharmacokinet Pharmacodyn ; 46(1): 75-87, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30673914

RESUMO

Cortisol is a steroid hormone relevant to immune function in horses and other species and shows a circadian rhythm. The glucocorticoid dexamethasone suppresses cortisol in horses. Pituitary pars intermedia dysfunction (PPID) is a disease in which the cortisol suppression mechanism through dexamethasone is challenged. Overnight dexamethasone suppression test (DST) protocols are used to test the functioning of this mechanism and to establish a diagnosis for PPID. However, existing DST protocols have been recognized to perform poorly in previous experimental studies, often indicating presence of PPID in healthy horses. This study uses a pharmacokinetic/pharmacodynamic (PK/PD) modelling approach to analyse the oscillatory cortisol response and its interaction with dexamethasone. Two existing DST protocols were then scrutinized using model simulations with particular focus on their ability to avoid false positive outcomes. Using a Bayesian population approach allowed for quantification of uncertainty and enabled predictions for a broader population of horses than the underlying sample. Dose selection and sampling time point were both determined to have large influence on the number of false positives. Advice on pitfalls in test protocols and directions for possible improvement of DST protocols were given. The presented methodology is also easily extended to other clinical test protocols.


Assuntos
Dexametasona/farmacologia , Hidrocortisona/metabolismo , Animais , Teorema de Bayes , Ritmo Circadiano/efeitos dos fármacos , Glucocorticoides/farmacologia , Cavalos , Doenças da Hipófise/tratamento farmacológico , Doenças da Hipófise/metabolismo
17.
Eur J Pharm Sci ; 128: 250-269, 2019 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-30453011

RESUMO

This study presents an extensive dose-response-time (DRT) meta-analysis of the nicotinic acid-induced inhibition of free fatty acids and insulin release. The purpose was to quantify the implications of lacking exposure data when analysing complex pharmacodynamic systems. The DRT model successfully characterised various response behaviours-including time-delays, rebound, feedback mechanisms, and adaptation-on both the individual and the population level. Comparing the fitted DRT model to an exposure-driven reference analysis showed that bias and uncertainty were introduced in the parameter estimates. However, most estimates were within one standard error from the reference. In both approaches, a few parameters suffered from practical identifiability issues, likely due to large differences in half-lives of the different rate processes. Moreover, the optimal dosing strategies predicted by the DRT model differed slightly from those of the exposure-driven analysis, having a lower optimal steady-state reduction of free fatty acids exposure.


Assuntos
Ácidos Graxos não Esterificados/metabolismo , Insulina/metabolismo , Niacina/farmacologia , Animais , Relação Dose-Resposta a Droga , Modelos Biológicos , Niacina/administração & dosagem , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
18.
Comput Methods Programs Biomed ; 171: 141-152, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27181677

RESUMO

BACKGROUND AND OBJECTIVE: Structural identifiability is a concept that considers whether the structure of a model together with a set of input-output relations uniquely determines the model parameters. In the mathematical modelling of biological systems, structural identifiability is an important concept since biological interpretations are typically made from the parameter estimates. For a system defined by ordinary differential equations, several methods have been developed to analyse whether the model is structurally identifiable or otherwise. Another well-used modelling framework, which is particularly useful when the experimental data are sparsely sampled and the population variance is of interest, is mixed-effects modelling. However, established identifiability analysis techniques for ordinary differential equations are not directly applicable to such models. METHODS: In this paper, we present and apply three different methods that can be used to study structural identifiability in mixed-effects models. The first method, called the repeated measurement approach, is based on applying a set of previously established statistical theorems. The second method, called the augmented system approach, is based on augmenting the mixed-effects model to an extended state-space form. The third method, called the Laplace transform mixed-effects extension, is based on considering the moment invariants of the systems transfer function as functions of random variables. RESULTS: To illustrate, compare and contrast the application of the three methods, they are applied to a set of mixed-effects models. CONCLUSIONS: Three structural identifiability analysis methods applicable to mixed-effects models have been presented in this paper. As method development of structural identifiability techniques for mixed-effects models has been given very little attention, despite mixed-effects models being widely used, the methods presented in this paper provides a way of handling structural identifiability in mixed-effects models previously not possible.


Assuntos
Bioestatística/métodos , Modelos Estatísticos , Algoritmos , Simulação por Computador
19.
Pharmacol Rev ; 71(1): 89-122, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30587536

RESUMO

The most common approach to in vivo pharmacokinetic and pharmacodynamic analyses involves sequential analysis of the plasma concentration- and response-time data, such that the plasma kinetic model provides an independent function, driving the dynamics. However, in situations when plasma sampling may jeopardize the effect measurements or is scarce, nonexistent, or unlinked to the effect (e.g., in intensive care units, pediatric or frail elderly populations, or drug discovery), focusing on the response-time course alone may be an adequate alternative for pharmacodynamic analyses. Response-time data inherently contain useful information about the turnover characteristics of response (target turnover rate, half-life of response), as well as the drug's biophase kinetics (biophase availability, absorption half-life, and disposition half-life) pharmacodynamic properties (potency, efficacy). The use of pharmacodynamic time-response data circumvents the need for a direct assay method for the drug and has the additional advantage of being applicable to cases of local drug administration close to its intended targets in the immediate vicinity of target, or when target precedes systemic plasma concentrations. This review exemplifies the potential of biophase functions in pharmacodynamic analyses in both preclinical and clinical studies, with the purpose of characterizing response data and optimizing subsequent study protocols. This article illustrates crucial determinants to the success of modeling dose-response-time (DRT) data, such as the dose selection, repeated dosing, and different input rates and routes. Finally, a literature search was also performed to gauge how frequently this technique has been applied in preclinical and clinical studies. This review highlights situations in which DRT should be carefully scrutinized and discusses future perspectives of the field.


Assuntos
Desenvolvimento de Medicamentos/métodos , Modelos Biológicos , Preparações Farmacêuticas/administração & dosagem , Idoso , Animais , Criança , Ensaios Clínicos como Assunto/métodos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Unidades de Terapia Intensiva , Preparações Farmacêuticas/metabolismo , Fatores de Tempo
20.
AAPS J ; 20(5): 88, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30069613

RESUMO

Nonlinear mixed effects (NLME) modeling based on stochastic differential equations (SDEs) have evolved into a promising approach for analysis of PK/PD data. SDE-NLME models go beyond the realm of standard population modeling as they consider stochastic dynamics, thereby introducing a probabilistic perspective on the state variables. This article presents a summary of the main contributions to SDE-NLME models found in the literature. The aims of this work were to develop an exact gradient version of the first-order conditional estimation (FOCE) method for SDE-NLME models and to investigate whether it enabled faster estimation and better gradient precision/accuracy compared to the use of gradients approximated by finite differences. A simulation-estimation study was set up whereby finite difference approximations of the gradients of each level were interchanged with the exact gradients. Following previous work, the uncertainty of the state variables was accounted for using the extended Kalman filter (EKF). The exact gradient FOCE method was implemented in Mathematica 11 and evaluated on SDE versions of three common PK/PD models. When finite difference gradients were replaced by exact gradients at both FOCE levels, relative runtimes improved between 6- and 32-fold, depending on model complexity. Additionally, gradient precision/accuracy was significantly better in the exact gradient case. We conclude that parameter estimation using FOCE with exact gradients can successfully be applied to SDE-NLME models.


Assuntos
Variação Biológica da População , Modelos Biológicos , Dinâmica não Linear , Farmacocinética , Processos Estocásticos , Algoritmos , Simulação por Computador , Humanos , Incerteza
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...