Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS Comput Biol ; 19(4): e1011000, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37053266

RESUMO

BACKGROUND: Antibiotic treatments are often associated with a late slowdown in bacterial killing. This separates the killing of bacteria into at least two distinct phases: a quick phase followed by a slower phase, the latter of which is linked to treatment success. Current mechanistic explanations for the in vitro slowdown are either antibiotic persistence or heteroresistance. Persistence is defined as the switching back and forth between susceptible and non-susceptible states, while heteroresistance is defined as the coexistence of bacteria with heterogeneous susceptibilities. Both are also thought to cause a slowdown in the decline of bacterial populations in patients and therefore complicate and prolong antibiotic treatments. Reduced bacterial death rates over time are also observed within tuberculosis patients, yet the mechanistic reasons for this are unknown and therefore the strategies to mitigate them are also unknown. METHODS AND FINDINGS: We analyse a dose ranging trial for rifampicin in tuberculosis patients and show that there is a slowdown in the decline of bacteria. We show that the late phase of bacterial killing depends more on the peak drug concentrations than the total drug exposure. We compare these to pharmacokinetic-pharmacodynamic models of rifampicin heteroresistance and persistence. We find that the observation on the slow phase's dependence on pharmacokinetic measures, specifically peak concentrations are only compatible with models of heteroresistance and incompatible with models of persistence. The quantitative agreement between heteroresistance models and observations is very good ([Formula: see text]). To corroborate the importance of the slowdown, we validate our results by estimating the time to sputum culture conversion and compare the results to a different dose ranging trial. CONCLUSIONS: Our findings indicate that higher doses, specifically higher peak concentrations may be used to optimize rifampicin treatments by accelerating bacterial killing in the slow phase. It adds to the growing body of literature supporting higher rifampicin doses for shortening tuberculosis treatments.


Assuntos
Mycobacterium tuberculosis , Tuberculose , Humanos , Rifampina/uso terapêutico , Rifampina/farmacocinética , Tuberculose/tratamento farmacológico , Antibacterianos/farmacologia
2.
BMC Bioinformatics ; 23(1): 22, 2022 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-34991453

RESUMO

BACKGROUND: As antibiotic resistance creates a significant global health threat, we need not only to accelerate the development of novel antibiotics but also to develop better treatment strategies using existing drugs to improve their efficacy and prevent the selection of further resistance. We require new tools to rationally design dosing regimens from data collected in early phases of antibiotic and dosing development. Mathematical models such as mechanistic pharmacodynamic drug-target binding explain mechanistic details of how the given drug concentration affects its targeted bacteria. However, there are no available tools in the literature that allow non-quantitative scientists to develop computational models to simulate antibiotic-target binding and its effects on bacteria. RESULTS: In this work, we have devised an extension of a mechanistic binding-kinetic model to incorporate clinical drug concentration data. Based on the extended model, we develop a novel and interactive web-based tool that allows non-quantitative scientists to create and visualize their own computational models of bacterial antibiotic target-binding based on their considered drugs and bacteria. We also demonstrate how Rifampicin affects bacterial populations of Tuberculosis bacteria using our vCOMBAT tool. CONCLUSIONS: The vCOMBAT online tool is publicly available at https://combat-bacteria.org/ .


Assuntos
Antibacterianos , Farmacorresistência Bacteriana , Antibacterianos/farmacologia , Bactérias/genética , Simulação por Computador , Modelos Biológicos
3.
Eur Respir Rev ; 30(160)2021 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-34039674

RESUMO

Standardised management of tuberculosis may soon be replaced by individualised, precision medicine-guided therapies informed with knowledge provided by the field of systems biology. Systems biology is a rapidly expanding field of computational and mathematical analysis and modelling of complex biological systems that can provide insights into mechanisms underlying tuberculosis, identify novel biomarkers, and help to optimise prevention, diagnosis and treatment of disease. These advances are critically important in the context of the evolving epidemic of drug-resistant tuberculosis. Here, we review the available evidence on the role of systems biology approaches - human and mycobacterial genomics and transcriptomics, proteomics, lipidomics/metabolomics, immunophenotyping, systems pharmacology and gut microbiomes - in the management of tuberculosis including prediction of risk for disease progression, severity of mycobacterial virulence and drug resistance, adverse events, comorbidities, response to therapy and treatment outcomes. Application of the Grading of Recommendations, Assessment, Development and Evaluation (GRADE) approach demonstrated that at present most of the studies provide "very low" certainty of evidence for answering clinically relevant questions. Further studies in large prospective cohorts of patients, including randomised clinical trials, are necessary to assess the applicability of the findings in tuberculosis prevention and more efficient clinical management of patients.


Assuntos
Biologia de Sistemas , Tuberculose , Genômica , Humanos , Metabolômica , Estudos Prospectivos , Tuberculose/diagnóstico , Tuberculose/tratamento farmacológico
5.
ERJ Open Res ; 6(4)2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33263043

RESUMO

Adherence to treatment for tuberculosis (TB) has been a concern for many decades, resulting in the World Health Organization's recommendation of the direct observation of treatment in the 1990s. Recent advances in digital adherence technologies (DATs) have renewed discussion on how to best address nonadherence, as well as offering important information on dose-by-dose adherence patterns and their variability between countries and settings. Previous studies have largely focussed on percentage thresholds to delineate sufficient adherence, but this is misleading and limited, given the complex and dynamic nature of adherence over the treatment course. Instead, we apply a standardised taxonomy - as adopted by the international adherence community - to dose-by-dose medication-taking data, which divides missed doses into 1) late/noninitiation (starting treatment later than expected/not starting), 2) discontinuation (ending treatment early), and 3) suboptimal implementation (intermittent missed doses). Using this taxonomy, we can consider the implications of different forms of nonadherence for intervention and regimen design. For example, can treatment regimens be adapted to increase the "forgiveness" of common patterns of suboptimal implementation to protect against treatment failure and the development of drug resistance? Is it reasonable to treat all missed doses of treatment as equally problematic and equally common when deploying DATs? Can DAT data be used to indicate the patients that need enhanced levels of support during their treatment course? Critically, we pinpoint key areas where knowledge regarding treatment adherence is sparse and impeding scientific progress.

6.
Cell Mol Life Sci ; 77(3): 381-394, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31768605

RESUMO

Optimizing drug therapies for any disease requires a solid understanding of pharmacokinetics (the drug concentration at a given time point in different body compartments) and pharmacodynamics (the effect a drug has at a given concentration). Mathematical models are frequently used to infer drug concentrations over time based on infrequent sampling and/or in inaccessible body compartments. Models are also used to translate drug action from in vitro to in vivo conditions or from animal models to human patients. Recently, mathematical models that incorporate drug-target binding and subsequent downstream responses have been shown to advance our understanding and increase predictive power of drug efficacy predictions. We here discuss current approaches of modeling drug binding kinetics that aim at improving model-based drug development in the future. This in turn might aid in reducing the large number of failed clinical trials.


Assuntos
Desenho de Fármacos , Preparações Farmacêuticas/metabolismo , Animais , Sistemas de Liberação de Medicamentos/métodos , Humanos , Cinética , Modelos Teóricos
7.
Int J Mol Sci ; 20(16)2019 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-31443146

RESUMO

Bacterial heteroresistance (i.e., the co-existence of several subpopulations with different antibiotic susceptibilities) can delay the clearance of bacteria even with long antibiotic exposure. Some proposed mechanisms have been successfully described with mathematical models of drug-target binding where the mechanism's downstream of drug-target binding are not explicitly modeled and subsumed in an empirical function, connecting target occupancy to antibiotic action. However, with current approaches it is difficult to model mechanisms that involve multi-step reactions that lead to bacterial killing. Here, we have a dual aim: first, to establish pharmacodynamic models that include multi-step reaction pathways, and second, to model heteroresistance and investigate which molecular heterogeneities can lead to delayed bacterial killing. We show that simulations based on Gillespie algorithms, which have been employed to model reaction kinetics for decades, can be useful tools to model antibiotic action via multi-step reactions. We highlight the strengths and weaknesses of current models and Gillespie simulations. Finally, we show that in our models, slight normally distributed variances in the rates of any event leading to bacterial death can (depending on parameter choices) lead to delayed bacterial killing (i.e., heteroresistance). This means that a slowly declining residual bacterial population due to heteroresistance is most likely the default scenario and should be taken into account when planning treatment length.


Assuntos
Antibacterianos/farmacologia , Algoritmos , Farmacorresistência Bacteriana , Cinética , Testes de Sensibilidade Microbiana
8.
PLoS One ; 13(10): e0205099, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30286168

RESUMO

In the human retina, rod and cone cells detect incoming light with a molecule called rhodopsin. After rhodopsin molecules are activated (by photon impact), these molecules activate the rest of the signalling process for a brief period of time until they are deactivated by a multistage process. First, active rhodopsin is phosphorylated multiple times. Following this, they are further inhibited by the binding of molecules called arrestins. Finally, they decay into opsins. The time required for each of these stages becomes progressively longer, and each stage further reduces the activity of rhodopsin. However, while this deactivation process itself is well researched, the roles of the above stages in signal (and image) processing are poorly understood. In this paper, we will show that the activity of rhodopsin molecules during the deactivation process can be described as the fractional integration of an incoming signal. Furthermore, we show how this affects an image; specifically, the effect of fractional integration in video and signal processing and how it reduces noise and the improves adaptability under different lighting conditions. Our experimental results provide a better understanding of vertebrate and human vision, and why the rods and cones of the retina differ from the light detectors in cameras.


Assuntos
Adaptação Fisiológica/fisiologia , Células Fotorreceptoras Retinianas Cones/metabolismo , Rodopsina/metabolismo , Visão Ocular/fisiologia , Algoritmos , Humanos , Processamento de Imagem Assistida por Computador , Modelos Biológicos , Fosforilação
9.
Pathog Dis ; 76(6)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-30107522

RESUMO

Treatment of infectious diseases is often long and requires patients to take drugs even after they have seemingly recovered. This is because of a phenomenon called persistence, which allows small fractions of the bacterial population to survive treatment despite being genetically susceptible. The surviving subpopulation is often below detection limit and therefore is empirically inaccessible but can cause treatment failure when treatment is terminated prematurely. Mathematical models could aid in predicting bacterial survival and thereby determine sufficient treatment length. However, the mechanisms of persistence are hotly debated, necessitating the development of multiple mechanistic models. Here we develop a generalized mathematical framework that can accommodate various persistence mechanisms from measurable heterogeneities in pathogen populations. It allows the estimation of the relative increase in treatment length necessary to eradicate persisters compared to the majority population. To simplify and generalize, we separate the model into two parts: the distribution of the molecular mechanism of persistence in the bacterial population (e.g. number of efflux pumps or target molecules, growth rates) and the elimination rate of single bacteria as a function of that phenotype. Thereby, we obtain an estimate of the required treatment length for each phenotypic subpopulation depending on its size and susceptibility.


Assuntos
Antibacterianos/administração & dosagem , Infecções Bacterianas/tratamento farmacológico , Tratamento Farmacológico/métodos , Viabilidade Microbiana/efeitos dos fármacos , Tempo , Humanos , Modelos Teóricos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA