Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
NPJ Syst Biol Appl ; 10(1): 51, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38750040

RESUMO

In vertical inhibition treatment strategies, multiple components of an intracellular pathway are simultaneously inhibited. Vertical inhibition of the BRAFV600E-MEK-ERK signalling pathway is a standard of care for treating BRAFV600E-mutated melanoma where two targeted cancer drugs, a BRAFV600E-inhibitor, and a MEK inhibitor, are administered in combination. Targeted therapies have been linked to early onsets of drug resistance, and thus treatment strategies of higher complexities and lower doses have been proposed as alternatives to current clinical strategies. However, finding optimal complex, low-dose treatment strategies is a challenge, as it is possible to design more treatment strategies than are feasibly testable in experimental settings. To quantitatively address this challenge, we develop a mathematical model of BRAFV600E-MEK-ERK signalling dynamics in response to combinations of the BRAFV600E-inhibitor dabrafenib (DBF), the MEK inhibitor trametinib (TMT), and the ERK-inhibitor SCH772984 (SCH). From a model of the BRAFV600E-MEK-ERK pathway, and a set of molecular-level drug-protein interactions, we extract a system of chemical reactions that is parameterised by in vitro data and converted to a system of ordinary differential equations (ODEs) using the law of mass action. The ODEs are solved numerically to produce simulations of how pathway-component concentrations change over time in response to different treatment strategies, i.e., inhibitor combinations and doses. The model can thus be used to limit the search space for effective treatment strategies that target the BRAFV600E-MEK-ERK pathway and warrant further experimental investigation. The results demonstrate that DBF and DBF-TMT-SCH therapies show marked sensitivity to BRAFV600E concentrations in silico, whilst TMT and SCH monotherapies do not.


Assuntos
Imidazóis , Sistema de Sinalização das MAP Quinases , Melanoma , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas B-raf , Piridonas , Pirimidinonas , Proteínas Proto-Oncogênicas B-raf/genética , Humanos , Piridonas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Melanoma/tratamento farmacológico , Melanoma/genética , Imidazóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Pirimidinonas/farmacologia , Oximas/farmacologia , Simulação por Computador , Modelos Biológicos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Mutação , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética
2.
J Math Biol ; 86(5): 68, 2023 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-37017776

RESUMO

Theoretical and applied cancer studies that use individual-based models (IBMs) have been limited by the lack of a mathematical formulation that enables rigorous analysis of these models. However, spatial cumulant models (SCMs), which have arisen from theoretical ecology, describe population dynamics generated by a specific family of IBMs, namely spatio-temporal point processes (STPPs). SCMs are spatially resolved population models formulated by a system of differential equations that approximate the dynamics of two STPP-generated summary statistics: first-order spatial cumulants (densities), and second-order spatial cumulants (spatial covariances). We exemplify how SCMs can be used in mathematical oncology by modelling theoretical cancer cell populations comprising interacting growth factor-producing and non-producing cells. To formulate model equations, we use computational tools that enable the generation of STPPs, SCMs and mean-field population models (MFPMs) from user-defined model descriptions (Cornell et al. Nat Commun 10:4716, 2019). To calculate and compare STPP, SCM and MFPM-generated summary statistics, we develop an application-agnostic computational pipeline. Our results demonstrate that SCMs can capture STPP-generated population density dynamics, even when MFPMs fail to do so. From both MFPM and SCM equations, we derive treatment-induced death rates required to achieve non-growing cell populations. When testing these treatment strategies in STPP-generated cell populations, our results demonstrate that SCM-informed strategies outperform MFPM-informed strategies in terms of inhibiting population growths. We thus demonstrate that SCMs provide a new framework in which to study cell-cell interactions, and can be used to describe and perturb STPP-generated cell population dynamics. We, therefore, argue that SCMs can be used to increase IBMs' applicability in cancer research.


Assuntos
Ecologia , Neoplasias , Humanos , Dinâmica Populacional , Crescimento Demográfico , Modelos Biológicos
3.
Sci Rep ; 13(1): 1211, 2023 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-36681762

RESUMO

A key feature distinguishing 3D bioprinting from other 3D cell culture techniques is its precise control over created structures. This property allows for the high-resolution fabrication of biomimetic structures with controlled structural and mechanical properties such as porosity, permeability, and stiffness. However, analyzing post-printing cellular dynamics and optimizing their functions within the 3D fabricated environment is only possible through trial and error and replicating several experiments. This issue motivated the development of a cellular automata model for the first time to simulate post-printing cell behaviour within the 3D bioprinted construct. To improve our model, we bioprinted a 3D construct using MDA-MB-231 cell-laden hydrogel and evaluated cellular functions, including viability and proliferation in 11 days. The results showed that our model successfully simulated the 3D bioprinted structure and captured in-vitro observations. We demonstrated that in-silico model could predict and elucidate post-printing biological functions for different initial cell numbers in bioink and different bioink formulations with gelatine and alginate, without replicating several costly and time-consuming in-vitro measurements. We believe such a computational framework will substantially impact 3D bioprinting's future application. We hope this study inspires researchers to further realize how an in-silico model might be utilized to advance in-vitro 3D bioprinting research.


Assuntos
Bioimpressão , Neoplasias , Hidrogéis/química , Porosidade , Gelatina/química , Sobrevivência Celular , Impressão Tridimensional , Bioimpressão/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais/química
4.
Br J Cancer ; 125(11): 1552-1560, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34621046

RESUMO

BACKGROUND: Simultaneous inhibition of multiple components of the BRAF-MEK-ERK cascade (vertical inhibition) has become a standard of care for treating BRAF-mutant melanoma. However, the molecular mechanism of how vertical inhibition synergistically suppresses intracellular ERK activity, and consequently cell proliferation, are yet to be fully elucidated. METHODS: We develop a mechanistic mathematical model that describes how the mutant BRAF inhibitor, dabrafenib, and the MEK inhibitor, trametinib, affect BRAFV600E-MEK-ERK signalling. The model is based on a system of chemical reactions that describes cascade signalling dynamics. Using mass action kinetics, the chemical reactions are re-expressed as ordinary differential equations that are parameterised by in vitro data and solved numerically to obtain the temporal evolution of cascade component concentrations. RESULTS: The model provides a quantitative method to compute how dabrafenib and trametinib can be used in combination to synergistically inhibit ERK activity in BRAFV600E-mutant melanoma cells. The model elucidates molecular mechanisms of vertical inhibition of the BRAFV600E-MEK-ERK cascade and delineates how elevated BRAF concentrations generate drug resistance to dabrafenib and trametinib. The computational simulations further suggest that elevated ATP levels could be a factor in drug resistance to dabrafenib. CONCLUSIONS: The model can be used to systematically motivate which dabrafenib-trametinib dose combinations, for treating BRAFV600E-mutated melanoma, warrant experimental investigation.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Modelos Biológicos , Modelos Químicos , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/química , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Imidazóis/química , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/tratamento farmacológico , Melanoma/enzimologia , Melanoma/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/química , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Mutação , Oximas/química , Oximas/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/química , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Piridonas/química , Piridonas/farmacologia , Pirimidinonas/química , Pirimidinonas/farmacologia
5.
Bull Math Biol ; 83(10): 103, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34459993

RESUMO

We combine a systems pharmacology approach with an agent-based modelling approach to simulate LoVo cells subjected to AZD6738, an ATR (ataxia-telangiectasia-mutated and rad3-related kinase) inhibiting anti-cancer drug that can hinder tumour proliferation by targeting cellular DNA damage responses. The agent-based model used in this study is governed by a set of empirically observable rules. By adjusting only the rules when moving between monolayer and multi-cellular tumour spheroid simulations, whilst keeping the fundamental mathematical model and parameters intact, the agent-based model is first parameterised by monolayer in vitro data and is thereafter used to simulate treatment responses in in vitro tumour spheroids subjected to dynamic drug delivery. Spheroid simulations are subsequently compared to in vivo data from xenografts in mice. The spheroid simulations are able to capture the dynamics of in vivo tumour growth and regression for approximately 8 days post-tumour injection. Translating quantitative information between in vitro and in vivo research remains a scientifically and financially challenging step in preclinical drug development processes. However, well-developed in silico tools can be used to facilitate this in vitro to in vivo translation, and in this article, we exemplify how data-driven, agent-based models can be used to bridge the gap between in vitro and in vivo research. We further highlight how agent-based models, that are currently underutilised in pharmaceutical contexts, can be used in preclinical drug development.


Assuntos
Neoplasias , Preparações Farmacêuticas , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Linhagem Celular Tumoral , Dano ao DNA , Conceitos Matemáticos , Camundongos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Farmacologia em Rede , Ensaios Antitumorais Modelo de Xenoenxerto
6.
PLoS Comput Biol ; 16(8): e1008041, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32745136

RESUMO

Hypoxia-activated prodrugs (HAPs) present a conceptually elegant approach to not only overcome, but better yet, exploit intra-tumoural hypoxia. Despite being successful in vitro and in vivo, HAPs are yet to achieve successful results in clinical settings. It has been hypothesised that this lack of clinical success can, in part, be explained by the insufficiently stringent clinical screening selection of determining which tumours are suitable for HAP treatments. Taking a mathematical modelling approach, we investigate how tumour properties and HAP-radiation scheduling influence treatment outcomes in simulated tumours. The following key results are demonstrated in silico: (i) HAP and ionising radiation (IR) monotherapies may attack tumours in dissimilar, and complementary, ways. (ii) HAP-IR scheduling may impact treatment efficacy. (iii) HAPs may function as IR treatment intensifiers. (iv) The spatio-temporal intra-tumoural oxygen landscape may impact HAP efficacy. Our in silico framework is based on an on-lattice, hybrid, multiscale cellular automaton spanning three spatial dimensions. The mathematical model for tumour spheroid growth is parameterised by multicellular tumour spheroid (MCTS) data.


Assuntos
Antineoplásicos/farmacologia , Hipóxia Celular/fisiologia , Modelos Biológicos , Pró-Fármacos/farmacologia , Microambiente Tumoral/fisiologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Biologia Computacional , Simulação por Computador , Humanos , Radiação Ionizante , Radioterapia , Esferoides Celulares , Células Tumorais Cultivadas
7.
JCO Clin Cancer Inform ; 3: 1-11, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30742485

RESUMO

Cancers present with high variability across patients and tumors; thus, cancer care, in terms of disease prevention, detection, and control, can highly benefit from a personalized approach. For a comprehensive personalized oncology practice, this personalization should ideally consider data gathered from various information levels, which range from the macroscale population level down to the microscale tumor level, without omission of the central patient level. Appropriate data mined from each of these levels can significantly contribute in devising personalized treatment plans tailored to the individual patient and tumor. Mathematical models of solid tumors, combined with patient-specific tumor profiles, present a unique opportunity to personalize cancer treatments after detection using a bottom-up approach. Here, we discuss how information harvested from mathematical models and from corresponding in silico experiments can be implemented in preclinical and clinical applications. To conceptually illustrate the power of these models, one such model is presented, and various pertinent tumor and treatment scenarios are demonstrated in silico. The presented model, specifically a multiscale, hybrid cellular automaton, has been fully validated in vitro using multiple cell-line-specific data. We discuss various insights provided by this model and other models like it and their role in designing predictive tools that are both patient, and tumor specific. After refinement and parametrization with appropriate data, such in silico tools have the potential to be used in a clinical setting to aid in treatment protocols and decision making.


Assuntos
Biomarcadores Tumorais/genética , Biologia Computacional/métodos , Modelos Teóricos , Neoplasias/diagnóstico , Neoplasias/terapia , Medicina de Precisão/métodos , Humanos , Neoplasias/genética , Pesquisa Translacional Biomédica
8.
J Theor Biol ; 454: 253-267, 2018 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-29909142

RESUMO

Tumour recurrence post chemotherapy is an established clinical problem and many cancer types are often observed to be increasingly drug resistant subsequent to chemotherapy treatments. Drug resistance in cancer is a multipart phenomenon which can be derived from several origins and in many cases it has been observed that cancer cells have the ability to possess, acquire and communicate drug resistant traits. Here, an in silico framework is developed in order to study drug resistance and drug response in cancer cell populations exhibiting various drug resistant features. The framework is based on an on-lattice hybrid multiscale mathematical model and is equipped to simulate multiple mechanisms on different scales that contribute towards chemotherapeutic drug resistance in cancer. This study demonstrates how drug resistant tumour features may depend on the interplay amongst intracellular, extracelluar and intercellular factors. On a cellular level, drug resistant cell phenotypes are here derived from inheritance or mutations that are spontaneous, drug-induced or communicated via exosomes. Furthermore intratumoural heterogeneity and spatio-temporal drug dynamics heavily influences drug delivery and the development of drug resistant cancer cell subpopulations. Chemotherapy treatment strategies are here optimised for various in silico tumour scenarios and treatment objectives. We demonstrate that optimal chemotherapy treatment strategies drastically depend on which drug resistant mechanisms are activated, and that furthermore suboptimal chemotherapy administration may promote drug resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos , Modelos Teóricos , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Contagem de Células , Ciclo Celular/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Simulação por Computador , Progressão da Doença , Relação Dose-Resposta a Droga , Exossomos/fisiologia , Humanos , Modelos Biológicos , Células Tumorais Cultivadas , Microambiente Tumoral/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA