Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
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 , Humanos , Antineoplásicos/farmacologia , Simulação por Computador , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Melanoma/tratamento farmacológico , Melanoma/genética , Modelos Biológicos , Mutação , Oximas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/efeitos dos fármacos , Proteínas Proto-Oncogênicas B-raf/metabolismo , Piridonas/farmacologia , Pirimidinonas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
2.
J Math Biol ; 87(1): 8, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37318599

RESUMO

Invasion of the surrounding tissue is a key aspect of cancer growth and spread involving a coordinated effort between cell migration and matrix degradation, and has been the subject of mathematical modelling for almost 30 years. In this current paper we address a long-standing question in the field of cancer cell migration modelling. Namely, identify the migratory pattern and spread of individual cancer cells, or small clusters of cancer cells, when the macroscopic evolution of the cancer cell colony is dictated by a specific partial differential equation (PDE). We show that the usual heuristic understanding of the diffusion and advection terms of the PDE being one-to-one responsible for the random and biased motion of the solitary cancer cells, respectively, is not precise. On the contrary, we show that the drift term of the correct stochastic differential equation scheme that dictates the individual cancer cell migration, should account also for the divergence of the diffusion of the PDE. We support our claims with a number of numerical experiments and computational simulations.


Assuntos
Modelos Biológicos , Neoplasias , Humanos , Modelos Teóricos , Movimento Celular , Difusão
3.
J Theor Biol ; 534: 110963, 2022 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-34838584

RESUMO

The formation of new vascular networks is essential for tissue development and regeneration, in addition to playing a key role in pathological settings such as ischemia and tumour development. Experimental findings in the past two decades have led to the identification of a new mechanism of neovascularisation, known as cluster-based vasculogenesis, during which endothelial progenitor cells (EPCs) mobilised from the bone marrow are capable of bridging distant vascular beds in a variety of hypoxic settings in vivo. This process is characterised by the formation of EPC clusters during its early stages and, while much progress has been made in identifying various mechanisms underlying cluster formation, we are still far from a comprehensive description of such spatio-temporal dynamics. In order to achieve this, we propose a novel mathematical model of the early stages of cluster-based vasculogenesis, comprising of a system of nonlocal partial differential equations including key mechanisms such as endogenous chemotaxis, matrix degradation, cell proliferation and cell-to-cell adhesion. We conduct a linear stability analysis on the system and solve the equations numerically. We then conduct a parametric analysis of the numerical solutions of the one-dimensional problem to investigate the role of underlying dynamics on the speed of cluster formation and the size of clusters, measured via appropriate metrics for the cluster width and compactness. We verify the key results of the parametric analysis with simulations of the two-dimensional problem. Our results, which qualitatively compare with data from in vitro experiments, elucidate the complementary role played by endogenous chemotaxis and matrix degradation in the formation of clusters, suggesting chemotaxis is responsible for the cluster topology while matrix degradation is responsible for the speed of cluster formation. Our results also indicate that the nonlocal cell-to-cell adhesion term in our model, even though it initially causes cells to aggregate, is not sufficient to ensure clusters are stable over long time periods. Consequently, new modelling strategies for cell-to-cell adhesion are required to stabilise in silico clusters. We end the paper with a thorough discussion of promising, fruitful future modelling and experimental research perspectives.


Assuntos
Células Progenitoras Endoteliais , Neovascularização Fisiológica , Diferenciação Celular , Células Progenitoras Endoteliais/metabolismo , Humanos , Neovascularização Patológica/metabolismo , Células-Tronco
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.
R Soc Open Sci ; 8(6): 202237, 2021 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-34150312

RESUMO

We present two different methods to estimate parameters within a partial differential equation model of cancer invasion. The model describes the spatio-temporal evolution of three variables-tumour cell density, extracellular matrix density and matrix degrading enzyme concentration-in a one-dimensional tissue domain. The first method is a likelihood-free approach associated with approximate Bayesian computation; the second is a two-stage gradient matching method based on smoothing the data with a generalized additive model (GAM) and matching gradients from the GAM to those from the model. Both methods performed well on simulated data. To increase realism, additionally we tested the gradient matching scheme with simulated measurement error and found that the ability to estimate some model parameters deteriorated rapidly as measurement error increased.

7.
J Theor Biol ; 522: 110677, 2021 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-33781776

RESUMO

We develop a three-dimensional genuinely hybrid atomistic-continuum model that describes the invasive growth dynamics of individual cancer cells in tissue. The framework explicitly accounts for phenotypic variation by distinguishing between cancer cells of an epithelial-like and a mesenchymal-like phenotype. It also describes mutations between these cell phenotypes in the form of epithelial-mesenchymal transition (EMT) and its reverse process mesenchymal-epithelial transition (MET). The proposed model consists of a hybrid system of partial and stochastic differential equations that describe the evolution of epithelial-like and mesenchymal-like cancer cells, respectively, under the consideration of matrix-degrading enzyme concentrations and the extracellular matrix density. With the help of inverse parameter estimation and a sensitivity analysis, this three-dimensional model is then calibrated to an in vitro organotypic invasion assay experiment of oral squamous cell carcinoma cells.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Linhagem Celular Tumoral , Simulação por Computador , Humanos , Invasividade Neoplásica
8.
Phys Med Biol ; 66(4): 045026, 2021 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-33339019

RESUMO

Understanding and designing clinical radiation therapy is one of the most important areas of state-of-the-art oncological treatment regimens. Decades of research have gone into developing sophisticated treatment devices and optimization protocols for schedules and dosages. In this paper, we presented a comprehensive computational platform that facilitates building of the sophisticated multi-cell-based model of how radiation affects the biology of living tissue. We designed and implemented a coupled simulation method, including a radiation transport model, and a cell biology model, to simulate the tumor response after irradiation. The radiation transport simulation was implemented through Geant4 which is an open-source Monte Carlo simulation platform that provides many flexibilities for users, as well as low energy DNA damage simulation physics, Geant4-DNA. The cell biology simulation was implemented using CompuCell3D (CC3D) which is a cell biology simulation platform. In order to couple Geant4 solver with CC3D, we developed a 'bridging' module, RADCELL, that extracts tumor cellular geometry of the CC3D simulation (including specification of the individual cells) and ported it to the Geant4 for radiation transport simulation. The cell dose and cell DNA damage distribution in multicellular system were obtained using Geant4. The tumor response was simulated using cell-based tissue models based on CC3D, and the cell dose and cell DNA damage information were fed back through RADCELL to CC3D for updating the cell properties. By merging two powerful and widely used modeling platforms, CC3D and Geant4, we delivered a novel tool that can give us the ability to simulate the dynamics of biological tissue in the presence of ionizing radiation, which provides a framework for quantifying the biological consequences of radiation therapy. In this introductory methods paper, we described our modeling platform in detail and showed how it can be applied to study the application of radiotherapy to a vascularized tumor.


Assuntos
Simulação por Computador , Neoplasias de Tecido Vascular/radioterapia , Radiobiologia/métodos , Radioterapia/métodos , Relação Dose-Resposta à Radiação , Humanos , Modelos Biológicos , Método de Monte Carlo , Neoplasias de Tecido Vascular/fisiopatologia , Doses de Radiação , Radiação Ionizante , Software
9.
J Math Biol ; 80(1-2): 343-371, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31183520

RESUMO

Continuum models for the spatial dynamics of growing cell populations have been widely used to investigate the mechanisms underpinning tissue development and tumour invasion. These models consist of nonlinear partial differential equations that describe the evolution of cellular densities in response to pressure gradients generated by population growth. Little prior work has explored the relation between such continuum models and related single-cell-based models. We present here a simple stochastic individual-based model for the spatial dynamics of multicellular systems whereby cells undergo pressure-driven movement and pressure-dependent proliferation. We show that nonlinear partial differential equations commonly used to model the spatial dynamics of growing cell populations can be formally derived from the branching random walk that underlies our discrete model. Moreover, we carry out a systematic comparison between the individual-based model and its continuum counterparts, both in the case of one single cell population and in the case of multiple cell populations with different biophysical properties. The outcomes of our comparative study demonstrate that the results of computational simulations of the individual-based model faithfully mirror the qualitative and quantitative properties of the solutions to the corresponding nonlinear partial differential equations. Ultimately, these results illustrate how the simple rules governing the dynamics of single cells in our individual-based model can lead to the emergence of complex spatial patterns of population growth observed in continuum models.


Assuntos
Movimento Celular/fisiologia , Modelos Biológicos , Ciclo Celular/fisiologia , Proliferação de Células/fisiologia , Simulação por Computador , Análise Espacial
10.
Bull Math Biol ; 81(6): 1965-2010, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30903592

RESUMO

Cancer is a complex disease that starts with mutations of key genes in one cell or a small group of cells at a primary site in the body. If these cancer cells continue to grow successfully and, at some later stage, invade the surrounding tissue and acquire a vascular network, they can spread to distant secondary sites in the body. This process, known as metastatic spread, is responsible for around 90% of deaths from cancer and is one of the so-called hallmarks of cancer. To shed light on the metastatic process, we present a mathematical modelling framework that captures for the first time the interconnected processes of invasion and metastatic spread of individual cancer cells in a spatially explicit manner-a multigrid, hybrid, individual-based approach. This framework accounts for the spatiotemporal evolution of mesenchymal- and epithelial-like cancer cells, membrane-type-1 matrix metalloproteinase (MT1-MMP) and the diffusible matrix metalloproteinase-2 (MMP-2), and for their interactions with the extracellular matrix. Using computational simulations, we demonstrate that our model captures all the key steps of the invasion-metastasis cascade, i.e. invasion by both heterogeneous cancer cell clusters and by single mesenchymal-like cancer cells; intravasation of these clusters and single cells both via active mechanisms mediated by matrix-degrading enzymes (MDEs) and via passive shedding; circulation of cancer cell clusters and single cancer cells in the vasculature with the associated risk of cell death and disaggregation of clusters; extravasation of clusters and single cells; and metastatic growth at distant secondary sites in the body. By faithfully reproducing experimental results, our simulations support the evidence-based hypothesis that the membrane-bound MT1-MMP is the main driver of invasive spread rather than diffusible MDEs such as MMP-2.


Assuntos
Modelos Biológicos , Invasividade Neoplásica , Metástase Neoplásica , Animais , Neoplasias da Mama/patologia , Neoplasias da Mama/fisiopatologia , Comunicação Celular/fisiologia , Simulação por Computador , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Humanos , Masculino , Conceitos Matemáticos , Metaloproteinase 14 da Matriz/fisiologia , Metaloproteinase 2 da Matriz/fisiologia , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/patologia , Metástase Neoplásica/fisiopatologia , Células Neoplásicas Circulantes/patologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/fisiologia , Análise Espaço-Temporal , Biologia de Sistemas
11.
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
12.
J Theor Biol ; 451: 101-110, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-29750997

RESUMO

We present here a space- and phenotype-structured model of selection dynamics between cancer cells within a solid tumour. In the framework of this model, we combine formal analyses with numerical simulations to investigate in silico the role played by the spatial distribution of abiotic components of the tumour microenvironment in mediating phenotypic selection of cancer cells. Numerical simulations are performed both on the 3D geometry of an in silico multicellular tumour spheroid and on the 3D geometry of an in vivo human hepatic tumour, which was imaged using computerised tomography. The results obtained show that inhomogeneities in the spatial distribution of oxygen, currently observed in solid tumours, can promote the creation of distinct local niches and lead to the selection of different phenotypic variants within the same tumour. This process fosters the emergence of stable phenotypic heterogeneity and supports the presence of hypoxic cells resistant to cytotoxic therapy prior to treatment. Our theoretical results demonstrate the importance of integrating spatial data with ecological principles when evaluating the therapeutic response of solid tumours to cytotoxic therapy.


Assuntos
Modelos Biológicos , Neoplasias/patologia , Humanos , Neoplasias Hepáticas/patologia , Modelos de Interação Espacial , Fenótipo , Esferoides Celulares , Microambiente Tumoral
13.
Bull Math Biol ; 80(6): 1539-1562, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29536427

RESUMO

A growing body of experimental evidence indicates that immune cells move in an unrestricted search pattern if they are in the pre-activated state, whilst they tend to stay within a more restricted area upon activation induced by the presence of tumour antigens. This change in movement is not often considered in the existing mathematical models of the interactions between immune cells and cancer cells. With the aim to fill such a gap in the existing literature, in this work we present a spatially structured individual-based model of tumour-immune competition that takes explicitly into account the difference in movement between inactive and activated immune cells. In our model, a Lévy walk is used to capture the movement of inactive immune cells, whereas Brownian motion is used to describe the movement of antigen-activated immune cells. The effects of activation of immune cells, the proliferation of cancer cells and the immune destruction of cancer cells are also modelled. We illustrate the ability of our model to reproduce qualitatively the spatial trajectories of immune cells observed in experimental data of single-cell tracking. Computational simulations of our model further clarify the conditions for the onset of a successful immune action against cancer cells and may suggest possible targets to improve the efficacy of cancer immunotherapy. Overall, our theoretical work highlights the importance of taking into account spatial interactions when modelling the immune response to cancer cells.


Assuntos
Modelos Imunológicos , Neoplasias/imunologia , Linfócitos T/imunologia , Movimento Celular/imunologia , Proliferação de Células , Simulação por Computador , Células Dendríticas/imunologia , Humanos , Imunoterapia , Ativação Linfocitária , Conceitos Matemáticos , Neoplasias/patologia , Neoplasias/terapia , Linfócitos T Citotóxicos/imunologia
14.
Bull Math Biol ; 80(4): 701-737, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29500719

RESUMO

Spatio-temporal models have long been used to describe biological systems of cancer, but it has not been until very recently that increased attention has been paid to structural dynamics of the interaction between cancer populations and the molecular mechanisms associated with local invasion. One system that is of particular interest is that of the urokinase plasminogen activator (uPA) wherein uPA binds uPA receptors on the cancer cell surface, allowing plasminogen to be cleaved into plasmin, which degrades the extracellular matrix and this way leads to enhanced cancer cell migration. In this paper, we develop a novel numerical approach and associated analysis for spatio-structuro-temporal modelling of the uPA system for up to two-spatial and two-structural dimensions. This is accompanied by analytical exploration of the numerical techniques used in simulating this system, with special consideration being given to the proof of stability within numerical regimes encapsulating a central differences approach to approximating numerical gradients. The stability analysis performed here reveals instabilities induced by the coupling of the structural binding and proliferative processes. The numerical results expound how the uPA system aids the tumour in invading the local stroma, whilst the inhibitor to this system may impede this behaviour and encourage a more sporadic pattern of invasion.


Assuntos
Modelos Biológicos , Invasividade Neoplásica/patologia , Invasividade Neoplásica/fisiopatologia , Neoplasias/metabolismo , Neoplasias/patologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Movimento Celular , Simulação por Computador , Matriz Extracelular/metabolismo , Humanos , Conceitos Matemáticos , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Análise Espaço-Temporal
15.
J Theor Biol ; 446: 87-100, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29524441

RESUMO

If improvements are to be made in tuberculosis (TB) treatment, an increased understanding of disease in the lung is needed. Studies have shown that bacteria in a less metabolically active state, associated with the presence of lipid bodies, are less susceptible to antibiotics, and recent results have highlighted the disparity in concentration of different compounds into lesions. Treatment success therefore depends critically on the responses of the individual bacteria that constitute the infection. We propose a hybrid, individual-based approach that analyses spatio-temporal dynamics at the cellular level, linking the behaviour of individual bacteria and host cells with the macroscopic behaviour of the microenvironment. The individual elements (bacteria, macrophages and T cells) are modelled using cellular automaton (CA) rules, and the evolution of oxygen, drugs and chemokine dynamics are incorporated in order to study the effects of the microenvironment in the pathological lesion. We allow bacteria to switch states depending on oxygen concentration, which affects how they respond to treatment. This is the first multiscale model of its type to consider both oxygen-driven phenotypic switching of the Mycobacterium tuberculosis and antibiotic treatment. Using this model, we investigate the role of bacterial cell state and of initial bacterial location on treatment outcome. We demonstrate that when bacteria are located further away from blood vessels, less favourable outcomes are more likely, i.e. longer time before infection is contained/cleared, treatment failure or later relapse. We also show that in cases where bacteria remain at the end of simulations, the organisms tend to be slower-growing and are often located within granulomas, surrounded by caseous material.


Assuntos
Antibacterianos/uso terapêutico , Granuloma , Modelos Biológicos , Mycobacterium tuberculosis/metabolismo , Tuberculose Pulmonar , Granuloma/tratamento farmacológico , Granuloma/metabolismo , Granuloma/microbiologia , Humanos , Tuberculose Pulmonar/tratamento farmacológico , Tuberculose Pulmonar/metabolismo
16.
Math Med Biol ; 35(4): 541-577, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-29346560

RESUMO

Cells adhere to each other and to the extracellular matrix (ECM) through protein molecules on the surface of the cells. The breaking and forming of adhesive bonds, a process critical in cancer invasion and metastasis, can be influenced by the mutation of cancer cells. In this paper, we develop a nonlocal mathematical model describing cancer cell invasion and movement as a result of integrin-controlled cell-cell adhesion and cell-matrix adhesion, for two cancer cell populations with different levels of mutation. The partial differential equations for cell dynamics are coupled with ordinary differential equations describing the ECM degradation and the production and decay of integrins. We use this model to investigate the role of cancer mutation on the possibility of cancer clonal competition with alternating dominance, or even competitive exclusion (phenomena observed experimentally). We discuss different possible cell aggregation patterns, as well as travelling wave patterns. In regard to the travelling waves, we investigate the effect of cancer mutation rate on the speed of cancer invasion.


Assuntos
Adesão Celular , Movimento Celular , Matriz Extracelular , Modelos Teóricos , Invasividade Neoplásica , Neoplasias , Humanos
17.
Bull Math Biol ; 80(5): 1366-1403, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28634857

RESUMO

In this paper, we present two mathematical models related to different aspects and scales of cancer growth. The first model is a stochastic spatiotemporal model of both a synthetic gene regulatory network (the example of a three-gene repressilator is given) and an actual gene regulatory network, the NF-[Formula: see text]B pathway. The second model is a force-based individual-based model of the development of a solid avascular tumour with specific application to tumour cords, i.e. a mass of cancer cells growing around a central blood vessel. In each case, we compare our computational simulation results with experimental data. In the final discussion section, we outline how to take the work forward through the development of a multiscale model focussed at the cell level. This would incorporate key intracellular signalling pathways associated with cancer within each cell (e.g. p53-Mdm2, NF-[Formula: see text]B) and through the use of high-performance computing be capable of simulating up to [Formula: see text] cells, i.e. the tissue scale. In this way, mathematical models at multiple scales would be combined to formulate a multiscale computational model.


Assuntos
Modelos Biológicos , Neoplasias/patologia , Animais , Simulação por Computador , Redes Reguladoras de Genes , Humanos , Conceitos Matemáticos , Neoplasias/genética , Transdução de Sinais , Análise Espaço-Temporal , Processos Estocásticos
18.
J Math Biol ; 75(6-7): 1517-1561, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28405746

RESUMO

The dynamic interplay between collective cell movement and the various molecules involved in the accompanying cell signalling mechanisms plays a crucial role in many biological processes including normal tissue development and pathological scenarios such as wound healing and cancer. Information about the various structures embedded within these processes allows a detailed exploration of the binding of molecular species to cell-surface receptors within the evolving cell population. In this paper we establish a general spatio-temporal-structural framework that enables the description of molecular binding to cell membranes coupled with the cell population dynamics. We first provide a general theoretical description for this approach and then illustrate it with three examples arising from cancer invasion.


Assuntos
Movimento Celular/fisiologia , Modelos Biológicos , Comunicação Celular/fisiologia , Proliferação de Células/fisiologia , Simulação por Computador , Matriz Extracelular/fisiologia , Humanos , Conceitos Matemáticos , Metaloproteinase 14 da Matriz/fisiologia , Invasividade Neoplásica/fisiopatologia , Receptores de Superfície Celular/fisiologia , Receptores de Ativador de Plasminogênio Tipo Uroquinase/fisiologia , Transdução de Sinais/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/fisiologia
19.
Bull Math Biol ; 79(3): 389-429, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28210916

RESUMO

Known as one of the hallmarks of cancer (Hanahan and Weinberg in Cell 100:57-70, 2000) cancer cell invasion of human body tissue is a complicated spatio-temporal multiscale process which enables a localised solid tumour to transform into a systemic, metastatic and fatal disease. This process explores and takes advantage of the reciprocal relation that solid tumours establish with the extracellular matrix (ECM) components and other multiple distinct cell types from the surrounding microenvironment. Through the secretion of various proteolytic enzymes such as matrix metalloproteinases or the urokinase plasminogen activator (uPA), the cancer cell population alters the configuration of the surrounding ECM composition and overcomes the physical barriers to ultimately achieve local cancer spread into the surrounding tissue. The active interplay between the tissue-scale tumour dynamics and the molecular mechanics of the involved proteolytic enzymes at the cell scale underlines the biologically multiscale character of invasion and raises the challenge of modelling this process with an appropriate multiscale approach. In this paper, we present a new two-scale moving boundary model of cancer invasion that explores the tissue-scale tumour dynamics in conjunction with the molecular dynamics of the urokinase plasminogen activation system. Building on the multiscale moving boundary method proposed in Trucu et al. (Multiscale Model Simul 11(1):309-335, 2013), the modelling that we propose here allows us to study the changes in tissue-scale tumour morphology caused by the cell-scale uPA microdynamics occurring along the invasive edge of the tumour. Our computational simulation results demonstrate a range of heterogeneous dynamics which are qualitatively similar to the invasive growth patterns observed in a number of different types of cancer, such as the tumour infiltrative growth patterns discussed in Ito et al. (J Gastroenterol 47:1279-1289, 2012).


Assuntos
Modelos Biológicos , Invasividade Neoplásica/fisiopatologia , Algoritmos , Simulação por Computador , Matriz Extracelular/metabolismo , Fibrinolisina/metabolismo , Humanos , Conceitos Matemáticos , Invasividade Neoplásica/patologia , Microambiente Tumoral/fisiologia , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
20.
Math Med Biol ; 34(1): 1-13, 2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-26443812

RESUMO

278: The Cytochrome P450 (CYP) system is involved in 90% of the human body's interactions with xenobiotics and due to this, it has become an area of avid research including the creation of transgenic mice. This paper proposes a three-compartment model which is used to explain the drug metabolism in the Hepatic Reductase Null (HRN) mouse developed by the University of Dundee (Henderson, C. J., Otto, D. M. E., Carrie, D., Magnuson, M. A., McLaren, A. W., Rosewell, I. and Wolf, C. R. (2003) Inactivation of the hepatic cytochrome p450 system by conditional deletion of hepatic cytochrome p450 reductase. J. Biol. Chem. , 13480-13486). The model is compared with a two-compartment model using experimental data from studies using wild-type and HRN mice. This comparison allowed for metabolic differences between the two types of mice to be isolated. The three sets of drug data (Gefitinib, Midazolam and Thalidomide) showed that the transgenic mouse has a decreased rate of metabolism.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Fígado/enzimologia , Modelos Biológicos , NADPH-Ferri-Hemoproteína Redutase/metabolismo , Inibidores da Angiogênese/metabolismo , Animais , Modelos Animais de Doenças , Moduladores GABAérgicos/metabolismo , Gefitinibe , Camundongos , Camundongos Transgênicos/metabolismo , Midazolam/metabolismo , Inibidores de Proteínas Quinases/metabolismo , Quinazolinas/metabolismo , Talidomida/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA