Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Int J Mol Sci ; 22(12)2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-34208139

RESUMO

Glioblastoma is the most malignant brain tumor among adults. Despite multimodality treatment, it remains incurable, mainly because of its extensive heterogeneity and infiltration in the brain parenchyma. Recent evidence indicates dysregulation of the expression of the Promyelocytic Leukemia Protein (PML) in primary Glioblastoma samples. PML is implicated in various ways in cancer biology. In the brain, PML participates in the physiological migration of the neural progenitor cells, which have been hypothesized to serve as the cell of origin of Glioblastoma. The role of PML in Glioblastoma progression has recently gained attention due to its controversial effects in overall Glioblastoma evolution. In this work, we studied the role of PML in Glioblastoma pathophysiology using the U87MG cell line. We genetically modified the cells to conditionally overexpress the PML isoform IV and we focused on its dual role in tumor growth and invasive capacity. Furthermore, we targeted a PML action mediator, the Enhancer of Zeste Homolog 2 (EZH2), via the inhibitory drug DZNeP. We present a combined in vitro-in silico approach, that utilizes both 2D and 3D cultures and cancer-predictive computational algorithms, in order to differentiate and interpret the observed biological results. Our overall findings indicate that PML regulates growth and invasion through distinct cellular mechanisms. In particular, PML overexpression suppresses cell proliferation, while it maintains the invasive capacity of the U87MG Glioblastoma cells and, upon inhibition of the PML-EZH2 pathway, the invasion is drastically eliminated. Our in silico simulations suggest that the underlying mechanism of PML-driven Glioblastoma physiology regulates invasion by differential modulation of the cell-to-cell adhesive and diffusive capacity of the cells. Elucidating further the role of PML in Glioblastoma biology could set PML as a potential molecular biomarker of the tumor progression and its mediated pathway as a therapeutic target, aiming at inhibiting cell growth and potentially clonal evolution regarding their proliferative and/or invasive phenotype within the heterogeneous tumor mass.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proteína da Leucemia Promielocítica/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Simulação por Computador , Humanos , Modelos Biológicos , Invasividade Neoplásica , Esferoides Celulares/patologia
2.
Sci Rep ; 14(1): 3759, 2024 02 14.
Artigo em Inglês | MEDLINE | ID: mdl-38355655

RESUMO

Adjuvant Temozolomide is considered the front-line Glioblastoma chemotherapeutic treatment; yet not all patients respond. Latest trends in clinical trials usually refer to Doxorubicin; yet it can lead to severe side-effects if administered in high doses. While Glioblastoma prognosis remains poor, little is known about the combination of the two chemotherapeutics. Patient-derived spheroids were generated and treated with a range of Temozolomide/Doxorubicin concentrations either as monotherapy or in combination. Optical microscopy was used to monitor the growth pattern and cell death. Based on the monotherapy experiments, we developed a probabilistic mathematical framework in order to describe the drug-induced effect at the single-cell level and simulate drug doses in combination assuming probabilistic independence. Doxorubicin was found to be effective in doses even four orders of magnitude less than Temozolomide in monotherapy. The combination therapy doses tested in vitro were able to lead to irreversible growth inhibition at doses where monotherapy resulted in relapse. In our simulations, we assumed both drugs are anti-mitotic; Temozolomide has a growth-arrest effect, while Doxorubicin is able to cumulatively cause necrosis. Interestingly, under no mechanistic synergy assumption, the in silico predictions underestimate the in vitro results. In silico models allow the exploration of a variety of potential underlying hypotheses. The simulated-biological discrepancy at certain doses indicates a supra-additive response when both drugs are combined. Our results suggest a Temozolomide-Doxorubicin dual chemotherapeutic scheme to both disable proliferation and increase cytotoxicity against Glioblastoma.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Linhagem Celular Tumoral , Recidiva Local de Neoplasia , Doxorrubicina/farmacologia , Doxorrubicina/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo
3.
J Pers Med ; 14(5)2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38793058

RESUMO

The massive amount of human biological, imaging, and clinical data produced by multiple and diverse sources necessitates integrative modeling approaches able to summarize all this information into answers to specific clinical questions. In this paper, we present a hypermodeling scheme able to combine models of diverse cancer aspects regardless of their underlying method or scale. Describing tissue-scale cancer cell proliferation, biomechanical tumor growth, nutrient transport, genomic-scale aberrant cancer cell metabolism, and cell-signaling pathways that regulate the cellular response to therapy, the hypermodel integrates mutation, miRNA expression, imaging, and clinical data. The constituting hypomodels, as well as their orchestration and links, are described. Two specific cancer types, Wilms tumor (nephroblastoma) and non-small cell lung cancer, are addressed as proof-of-concept study cases. Personalized simulations of the actual anatomy of a patient have been conducted. The hypermodel has also been applied to predict tumor control after radiotherapy and the relationship between tumor proliferative activity and response to neoadjuvant chemotherapy. Our innovative hypermodel holds promise as a digital twin-based clinical decision support system and as the core of future in silico trial platforms, although additional retrospective adaptation and validation are necessary.

4.
IEEE Rev Biomed Eng ; 16: 456-471, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-34506292

RESUMO

The main reason why therapeutic schemes fail in Glioblastoma lies on its own peculiarities as a cancer and on our failure to fully decipher them. Fast tumor evolution, invasiveness and incomplete surgical resection contribute to disease recurrence, therapy resistance and high mortality. More faithful models must be developed to address Glioblastoma biology and better clinical guidance. Research studies are discussed in this review that: i) improve understanding and assessment of the growth mechanisms of Glioblastoma and ii) develop preclinical models (in vitro-in vivo-in silico) that mimic patient's tumor (phenocopying) in order to provide better prediction of response to therapies.


Assuntos
Glioblastoma , Humanos , Glioblastoma/patologia , Glioblastoma/terapia
5.
Vaccines (Basel) ; 11(4)2023 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-37112635

RESUMO

The regulation policies implemented, the characteristics of vaccines, and the evolution of the virus continue to play a significant role in the progression of the SARS-CoV-2 pandemic. Numerous research articles have proposed using mathematical models to predict the outcomes of different scenarios, with the aim of improving awareness and informing policy-making. In this work, we propose an expansion to the classical SEIR epidemiological model that is designed to fit the complex epidemiological data of COVID-19. The model includes compartments for vaccinated, asymptomatic, hospitalized, and deceased individuals, splitting the population into two branches based on the severity of progression. In order to investigate the impact of the vaccination program on the spread of COVID-19 in Greece, this study takes into account the realistic vaccination program implemented in Greece, which includes various vaccination rates, different dosages, and the administration of booster shots. It also examines for the first time policy scenarios at crucial time-intervention points for Greece. In particular, we explore how alterations in the vaccination rate, immunity loss, and relaxation of measures regarding the vaccinated individuals affect the dynamics of COVID-19 spread. The modeling parameters revealed an alarming increase in the death rate during the dominance of the delta variant and before the initiation of the booster shot program in Greece. The existing probability of vaccinated people becoming infected and transmitting the virus sets them as catalytic players in COVID-19 progression. Overall, the modeling observations showcase how the criticism of different intervention measures, the vaccination program, and the virus evolution has been present throughout the various stages of the pandemic. As long as immunity declines, new variants emerge, and vaccine protection in reducing transmission remains incompetent; monitoring the complex vaccine and virus evolution is critical to respond proactively in the future.

6.
Int J Numer Method Biomed Eng ; 39(7): e3734, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37203371

RESUMO

Glioblastoma is the most aggressive and infiltrative glioma, classified as Grade IV, with the poorest survival rate among patients. Accurate and rigorously tested mechanistic in silico modeling offers great value to understand and quantify the progression of primary brain tumors. This paper presents a continuum-based finite element framework that is built on high performance computing, open-source libraries to simulate glioblastoma progression. We adopt the established proliferation invasion hypoxia necrosis angiogenesis model in our framework to realize scalable simulations of cancer, and has demonstrated to produce accurate and efficient solutions in both two- and three-dimensional brain models. The in silico solver can successfully implement arbitrary order discretization schemes and adaptive remeshing algorithms. A model sensitivity analysis is conducted to test the impact of vascular density, cancer cell invasiveness and aggressiveness, the phenotypic transition potential, including that of necrosis, and the effect of tumor-induced angiogenesis in the evolution of glioblastoma. Additionally, individualized simulations of brain cancer progression are carried out using pertinent magnetic resonance imaging data, where the in silico model is used to investigate the complex dynamics of the disease. We conclude by arguing how the proposed framework can deliver patient-specific simulations of cancer prognosis and how it could bridge clinical imaging with modeling.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Análise de Elementos Finitos , Neoplasias Encefálicas/diagnóstico por imagem , Simulação por Computador , Neovascularização Patológica , Necrose , Encéfalo/patologia
7.
IEEE J Biomed Health Inform ; 26(3): 1188-1195, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34379601

RESUMO

The development of label-free non-destructive techniques to be used as diagnostic tools in cancer research is of great importance for improving the quality of life for millions of patients. Previous studies have demonstrated that Third Harmonic Generation (THG) imaging could differentiate malignant from benign unlabeled human breast biopsies and distinguish the different grades of cancer. Towards the application of such technologies to clinic, in the present report, a deep learning technique was applied to THG images recorded from breast cancer tissues of grades 0, I, II, and III. By the implementation of a convolutional neural network (CNN) model, the differentiation of malignant from benign breast tissue samples and the discrimination of the different grades of cancer in a fast and accurate way were achieved. The obtained results provide a step ahead towards the use of optical diagnostic tools in conjunction with the CNN image classifier for the reliable and rapid malignancy diagnosis in clinic.


Assuntos
Neoplasias da Mama , Aprendizado Profundo , Biópsia , Mama/diagnóstico por imagem , Mama/patologia , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/patologia , Feminino , Humanos , Qualidade de Vida
8.
Front Oncol ; 10: 1552, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042800

RESUMO

Tumors are complex, dynamic, and adaptive biological systems characterized by high heterogeneity at genetic, epigenetic, phenotypic, as well as tissue microenvironmental level. In this work, utilizing cellular automata methods, we focus on intrinsic heterogeneity with respect to cell cycle duration and explore whether and to what extent this heterogeneity affects cancer cell growth dynamics when cytotoxic treatment is applied. We assume that treatment acts on cancer cells specifically during mitosis and compare it with a (cell cycle-non-specific) cytotoxic treatment that acts randomly regardless of the cell cycle phase. We simulate the spatiotemporal evolution of tumor cells with different initial spatial configurations and different cell length probability distributions. We observed that in heterogeneous populations, strong selection forces act on cancer cells favoring the faster cells, when the death rates are lower than the proliferation rates. However, at higher mitotic death rates, selection of the slower proliferative cells is favored, leading to slower post-treatment regrowth rates, as compared to untreated growth. Of note, random cell death progressively eliminates the slower proliferative cells, consistently, favoring highly proliferative phenotypes. Interestingly, compared to the monoclonal populations that exhibit complete response at high random death rates, emergent resistance arises naturally in heterogeneous populations during treatment. As divergent selection forces may act on a heterogeneous cancer cell population, we argue that treatment plan selection can considerably alter the post-treatment tumor dynamics, cell survival, and emergence of resistance, proving its significant biological and therapeutic impact.

9.
IEEE J Biomed Health Inform ; 23(5): 1844-1854, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30605113

RESUMO

Metabolic reprogramming is a hallmark of cancer. The main aim of this paper is to integrate a genome-scale metabolic description of tumor cells into a tumor growth model that accounts for the spatiotemporally heterogeneous tumor microenvironment, in order to study the effects of microscopic characteristics on tumor evolution. A lactate maximization metabolic strategy that allows near-optimal growth solution, while maximizing lactate secretion, is assumed. The proposed sub-cellular metabolic model is then incorporated into a hybrid discrete-continuous model of tumor growth. We produced several phenotypes by applying different constraints and optimization criteria in the metabolic model and explored the tumor evolution of the various phenotypes in different vasculature conditions and extracellular matrix densities. At first, we showed that the metabolic capabilities of phenotypes depending on resource availability can vary in a counter-intuitive manner. We then showed that: first, tumor population, morphology, and spread are affected differently in different conditions, allowing thus phenotypes to be superior than others in different conditions; and second, polyclonal tumors consisting of different phenotypes can exploit their different metabolic capabilities to enhance further tumor evolution. The proposed framework comprises a proof-of-concept demonstration showing the importance of considering the metabolic capabilities of phenotypes on predicting tumor evolution. The proposed framework allows the incorporation of context-specific and patient-specific data for the study of personalized tumor evolution and therapy efficacy, linking genome to metabolic capabilities and tumor dynamics.


Assuntos
Biologia Computacional/métodos , Glucose/metabolismo , Glicólise/fisiologia , Modelos Biológicos , Neoplasias , Proliferação de Células , Humanos , Análise do Fluxo Metabólico , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Células Tumorais Cultivadas , Microambiente Tumoral/fisiologia
10.
Tissue Cell ; 59: 39-43, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31383287

RESUMO

Major Glioblastoma's hallmarks include proliferation, invasion and heterogeneity. Biological 3D tumor spheroid models can serve as intermediate systems between traditional 2D cell culture and complex in vivo models. Tumor spheroids have been shown to more accurately reproduce the spatial organization and microenvironmental factors of in vivo micro-tumors, such as relevant gradients of nutrients and other molecular agents, while they maintain cell-to-cell and cell-to-matrix interactions. In vitro 3D assays are useful to monitor these properties. Here, we test the suitability of the well-known T98 G Glioblastoma cell line in such a 3D assay. The doubling time and death rate parameters of T98 G are estimated, as well as their spheroidal growth-expansion curves with and without the presence of basement membrane substrate. The T98 G invasive profile is characterized by collective morphology and proliferation-associated invasion. We show that the T98 G secondary GB cell line exhibits both invasive and proliferative capabilities in 3D and thus, can serve as control cell line for the 3D in vitro study of primary GB cell cultures.


Assuntos
Glioblastoma , Modelos Biológicos , Esferoides Celulares , Linhagem Celular Tumoral , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia
11.
Annu Int Conf IEEE Eng Med Biol Soc ; 2016: 6142-6145, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28269654

RESUMO

Anti-cancer therapy efficacy in solid tumors mainly depends on drug transportation through the vasculature system and the extracellular matrix, on diffusion gradients and clonal heterogeneity within the tumor mass, as well as on the responses of the individual tumor cells to drugs and their interactions with each other and their local microenvironment. In this work, we develop a mathematical predictive model for tumor growth and drug response based on 3D spheroids experiments that possess several in vivo features of tumors and are considered better for drug screening. The model takes into account the diffusion gradients of both oxygen and drug through the tumor volume, describes the tumor population at cell level and assumes a simple underlying cellular dose-response curve that is translated to a cell death probability. The model shows that although the endpoint tumor regression can be well approximated, the effects of the drug on cell fate necessitate a more sophisticated model to explain the temporal evolution of tumor regression and more quantitative information regarding the number and topology of dead and living cells, which is highly important for in vivo clinical relevant predictions. The model is built in a way that can be constrained by experimentally derived set of parameters and is capable of accommodating cell heterogeneity, sub-cellular regulatory mechanisms and drug-induced signaling cascades, as well as additional mechanisms of adapted resistance.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais/métodos , Esferoides Celulares/efeitos dos fármacos , Técnicas de Cultura de Células , Difusão , Humanos , Modelos Teóricos , Neoplasias/tratamento farmacológico , Oxigênio/metabolismo , Células Tumorais Cultivadas
12.
Cancer Inform ; 14(Suppl 4): 67-81, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26396490

RESUMO

Modeling tumor growth has proven a very challenging problem, mainly due to the fact that tumors are highly complex systems that involve dynamic interactions spanning multiple scales both in time and space. The desire to describe interactions in various scales has given rise to modeling approaches that use both continuous and discrete variables, known as hybrid approaches. This work refers to a hybrid model on a 2D square lattice focusing on cell movement dynamics as they play an important role in tumor morphology, invasion and metastasis and are considered as indicators for the stage of malignancy used for early prognosis and effective treatment. Considering various distributions of the microenvironment, we explore how Neumann vs. Moore neighborhood schemes affects tumor growth and morphology. The results indicate that the importance of neighborhood selection is critical under specific conditions that include i) increased hapto/chemo-tactic coefficient, ii) a rugged microenvironment and iii) ECM degradation.

13.
Cancer Inform ; 14(Suppl 4): 7-18, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26085787

RESUMO

Glioblastoma multiforme is the most aggressive type of glioma and the most common malignant primary intra-axial brain tumor. In an effort to predict the evolution of the disease and optimize therapeutical decisions, several models have been proposed for simulating the growth pattern of glioma. One of the latest models incorporates cell proliferation and invasion, angiogenic net rates, oxygen consumption, and vasculature. These factors, particularly oxygenation levels, are considered fundamental factors of tumor heterogeneity and compartmentalization. This paper focuses on the initialization of the cancer cell populations and vasculature based on imaging examinations of the patient and presents a feasibility study on vasculature prediction over time. To this end, pharmacokinetic parameters derived from dynamic contrast-enhanced magnetic resonance imaging using Toft's model are used in order to feed the model. K (trans) is used as a metric of the density of endothelial cells (vasculature); at the same time, it also helps to discriminate distinct image areas of interest, under a set of assumptions. Feasibility results of applying the model to a real clinical case are presented, including a study on the effect of certain parameters on the pattern of the simulated tumor.

14.
PLoS One ; 9(8): e103191, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25099885

RESUMO

Tumor is characterized by extensive heterogeneity with respect to its microenvironment and its genetic composition. We extend a previously developed monoclonal continuous spatial model of tumor growth to account for polyclonal cell populations and investigate the interplay between a more proliferative and a more invasive phenotype under different conditions. The model simulations demonstrate a transition from the dominance of the proliferative to the dominance of the invasive phenotype resembling malignant tumor progression and show a time period where both subpopulations are abundant. As the dominant phenotype switches from proliferative to invasive, the geometry of tumor changes from a compact and almost spherical shape to a more diffusive and fingered morphology with the proliferative phenotype to be restricted in the tumor bulk and the invasive to dominate at tumor edges. Different micro-environmental conditions and different phenotypic properties can promote or inhibit invasion demonstrating their mutual importance. The model provides a computational framework to investigate tumor heterogeneity and the constant interplay between the environment and the specific characteristics of phenotypes that should be taken into account for the prediction of tumor evolution, morphology and effective treatment.


Assuntos
Proliferação de Células , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Humanos , Modelos Biológicos , Invasividade Neoplásica
15.
IEEE J Biomed Health Inform ; 18(3): 824-31, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24808225

RESUMO

Significant Virtual Physiological Human efforts and projects have been concerned with cancer modeling, especially in the European Commission Seventh Framework research program, with the ambitious goal to approach personalized cancer simulation based on patient-specific data and thereby optimize therapy decisions in the clinical setting. However, building realistic in silico predictive models targeting the clinical practice requires interactive, synergetic approaches to integrate the currently fragmented efforts emanating from the systems biology and computational oncology communities all around the globe. To further this goal, we propose an intelligent graphical workflow planning system that exploits the multiscale and modular nature of cancer and allows building complex cancer models by intuitively linking/interchanging highly specialized models. The system adopts and extends current standardization efforts, key tools, and infrastructure in view of building a pool of reliable and reproducible models capable of improving current therapies and demonstrating the potential for clinical translation of these technologies.


Assuntos
Simulação por Computador , Internet , Modelos Biológicos , Neoplasias , Software , Biologia de Sistemas/métodos , Humanos , Medicina de Precisão , Transdução de Sinais , Interface Usuário-Computador
16.
Artigo em Inglês | MEDLINE | ID: mdl-24110990

RESUMO

During the last decades, especially via the EU initiative related to the Virtual Physiological Human, significant progress has been made in advancing "in-silico" computational models to produce accurate and reliable tumor growth simulations. However, currently most attempts to validate the outcome of the models are either done in-vitro or ex-vivo after tumor resection. In this work, we incorporate information provided by fluorescence molecular tomography performed in-vivo into a mathematical model that describes tumor growth. The outcome is validated against tumor evolution snapshots captured in-vivo using advanced molecular probes in laboratory animals. The simulations are inline with the actual in-vivo growth and although alternative modeling parameters can lead to similar results challenging for additional microscopic information and imaging modalities to drive the in-silico models, they all show that hypoxia plays a dominant role in the evolution of the tumor under study.


Assuntos
Simulação por Computador , Imagem Molecular/métodos , Neoplasias/patologia , Animais , Proliferação de Células , Diagnóstico por Imagem , Modelos Animais de Doenças , Fluorescência , Células HeLa , Humanos , Camundongos , Reprodutibilidade dos Testes
17.
BMC Syst Biol ; 5: 167, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-22008379

RESUMO

BACKGROUND: Metabolic interactions involve the exchange of metabolic products among microbial species. Most microbes live in communities and usually rely on metabolic interactions to increase their supply for nutrients and better exploit a given environment. Constraint-based models have successfully analyzed cellular metabolism and described genotype-phenotype relations. However, there are only a few studies of genome-scale multi-species interactions. Based on genome-scale approaches, we present a graph-theoretic approach together with a metabolic model in order to explore the metabolic variability among bacterial strains and identify and describe metabolically interacting strain communities in a batch culture consisting of two or more strains. We demonstrate the applicability of our approach to the bacterium E. coli across different single-carbon-source conditions. RESULTS: A different diversity graph is constructed for each growth condition. The graph-theoretic properties of the constructed graphs reflect the inherent high metabolic redundancy of the cell to single-gene knockouts, reveal mutant-hubs of unique metabolic capabilities regarding by-production, demonstrate consistent metabolic behaviors across conditions and show an evolutionary difficulty towards the establishment of polymorphism, while suggesting that communities consisting of strains specifically adapted to a given condition are more likely to evolve. We reveal several strain communities of improved growth relative to corresponding monocultures, even though strain communities are not modeled to operate towards a collective goal, such as the community growth and we identify the range of metabolites that are exchanged in these batch co-cultures. CONCLUSIONS: This study provides a genome-scale description of the metabolic variability regarding by-production among E. coli strains under different conditions and shows how metabolic differences can be used to identify metabolically interacting strain communities. This work also extends the existing stoichiometric models in order to describe batch co-cultures and provides the extent of metabolic interactions in a strain community revealing their importance for growth.


Assuntos
Escherichia coli/metabolismo , Redes e Vias Metabólicas , Escherichia coli/genética , Escherichia coli/crescimento & desenvolvimento , Técnicas de Inativação de Genes , Genoma Bacteriano , Interações Microbianas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA