Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Bull Math Biol ; 86(8): 89, 2024 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-38884815

RESUMEN

Cancer metastasis accounts for a majority of cancer-related deaths worldwide. Metastasis occurs when the primary tumor sheds cells into the blood and lymphatic circulation, thereby becoming circulating tumor cells (CTCs) that transverse through the circulatory system, extravasate the circulation and establish a secondary distant tumor. Accumulating evidence suggests that circulating effector CD 8 + T cells are able to recognize and attack arrested or extravasating CTCs, but this important antitumoral effect remains largely undefined. Recent studies highlighted the supporting role of activated platelets in CTCs's extravasation from the bloodstream, contributing to metastatic progression. In this work, a simple mathematical model describes how the primary tumor, CTCs, activated platelets and effector CD 8 + T cells participate in metastasis. The stability analysis reveals that for early dissemination of CTCs, effector CD 8 + T cells can present or keep secondary metastatic tumor burden at low equilibrium state. In contrast, for late dissemination of CTCs, effector CD 8 + T cells are unlikely to inhibit secondary tumor growth. Moreover, global sensitivity analysis demonstrates that the rate of the primary tumor growth, intravascular CTC proliferation, as well as the CD 8 + T cell proliferation, strongly affects the number of the secondary tumor cells. Additionally, model simulations indicate that an increase in CTC proliferation greatly contributes to tumor metastasis. Our simulations further illustrate that the higher the number of activated platelets on CTCs, the higher the probability of secondary tumor establishment. Intriguingly, from a mathematical immunology perspective, our simulations indicate that if the rate of effector CD 8 + T cell proliferation is high, then the secondary tumor formation can be considerably delayed, providing a window for adjuvant tumor control strategies. Collectively, our results suggest that the earlier the effector CD 8 + T cell response is enhanced the higher is the probability of preventing or delaying secondary tumor metastases.


Asunto(s)
Plaquetas , Linfocitos T CD8-positivos , Conceptos Matemáticos , Modelos Inmunológicos , Metástasis de la Neoplasia , Células Neoplásicas Circulantes , Células Neoplásicas Circulantes/patología , Células Neoplásicas Circulantes/inmunología , Humanos , Plaquetas/inmunología , Plaquetas/patología , Metástasis de la Neoplasia/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias/inmunología , Neoplasias/patología , Simulación por Computador , Activación Plaquetaria/inmunología
2.
Bull Math Biol ; 83(7): 75, 2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-34008149

RESUMEN

In this paper, we investigate how natural killer (NK) cell recruitment to the tumor microenvironment (TME) affects oncolytic virotherapy. NK cells play a major role against viral infections. They are, however, known to induce early viral clearance of oncolytic viruses, which hinders the overall efficacy of oncolytic virotherapy. Here, we formulate and analyze a simple mathematical model of the dynamics of the tumor, OV and NK cells using currently available preclinical information. The aim of this study is to characterize conditions under which the synergistic balance between OV-induced NK responses and required viral cytopathicity may or may not result in a successful treatment. In this study, we found that NK cell recruitment to the TME must take place neither too early nor too late in the course of OV infection so that treatment will be successful. NK cell responses are most influential at either early (partly because of rapid response of NK cells to viral infections or antigens) or later (partly because of antitumoral ability of NK cells) stages of oncolytic virotherapy. The model also predicts that: (a) an NK cell response augments oncolytic virotherapy only if viral cytopathicity is weak; (b) the recruitment of NK cells modulates tumor growth; and (c) the depletion of activated NK cells within the TME enhances the probability of tumor escape in oncolytic virotherapy. Taken together, our model results demonstrate that OV infection is crucial, not just to cytoreduce tumor burden, but also to induce the stronger NK cell response necessary to achieve complete or at least partial tumor remission. Furthermore, our modeling framework supports combination therapies involving NK cells and OV which are currently used in oncolytic immunovirotherapy to treat several cancer types.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Humanos , Células Asesinas Naturales , Conceptos Matemáticos , Modelos Teóricos , Neoplasias/terapia , Microambiente Tumoral
3.
J Theor Biol ; 404: 312-330, 2016 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-27317864

RESUMEN

We present a novel mathematical model involving various immune cell populations and tumor cell populations. The model describes how tumor cells evolve and survive the brief encounter with the immune system mediated by natural killer (NK) cells and the activated CD8(+) cytotoxic T lymphocytes (CTLs). The model is composed of ordinary differential equations describing the interactions between these important immune lymphocytes and various tumor cell populations. Based on up-to-date knowledge of immune evasion and rational considerations, the model is designed to illustrate how tumors evade both arms of host immunity (i.e. innate and adaptive immunity). The model predicts that (a) an influx of an external source of NK cells might play a crucial role in enhancing NK-cell immune surveillance; (b) the host immune system alone is not fully effective against progression of tumor cells; (c) the development of immunoresistance by tumor cells is inevitable in tumor immune surveillance. Our model also supports the importance of infiltrating NK cells in tumor immune surveillance, which can be enhanced by NK cell-based immunotherapeutic approaches.


Asunto(s)
Modelos Inmunológicos , Neoplasias/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Comunicación Celular , Simulación por Computador , Citotoxicidad Inmunológica , Humanos , Evasión Inmune , Inmunoterapia , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Neoplasias/patología
4.
Methods ; 66(2): 139-52, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23994873

RESUMEN

The overexpression of certain membrane-bound receptors is a hallmark of cancer progression and it has been suggested to affect the organization, activation, recycling and down-regulation of receptor-ligand complexes in human cancer cells. Thus, comparing receptor trafficking pathways in normal vs. cancer cells requires the ability to image cells expressing dramatically different receptor expression levels. Here, we have presented a significant technical advance to the analysis and processing of images collected using intensity based Förster resonance energy transfer (FRET) confocal microscopy. An automated Image J macro was developed to select region of interests (ROI) based on intensity and statistical-based thresholds within cellular images with reduced FRET signal. Furthermore, SSMD (strictly standardized mean differences), a statistical signal-to-noise ratio (SNR) evaluation parameter, was used to validate the quality of FRET analysis, in particular of ROI database selection. The Image J ROI selection macro together with SSMD as an evaluation parameter of SNR levels, were used to investigate the endocytic recycling of Tfn-TFR complexes at nanometer range resolution in human normal vs. breast cancer cells expressing significantly different levels of endogenous TFR. Here, the FRET-based assay demonstrates that Tfn-TFR complexes in normal epithelial vs. breast cancer cells show a significantly different E% behavior during their endocytic recycling pathway. Since E% is a relative measure of distance, we propose that these changes in E% levels represent conformational changes in Tfn-TFR complexes during endocytic pathway. Thus, our results indicate that Tfn-TFR complexes undergo different conformational changes in normal vs. cancer cells, indicating that the organization of Tfn-TFR complexes at the nanometer range is significantly altered during the endocytic recycling pathway in cancer cells. In summary, improvements in the automated selection of FRET ROI datasets allowed us to detect significant changes in E% with potential biological significance in human normal vs. cancer cells.


Asunto(s)
Endocitosis , Procesamiento de Imagen Asistido por Computador , Transferrinas/metabolismo , Animales , Línea Celular Tumoral , Perros , Transferencia Resonante de Energía de Fluorescencia , Colorantes Fluorescentes/química , Humanos , Células de Riñón Canino Madin Darby , Microscopía Fluorescente , Transporte de Proteínas , Programas Informáticos
5.
iScience ; 26(7): 107229, 2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37519903

RESUMEN

Genomics and proteomics have been central to identify tumor cell populations, but more accurate approaches to classify cell subtypes are still lacking. We propose a new methodology to accurately classify cancer cells based on their organelle spatial topology. Herein, we developed an organelle topology-based cell classification pipeline (OTCCP), which integrates artificial intelligence (AI) and imaging quantification to analyze organelle spatial distribution and inter-organelle topology. OTCCP was used to classify a panel of human breast cancer cells, grown as 2D monolayer or 3D tumor spheroids using early endosomes, mitochondria, and their inter-organelle contacts. Organelle topology allows for a highly precise differentiation between cell lines of different subtypes and aggressiveness. These findings lay the groundwork for using organelle topological profiling as a fast and efficient method for phenotyping breast cancer function as well as a discovery tool to advance our understanding of cancer cell biology at the subcellular level.

6.
Math Biosci Eng ; 19(5): 4429-4457, 2022 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-35430822

RESUMEN

Combining chimeric antigen receptor T (CAR-T) cells with oncolytic viruses (OVs) has recently emerged as a promising treatment approach in preclinical studies that aim to alleviate some of the barriers faced by CAR-T cell therapy. In this study, we address by means of mathematical modeling the main question of whether a single dose or multiple sequential doses of CAR-T cells during the OVs therapy can have a synergetic effect on tumor reduction. To that end, we propose an ordinary differential equations-based model with virus-induced synergism to investigate potential effects of different regimes that could result in efficacious combination therapy against tumor cell populations. Model simulations show that, while the treatment with a single dose of CAR-T cells is inadequate to eliminate all tumor cells, combining the same dose with a single dose of OVs can successfully eliminate the tumor in the absence of virus-induced synergism. However, in the presence of virus-induced synergism, the same combination therapy fails to eliminate the tumor. Furthermore, it is shown that if the intensity of virus-induced synergy and/or virus oncolytic potency is high, then the induced CAR-T cell response can inhibit virus oncolysis. Additionally, the simulations show a more robust synergistic effect on tumor cell reduction when OVs and CAR-T cells are administered simultaneously compared to the combination treatment where CAR-T cells are administered first or after OV injection. Our findings suggest that the combination therapy of CAR-T cells and OVs seems unlikely to be effective if the virus-induced synergistic effects are included when genetically engineering oncolytic viral vectors.


Asunto(s)
Neoplasias , Viroterapia Oncolítica , Virus Oncolíticos , Receptores Quiméricos de Antígenos , Humanos , Modelos Teóricos , Neoplasias/terapia , Virus Oncolíticos/fisiología , Linfocitos T
7.
Bull Math Biol ; 73(12): 2865-87, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21814880

RESUMEN

In this paper, we present a mathematical model predicting the fraction of proliferating cells in G1, S, and G2/M phases of the cell cycle as a function of EGFR and HER2. We show that it is possible to find parameters for the mathematical model so that its predictions agree with the experimental observations that HER2 over-expression results in: (1) a shorter G1-phase and early S-phase entry; (2) and that with a 1-to-1 ration between EGFR and HER2, the growth advantage in HER2 over-expressing cells is indeed associated with the increase of the HER2 expression level.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Ciclo Celular/genética , Genes erbB-2 , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Expresión Génica , Humanos , Conceptos Matemáticos , Modelos Biológicos , Receptor ErbB-2/metabolismo
8.
J Biol Dyn ; 14(1): 454-478, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32589121

RESUMEN

In this paper, we apply a new approach to a special class of discrete time evolution models and establish a solid mathematical foundation to analyse them. We propose new single and multi-species evolutionary competition models using the evolutionary game theory that require a more advanced mathematical theory to handle effectively. A key feature of this new approach is to consider the discrete models as non-autonomous difference equations. Using the powerful tools and results developed in our recent work [E. D'Aniello and S. Elaydi, The structure of ω-limit sets of asymptotically non-autonomous discrete dynamical systems, Discr. Contin. Dyn. Series B. 2019 (to appear).], we embed the non-autonomous difference equations in an autonomous discrete dynamical systems in a higher dimension space, which is the product space of the phase space and the space of the functions defining the non-autonomous system. Our current approach applies to two scenarios. In the first scenario, we assume that the trait equations are decoupled from the equations of the populations. This requires specialized biological and ecological assumptions which we clearly state. In the second scenario, we do not assume decoupling, but rather we assume that the dynamics of the trait is known, such as approaching a positive stable equilibrium point which may apply to a much broader evolutionary dynamics.


Asunto(s)
Evolución Biológica , Modelos Biológicos , Animales , Conducta Competitiva , Conducta Predatoria/fisiología
9.
Sci Rep ; 10(1): 425, 2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31949228

RESUMEN

Mesenchymal stem cells (MSCs) loaded with oncolytic viruses are presently being investigated as a new modality of advanced/metastatic tumors treatment and enhancement of virotherapy. MSCs can, however, either promote or suppress tumor growth. To address the critical question of how MSCs loaded with oncolytic viruses affect virotherapy outcomes and tumor growth patterns in a tumor microenvironment, we developed and analyzed an integrated mathematical-experimental model. We used the model to describe both the growth dynamics in our experiments of firefly luciferase-expressing Hep3B tumor xenografts and the effects of the immune response during the MSCs-based virotherapy. We further employed it to explore the conceptual clinical feasibility, particularly, in evaluating the relative significance of potential immune promotive/suppressive mechanisms induced by MSCs loaded with oncolytic viruses. We were able to delineate conditions which may significantly contribute to the success or failure of MSC-based virotherapy as well as generate new hypotheses. In fact, one of the most impactful outcomes shown by this investigation, not inferred from the experiments alone, was the initially counter-intuitive fact that using tumor-promoting MSCs as carriers is not only helpful but necessary in achieving tumor control. Considering the fact that it is still currently a controversial debate whether MSCs exert a pro- or anti-tumor action, mathematical models such as this one help to quantitatively predict the consequences of using MSCs for delivering virotherapeutic agents in vivo. Taken together, our results show that MSC-mediated systemic delivery of oncolytic viruses is a promising strategy for achieving synergistic anti-tumor efficacy with improved safety profiles.


Asunto(s)
Adenoviridae/fisiología , Células Madre Mesenquimatosas/metabolismo , Modelos Biológicos , Viroterapia Oncolítica/métodos , Virus Oncolíticos/fisiología , Adenoviridae/metabolismo , Proliferación Celular , Virus Oncolíticos/metabolismo
10.
Bull Math Biol ; 70(6): 1707-29, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18648887

RESUMEN

We present a mathematical model to study the effects of HER2 over-expression on cell proliferation in breast cancer. The model illustrates the proliferative behavior of cells as a function of HER2 and EGFR receptors numbers, and the growth factor EGF. This mathematical model comprises kinetic equations describing the cell surface binding of EGF growth factor to EGFR and HER2 receptors, coupled to a model for the dependence of cell proliferation rate on growth factor receptors binding. The simulation results from this model predict: (1) a growth advantage associated with excess HER2 receptors; (2) that HER2-over-expression is an insufficient parameter to predict the proliferation response of cancer cells to epidermal growth factors; and (3) the EGFR receptor expression level in HER2-over-expressing cells plays a key role in mediating the proliferation response to receptor-ligand signaling. This mathematical model also elucidates the interaction and roles of other model parameters in determining cell proliferation rate of HER2-over-expressing cells.


Asunto(s)
Neoplasias de la Mama/patología , Proliferación Celular , Modelos Biológicos , Receptor ErbB-2/genética , Algoritmos , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Simulación por Computador , Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Cinética , Receptor ErbB-2/metabolismo
11.
Math Biosci Eng ; 15(6): 1435-1463, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30418793

RESUMEN

Oncolytic virotherapy has been emerging as a promising novel cancer treatment which may be further combined with the existing therapeutic modalities to enhance their effects. To investigate how virotherapy could enhance chemotherapy, we propose an ODE based mathematical model describing the interactions between tumour cells, the immune response, and a treatment combination with chemotherapy and oncolytic viruses. Stability analysis of the model with constant chemotherapy treatment rates shows that without any form of treatment, a tumour would grow to its maximum size. It also demonstrates that chemotherapy alone is capable of clearing tumour cells provided that the drug efficacy is greater than the intrinsic tumour growth rate. Furthermore, virotherapy alone may not be able to clear tumour cells from body tissue but would rather enhance chemotherapy if viruses with high viral potency are used. To assess the combined effect of virotherapy and chemotherapy we use the forward sensitivity index to perform a sensitivity analysis, with respect to chemotherapy key parameters, of the virus basic reproductive number and the tumour endemic equilibrium. The results from this sensitivity analysis indicate the existence of a critical dose of chemotherapy above which no further significant reduction in the tumour population can be observed. Numerical simulations show that a successful combinational therapy of the chemotherapeutic drugs and viruses depends mostly on the virus burst size, infection rate, and the amount of drugs supplied. Optimal control analysis was performed, by means of the Pontryagin's maximum principle, to further refine predictions of the model with constant treatment rates by accounting for the treatment costs and sides effects. Results from this analysis suggest that the optimal drug and virus combination correspond to half their maximum tolerated doses. This is in agreement with the results from stability and sensitivity analyses.


Asunto(s)
Modelos Biológicos , Neoplasias/terapia , Viroterapia Oncolítica , Animales , Terapia Combinada/métodos , Terapia Combinada/estadística & datos numéricos , Simulación por Computador , Humanos , Conceptos Matemáticos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Viroterapia Oncolítica/estadística & datos numéricos
12.
J Biol Dyn ; 11(1): 244-274, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28537127

RESUMEN

Chemovirotherapy is a combination therapy with chemotherapy and oncolytic viruses. It is gaining more interest and attracting more attention in the clinical setting due to its effective therapy and potential synergistic interactions against cancer. In this paper, we develop and analyse a mathematical model in the form of parabolic non-linear partial differential equations to investigate the spatiotemporal dynamics of tumour cells under chemovirotherapy treatment. The proposed model consists of uninfected and infected tumour cells, a free virus, and a chemotherapeutic drug. The analysis of the model is carried out for both the temporal and spatiotemporal cases. Travelling wave solutions to the spatiotemporal model are used to determine the minimum wave speed of tumour invasion. A sensitivity analysis is performed on the model parameters to establish the key parameters that promote cancer remission during chemovirotherapy treatment. Model analysis of the temporal model suggests that virus burst size and virus infection rate determine the success of the virotherapy treatment, whereas travelling wave solutions to the spatiotemporal model show that tumour diffusivity and growth rate are critical during chemovirotherapy. Simulation results reveal that chemovirotherapy is more effective and a good alternative to either chemotherapy or virotherapy, which is in agreement with the recent experimental studies.


Asunto(s)
Modelos Biológicos , Neoplasias/terapia , Viroterapia Oncolítica , Proliferación Celular , Simulación por Computador , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Factores de Tiempo , Incertidumbre
13.
PLoS One ; 12(9): e0184347, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28934210

RESUMEN

In the present paper, we address by means of mathematical modeling the following main question: How can oncolytic virus infection of some normal cells in the vicinity of tumor cells enhance oncolytic virotherapy? We formulate a mathematical model describing the interactions between the oncolytic virus, the tumor cells, the normal cells, and the antitumoral and antiviral immune responses. The model consists of a system of delay differential equations with one (discrete) delay. We derive the model's basic reproductive number within tumor and normal cell populations and use their ratio as a metric for virus tumor-specificity. Numerical simulations are performed for different values of the basic reproduction numbers and their ratios to investigate potential trade-offs between tumor reduction and normal cells losses. A fundamental feature unravelled by the model simulations is its great sensitivity to parameters that account for most variation in the early or late stages of oncolytic virotherapy. From a clinical point of view, our findings indicate that designing an oncolytic virus that is not 100% tumor-specific can increase virus particles, which in turn, can further infect tumor cells. Moreover, our findings indicate that when infected tissues can be regenerated, oncolytic viral infection of normal cells could improve cancer treatment.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Algoritmos , Simulación por Computador , Humanos , Modelos Biológicos , Neoplasias/inmunología , Neoplasias/virología
14.
Math Biosci Eng ; 10(3): 821-42, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23906151

RESUMEN

Biofilms are present in all natural, medical and industrial surroundings where bacteria live. Biofilm formation is a key factor in the growth and transport of both beneficial and harmful bacteria. While much is known about the later stages of biofilm formation, less is known about its initiation which is an important first step in the biofilm formation. In this paper, we develop a non-linear system of partial differential equations of Keller-Segel type model in one-dimensional space, which couples the dynamics of bacterial movement to that of the sensing molecules. In this case, bacteria perform a biased random walk towards the sensing molecules. We derive the boundary conditions of the adhesion of bacteria to a surface using zero-Dirichlet boundary conditions, while the equation describing sensing molecules at the interface needed particular conditions to be set. The numerical results show the profile of bacteria within the space and the time evolution of the density within the free-space and on the surface. Testing different parameter values indicate that significant amount of sensing molecules present on the surface leads to a faster bacterial movement toward the surface which is the first step of biofilm initiation. Our work gives rise to results that agree with the biological description of the early stages of biofilm formation.


Asunto(s)
Adhesión Bacteriana/fisiología , Biopelículas/crecimiento & desarrollo , Modelos Biológicos , Conceptos Matemáticos , Dinámicas no Lineales , Dinámica Poblacional , Percepción de Quorum/fisiología , Biología de Sistemas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA