Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Bull Math Biol ; 86(8): 89, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884815

ABSTRACT

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.


Subject(s)
Blood Platelets , CD8-Positive T-Lymphocytes , Mathematical Concepts , Models, Immunological , Neoplasm Metastasis , Neoplastic Cells, Circulating , Neoplastic Cells, Circulating/pathology , Neoplastic Cells, Circulating/immunology , Humans , Blood Platelets/immunology , Blood Platelets/pathology , Neoplasm Metastasis/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/pathology , Computer Simulation , Platelet Activation/immunology
2.
J Theor Biol ; 546: 111159, 2022 08 07.
Article in English | MEDLINE | ID: mdl-35577102

ABSTRACT

Increasingly-sophisticated parameter-sensitivity analysis techniques continue to be developed, and each technique comes with its own set of advantages and disadvantages. Selecting which parameter-sensitivity method to use for a particular model, however, is not a straightforward task. In this work, we present a multi-method framework that incorporates three global sensitivity analysis methods: two variance-based methods and one derivative-based method. The two variance-based methods are Sobol's method and MeFAST. The derivative-based method is known as DGSM (Derivative-based Global Sensitivity Measures). MeFAST (Multi test eFAST) is a new parameter sensitivity analysis implementation we built upon the eFAST (Extended Fourier Amplitude Sensitivity Test) algorithm. The improvements incorporated into MeFAST address some important aspects of prior eFAST implementations. We present an intuitive description of each implemented algorithm along with MATLAB codes and a guide to tuning algorithm hyper-parameters for better efficiency. We demonstrate the full methodology and workflow using two example mathematical models of different complexity: the first is a model of HIV disease progression and the second is a model of tumor growth. The computational framework we provide generates graphics for visualizing and comparing the results of all three sensitivity analysis algorithms (DGSM, Sobol, and MeFAST). This algorithm output comparison tool allows one to make a more informed decision when assessing which parameters most importantly influence model outcomes.


Subject(s)
Algorithms , Models, Theoretical , Computer Simulation
3.
Bull Math Biol ; 83(7): 75, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34008149

ABSTRACT

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.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Killer Cells, Natural , Mathematical Concepts , Models, Theoretical , Neoplasms/therapy , Tumor Microenvironment
4.
Sci Rep ; 10(1): 425, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949228

ABSTRACT

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.


Subject(s)
Adenoviridae/physiology , Mesenchymal Stem Cells/metabolism , Models, Biological , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , Adenoviridae/metabolism , Cell Proliferation , Oncolytic Viruses/metabolism
5.
Front Physiol ; 10: 1107, 2019.
Article in English | MEDLINE | ID: mdl-31555144

ABSTRACT

Type I diabetes (T1D) is an autoimmune disease that can be managed, but for which there is currently no cure. Recent discoveries, particularly in mouse models, indicate that targeted modulation of the immune response has the potential to move an individual from a diabetic to a long-term, if not permanent, healthy state. In this paper we develop a single compartment mathematical model that captures the dynamics of dendritic cells (DC and tDC), T cells (effector and regulatory), and macrophages in the development of type I diabetes. The model supports the hypothesis that differences in macrophage clearance rates play a significant role in determining whether or not an individual is likely to become diabetic subsequent to a significant immune challenge. With this model we are able to explore the effects of strengthening the anti-inflammatory component of the immune system in a vulnerable individual. Simulations indicate that there are windows of opportunity in which treatment intervention is more likely to be beneficial in protecting an individual from entering a diabetic state. This model framework can be used as a foundation for modeling future T1D treatments as they are developed.

6.
PLoS One ; 12(9): e0184347, 2017.
Article in English | MEDLINE | ID: mdl-28934210

ABSTRACT

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.


Subject(s)
Neoplasms/therapy , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Algorithms , Computer Simulation , Humans , Models, Biological , Neoplasms/immunology , Neoplasms/virology
7.
Methods Mol Biol ; 929: 51-74, 2012.
Article in English | MEDLINE | ID: mdl-23007426

ABSTRACT

Mathematical modeling is a vehicle that allows for explanation and prediction of natural phenomena. In this chapter we present guidelines and best practices for developing and implementing mathematical models, using cancer growth, chemotherapy, and immunotherapy modeling as examples.


Subject(s)
Models, Theoretical , Animals , Drug Therapy , Humans , Immunotherapy , Neoplasms/drug therapy , Neoplasms/therapy
9.
CBE Life Sci Educ ; 9(3): 165-71, 2010.
Article in English | MEDLINE | ID: mdl-20810946

ABSTRACT

In response to the call of BIO2010 for integrating quantitative skills into undergraduate biology education, 30 Howard Hughes Medical Institute (HHMI) Program Directors at the 2006 HHMI Program Directors Meeting established a consortium to investigate, implement, develop, and disseminate best practices resulting from the integration of math and biology. With the assistance of an HHMI-funded mini-grant, led by Karl Joplin of East Tennessee State University, and support in institutional HHMI grants at Emory and University of Delaware, these institutions held a series of summer institutes and workshops to document progress toward and address the challenges of implementing a more quantitative approach to undergraduate biology education. This report summarizes the results of the four summer institutes (2007-2010). The group developed four draft white papers, a wiki site, and a listserv. One major outcome of these meetings is this issue of CBE-Life Sciences Education, which resulted from proposals at our 2008 meeting and a January 2009 planning session. Many of the papers in this issue emerged from or were influenced by these meetings.


Subject(s)
Biology/education , Mathematics/education , Congresses as Topic , Periodicals as Topic , Universities
10.
CBE Life Sci Educ ; 9(3): 316-22, 2010.
Article in English | MEDLINE | ID: mdl-20810964

ABSTRACT

The success of interdisciplinary research teams depends largely upon skills related to team performance. We evaluated student and team performance for undergraduate biology and mathematics students who participated in summer research projects conducted in off-campus laboratories. The student teams were composed of a student with a mathematics background and an experimentally oriented biology student. The team mentors typically ranked the students' performance very good to excellent over a range of attributes that included creativity and ability to conduct independent research. However, the research teams experienced problems meeting prespecified deadlines due to poor time and project management skills. Because time and project management skills can be readily taught and moreover typically reflect good research practices, simple modifications should be made to undergraduate curricula so that the promise of initiatives, such as MATH-BIO 2010, can be implemented.


Subject(s)
Biology/education , Cooperative Behavior , Mathematics/education , Research/education , Teaching/organization & administration , Educational Measurement , Program Evaluation , Students
11.
Cancer Res ; 65(17): 7950-8, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16140967

ABSTRACT

Mathematical models of tumor-immune interactions provide an analytic framework in which to address specific questions about tumor-immune dynamics. We present a new mathematical model that describes tumor-immune interactions, focusing on the role of natural killer (NK) and CD8+ T cells in tumor surveillance, with the goal of understanding the dynamics of immune-mediated tumor rejection. The model describes tumor-immune cell interactions using a system of differential equations. The functions describing tumor-immune growth, response, and interaction rates, as well as associated variables, are developed using a least-squares method combined with a numerical differential equations solver. Parameter estimates and model validations use data from published mouse and human studies. Specifically, CD8+ T-tumor and NK-tumor lysis data from chromium release assays as well as in vivo tumor growth data are used. A variable sensitivity analysis is done on the model. The new functional forms developed show that there is a clear distinction between the dynamics of NK and CD8+ T cells. Simulations of tumor growth using different levels of immune stimulating ligands, effector cells, and tumor challenge are able to reproduce data from the published studies. A sensitivity analysis reveals that the variable to which the model is most sensitive is patient specific, and can be measured with a chromium release assay. The variable sensitivity analysis suggests that the model can predict which patients may positively respond to treatment. Computer simulations highlight the importance of CD8+ T-cell activation in cancer therapy.


Subject(s)
Models, Immunological , Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Growth Processes/immunology , Humans , Immunity, Cellular/immunology , Killer Cells, Natural/immunology , Mice , Neoplasms/therapy , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...