1
|
Mongeon B, Hébert-Doutreloux J, Surendran A, Karimi E, Fiset B, Quail DF, Walsh LA, Jenner AL, Craig M. Spatial computational modelling illuminates the role of the tumour microenvironment for treating glioblastoma with immunotherapies. NPJ Syst Biol Appl 2024; 10:91. [PMID: 39155294 PMCID: PMC11330976 DOI: 10.1038/s41540-024-00419-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Glioblastoma is the most common and deadliest brain tumour in adults, with a median survival of 15 months under the current standard of care. Immunotherapies like immune checkpoint inhibitors and oncolytic viruses have been extensively studied to improve this endpoint. However, most thus far have failed. To improve the efficacy of immunotherapies to treat glioblastoma, new single-cell imaging modalities like imaging mass cytometry can be leveraged and integrated with computational models. This enables a better understanding of the tumour microenvironment and its role in treatment success or failure in this hard-to-treat tumour. Here, we implemented an agent-based model that allows for spatial predictions of combination chemotherapy, oncolytic virus, and immune checkpoint inhibitors against glioblastoma. We initialised our model with patient imaging mass cytometry data to predict patient-specific responses and found that oncolytic viruses drive combination treatment responses determined by intratumoral cell density. We found that tumours with higher tumour cell density responded better to treatment. When fixing the number of cancer cells, treatment efficacy was shown to be a function of CD4 + T cell and, to a lesser extent, of macrophage counts. Critically, our simulations show that care must be put into the integration of spatial data and agent-based models to effectively capture intratumoral dynamics. Together, this study emphasizes the use of predictive spatial modelling to better understand cancer immunotherapy treatment dynamics, while highlighting key factors to consider during model design and implementation.
Collapse
Affiliation(s)
- Blanche Mongeon
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
| | | | - Anudeep Surendran
- Center for Advanced Systems Understanding, Görlitz, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Adrianne L Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Morgan Craig
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada.
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
2
|
Mohammad Mirzaei N, Shahriyari L. Modeling cancer progression: an integrated workflow extending data-driven kinetic models to bio-mechanical PDE models. Phys Biol 2024; 21:022001. [PMID: 38330444 DOI: 10.1088/1478-3975/ad2777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/08/2024] [Indexed: 02/10/2024]
Abstract
Computational modeling of cancer can help unveil dynamics and interactions that are hard to replicate experimentally. Thanks to the advancement in cancer databases and data analysis technologies, these models have become more robust than ever. There are many mathematical models which investigate cancer through different approaches, from sub-cellular to tissue scale, and from treatment to diagnostic points of view. In this study, we lay out a step-by-step methodology for a data-driven mechanistic model of the tumor microenvironment. We discuss data acquisition strategies, data preparation, parameter estimation, and sensitivity analysis techniques. Furthermore, we propose a possible approach to extend mechanistic ordinary differential equation models to PDE models coupled with mechanical growth. The workflow discussed in this article can help understand the complex temporal and spatial interactions between cells and cytokines in the tumor microenvironment and their effect on tumor growth.
Collapse
Affiliation(s)
- Navid Mohammad Mirzaei
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, United States of America
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, United States of America
| |
Collapse
|
3
|
Arabameri A, Arab S. Understanding the Interplay of CAR-NK Cells and Triple-Negative Breast Cancer: Insights from Computational Modeling. Bull Math Biol 2024; 86:20. [PMID: 38240892 DOI: 10.1007/s11538-023-01247-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024]
Abstract
Chimeric antigen receptor (CAR)-engineered natural killer (NK) cells have recently emerged as a promising and safe alternative to CAR-T cells for targeting solid tumors. In the case of triple-negative breast cancer (TNBC), traditional cancer treatments and common immunotherapies have shown limited effectiveness. However, CAR-NK cells have been successfully employed to target epidermal growth factor receptor (EGFR) on TNBC cells, thereby enhancing the efficacy of immunotherapy. The effectiveness of CAR-NK-based immunotherapy is influenced by various factors, including the vaccination dose, vaccination pattern, and tumor immunosuppressive factors in the microenvironment. To gain insights into the dynamics and effects of CAR-NK-based immunotherapy, we propose a computational model based on experimental data and immunological theories. This model integrates an individual-based model that describes the interplay between the tumor and the immune system, along with an ordinary differential equation model that captures the variation of inflammatory cytokines. Computational results obtained from the proposed model shed light on the conditions necessary for initiating an effective anti-tumor response. Furthermore, global sensitivity analysis highlights the issue of low persistence of CAR-NK cells in vivo, which poses a significant challenge for the successful clinical application of these cells. Leveraging the model, we identify the optimal vaccination time, vaccination dose, and time interval between injections for maximizing therapeutic outcomes.
Collapse
Affiliation(s)
- Abazar Arabameri
- Department of Electrical Engineering, University of Zanjan, Zanjan, Iran.
| | - Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
4
|
Chemoimmunotherapy Administration Protocol Design for the Treatment of Leukemia through Mathematical Modeling and In Silico Experimentation. Pharmaceutics 2022; 14:pharmaceutics14071396. [PMID: 35890295 PMCID: PMC9316854 DOI: 10.3390/pharmaceutics14071396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/25/2022] [Accepted: 06/27/2022] [Indexed: 02/04/2023] Open
Abstract
Cancer with all its more than 200 variants continues to be a major health problem around the world with nearly 10 million deaths recorded in 2020, and leukemia accounted for more than 300,000 cases according to the Global Cancer Observatory. Although new treatment strategies are currently being developed in several ongoing clinical trials, the high complexity of cancer evolution and its survival mechanisms remain as an open problem that needs to be addressed to further enhanced the application of therapies. In this work, we aim to explore cancer growth, particularly chronic lymphocytic leukemia, under the combined application of CAR-T cells and chlorambucil as a nonlinear dynamical system in the form of first-order Ordinary Differential Equations. Therefore, by means of nonlinear theories, sufficient conditions are established for the eradication of leukemia cells, as well as necessary conditions for the long-term persistence of both CAR-T and cancer cells. Persistence conditions are important in treatment protocol design as these provide a threshold below which the dose will not be enough to produce a cytotoxic effect in the tumour population. In silico experimentations allowed us to design therapy administration protocols to ensure the complete eradication of leukemia cells in the system under study when considering only the infusion of CAR-T cells and for the combined application of chemoimmunotherapy. All results are illustrated through numerical simulations. Further, equations to estimate cytotoxicity of chlorambucil and CAR-T cells to leukemia cancer cells were formulated and thoroughly discussed with a 95% confidence interval for the parameters involved in each formula.
Collapse
|
5
|
Khajanchi S, Nieto JJ. Spatiotemporal dynamics of a glioma immune interaction model. Sci Rep 2021; 11:22385. [PMID: 34789751 PMCID: PMC8599515 DOI: 10.1038/s41598-021-00985-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 10/20/2021] [Indexed: 12/20/2022] Open
Abstract
We report a mathematical model which depicts the spatiotemporal dynamics of glioma cells, macrophages, cytotoxic-T-lymphocytes, immuno-suppressive cytokine TGF-β and immuno-stimulatory cytokine IFN-γ through a system of five coupled reaction-diffusion equations. We performed local stability analysis of the biologically based mathematical model for the growth of glioma cell population and their environment. The presented stability analysis of the model system demonstrates that the temporally stable positive interior steady state remains stable under the small inhomogeneous spatiotemporal perturbations. The irregular spatiotemporal dynamics of gliomas, macrophages and cytotoxic T-lymphocytes are discussed extensively and some numerical simulations are presented. Performed some numerical simulations in both one and two dimensional spaces. The occurrence of heterogeneous pattern formation of the system has both biological and mathematical implications and the concepts of glioma cell progression and invasion are considered. Simulation of the model shows that by increasing the value of time, the glioma cell population, macrophages and cytotoxic-T-lymphocytes spread throughout the domain.
Collapse
Affiliation(s)
- Subhas Khajanchi
- Department of Mathematics, Presidency University, 86/1 College Street, Kolkata, 700073, India.
| | - Juan J Nieto
- Instituto de Matem\acute{a}ticas, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
6
|
Dzwinel W, Kłusek A, Siwik L. Supermodeling in predictive diagnostics of cancer under treatment. Comput Biol Med 2021; 137:104797. [PMID: 34488027 DOI: 10.1016/j.compbiomed.2021.104797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 08/20/2021] [Accepted: 08/21/2021] [Indexed: 11/28/2022]
Abstract
Classical data assimilation (DA) techniques, synchronizing a computer model with observations, are highly demanding computationally, particularly, for complex over-parametrized cancer models. Consequently, current models are not sufficiently flexible to interactively explore various therapy strategies, and to become a key tool of predictive oncology. We show that, by using supermodeling, it is possible to develop a prediction/correction scheme that could attain the required time regimes and be directly used to support decision-making in anticancer therapies. A supermodel is an interconnected ensemble of individual models (sub-models); in this case, the variously parametrized baseline tumor models. The sub-model connection weights are trained from data, thereby incorporating the advantages of the individual models. Simultaneously, by optimizing the strengths of the connections, the sub-models tend to partially synchronize with one another. As a result, during the evolution of the supermodel, the systematic errors of the individual models partially cancel each other. We find that supermodeling allows for a radical increase in the accuracy and efficiency of data assimilation. We demonstrate that it can be considered as a meta-procedure for any classical parameter fitting algorithm, thus it represents the next - latent - level of abstraction of data assimilation. We conclude that supermodeling is a very promising paradigm that can considerably increase the quality of prognosis in predictive oncology.
Collapse
Affiliation(s)
- Witold Dzwinel
- AGH University of Science and Technology, Krakow, Poland.
| | - Adrian Kłusek
- AGH University of Science and Technology, Krakow, Poland.
| | - Leszek Siwik
- AGH University of Science and Technology, Krakow, Poland.
| |
Collapse
|
7
|
Zhang Z, Liu L, Ma C, Cui X, Lam RHW, Chen W. An in silico glioblastoma microenvironment model dissects the immunological mechanisms of resistance to PD-1 checkpoint blockade immunotherapy. SMALL METHODS 2021; 5:2100197. [PMID: 34423116 PMCID: PMC8372235 DOI: 10.1002/smtd.202100197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Indexed: 05/02/2023]
Abstract
The PD-1 immune checkpoint-based therapy has emerged as a promising therapy strategy for treating the malignant brain tumor glioblastoma (GBM). However, patient response varies in clinical trials due in large to the tumor heterogeneity and immunological resistance in the tumor microenvironment. To further understand how mechanistically the niche interplay and competition drive anti-PD-1 resistance, we established an in-silico model to quantitatively describe the biological rationale of critical GBM-immune interactions, such as tumor growth and apoptosis, T cell activation and cytotoxicity, and tumor-associated macrophage (TAM) mediated immunosuppression. Such an in-silico experimentation and predictive model, based on the in vitro microfluidic chip-measured end-point data and patient-specific immunological characteristics, allowed for a comprehensive and dynamic analysis of multiple TAM-associated immunosuppression mechanisms against the anti-PD-1 immunotherapy. Our computational model demonstrated that the TAM-associated immunosuppression varied in severity across different GBM subtypes, which resulted in distinct tumor responses. Our prediction results indicated that a combination therapy co-targeting of PD-1 checkpoint and TAM-associated CSF-1R signaling could enhance the immune responses of GBM patients, especially those patients with mesenchymal GBM who are irresponsive to the single anti-PD-1 therapy. The development of a patient-specific in silico-in vitro GBM model would help navigate and personalize immunotherapies for GBM patients.
Collapse
Affiliation(s)
- Zhuoyu Zhang
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Lunan Liu
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Chao Ma
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Xin Cui
- Department of Biomedical Engineering, Jinan University, Guangzhou, China
| | - Raymond H W Lam
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY
| |
Collapse
|
8
|
Barros LRC, Paixão EA, Valli AMP, Naozuka GT, Fassoni AC, Almeida RC. CART math-A Mathematical Model of CAR-T Immunotherapy in Preclinical Studies of Hematological Cancers. Cancers (Basel) 2021; 13:2941. [PMID: 34208323 PMCID: PMC8231202 DOI: 10.3390/cancers13122941] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has gained great momentum with chimeric antigen receptor T cell (CAR-T) therapy, in which patient's T lymphocytes are genetically manipulated to recognize tumor-specific antigens, increasing tumor elimination efficiency. In recent years, CAR-T cell immunotherapy for hematological malignancies achieved a great response rate in patients and is a very promising therapy for several other malignancies. Each new CAR design requires a preclinical proof-of-concept experiment using immunodeficient mouse models. The absence of a functional immune system in these mice makes them simple and suitable for use as mathematical models. In this work, we develop a three-population mathematical model to describe tumor response to CAR-T cell immunotherapy in immunodeficient mouse models, encompassing interactions between a non-solid tumor and CAR-T cells (effector and long-term memory). We account for several phenomena, such as tumor-induced immunosuppression, memory pool formation, and conversion of memory into effector CAR-T cells in the presence of new tumor cells. Individual donor and tumor specificities are considered uncertainties in the model parameters. Our model is able to reproduce several CAR-T cell immunotherapy scenarios, with different CAR receptors and tumor targets reported in the literature. We found that therapy effectiveness mostly depends on specific parameters such as the differentiation of effector to memory CAR-T cells, CAR-T cytotoxic capacity, tumor growth rate, and tumor-induced immunosuppression. In summary, our model can contribute to reducing and optimizing the number of in vivo experiments with in silico tests to select specific scenarios that could be tested in experimental research. Such an in silico laboratory is an easy-to-run open-source simulator, built on a Shiny R-based platform called CARTmath. It contains the results of this manuscript as examples and documentation. The developed model together with the CARTmath platform have potential use in assessing different CAR-T cell immunotherapy protocols and its associated efficacy, becoming an accessory for in silico trials.
Collapse
Affiliation(s)
- Luciana R. C. Barros
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina ds Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Emanuelle A. Paixão
- Graduate Program, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil; (E.A.P.); (G.T.N.)
| | - Andrea M. P. Valli
- Computer Science Department, Universidade Federal do Espírito Santo, Vitória 29075-910, Brazil;
| | - Gustavo T. Naozuka
- Graduate Program, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil; (E.A.P.); (G.T.N.)
| | - Artur C. Fassoni
- Institute for Mathematics and Computer Science, Universidade Federal de Itajubá, Itajubá 37500-903, Brazil;
| | - Regina C. Almeida
- Computational Modeling Department, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil;
| |
Collapse
|
9
|
Personalized Immunotherapy Treatment Strategies for a Dynamical System of Chronic Myelogenous Leukemia. Cancers (Basel) 2021; 13:cancers13092030. [PMID: 33922302 PMCID: PMC8122842 DOI: 10.3390/cancers13092030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022] Open
Abstract
Simple Summary As computer performance continues to grow at more affordable costs, mathematical modelling and in silico experimentation begin to play a larger role in understanding cancer evolution. The aim of our work is to formulate a control strategy for the Adaptive Cellular Therapy (ACT) that can fully eradicates the Chronic Myelogenous Leukemia (CML) cells population in a mathematical model describing interactions between naive T cells, effector T cells and CML cancer cells in the circulatory blood system. Mathematical analysis and numerical simulations allow us to conclude that it is possible to design a personalized administration protocol for the ACT in the form of a pulse train with asymmetrical waves and a fixed amplitude to achieve complete CML cancer cells eradication. The amplitude of the impulse on which the treatment is applied is given by an arithmetical combination of the parameters of the system with at least a duty cycle of 45 min/day. Abstract This paper is devoted to exploring personalized applications of cellular immunotherapy as a control strategy for the treatment of chronic myelogenous leukemia described by a dynamical system of three first-order ordinary differential equations. The latter was achieved by applying both the Localization of Compact Invariant Sets and Lyapunov’s stability theory. Combination of these two approaches allows us to establish sufficient conditions on the immunotherapy treatment parameter to ensure the complete eradication of the leukemia cancer cells. These conditions are given in terms of the system parameters and by performing several in silico experimentations, we formulated a protocol for the therapy application that completely eradicates the leukemia cancer cells population for different initial tumour concentrations. The formulated protocol does not dangerously increase the effector T cells population. Further, complete eradication is considered when solutions go below a finite critical value below which cancer cells cannot longer persist; i.e., one cancer cell. Numerical simulations are consistent with our analytical results.
Collapse
|
10
|
Butner JD, Wang Z, Elganainy D, Al Feghali KA, Plodinec M, Calin GA, Dogra P, Nizzero S, Ruiz-Ramírez J, Martin GV, Tawbi HA, Chung C, Koay EJ, Welsh JW, Hong DS, Cristini V. A mathematical model for the quantification of a patient's sensitivity to checkpoint inhibitors and long-term tumour burden. Nat Biomed Eng 2021; 5:297-308. [PMID: 33398132 PMCID: PMC8669771 DOI: 10.1038/s41551-020-00662-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 11/14/2020] [Indexed: 02/06/2023]
Abstract
A large proportion of patients with cancer are unresponsive to treatment with immune checkpoint blockade and other immunotherapies. Here, we report a mathematical model of the time course of tumour responses to immune checkpoint inhibitors. The model takes into account intrinsic tumour growth rates, the rates of immune activation and of tumour-immune cell interactions, and the efficacy of immune-mediated tumour killing. For 124 patients, four cancer types and two immunotherapy agents, the model reliably described the immune responses and final tumour burden across all different cancers and drug combinations examined. In validation cohorts from four clinical trials of checkpoint inhibitors (with a total of 177 patients), the model accurately stratified the patients according to reduced or increased long-term tumour burden. We also provide model-derived quantitative measures of treatment sensitivity for specific drug-cancer combinations. The model can be used to predict responses to therapy and to quantify specific drug-cancer sensitivities in individual patients.
Collapse
Affiliation(s)
- Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Dalia Elganainy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karine A Al Feghali
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Basel, Switzerland
| | - George A Calin
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Javier Ruiz-Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA
| | - Geoffrey V Martin
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Tawbi
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eugene J Koay
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David S Hong
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
11
|
BUNIMOVICH-MENDRAZITSKY SVETLANA, SHAIKHET LEONID. STABILITY ANALYSIS OF A MATHEMATICAL MODEL FOR CHRONIC MYELOID LEUKEMIA ERADICATION. J BIOL SYST 2021. [DOI: 10.1142/s0218339021500078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We analyze a mathematical model for the treatment of chronic myeloid leukemia (CML). The model is designed for complete recovery of CML patients after treatment. The model developed in the paper [Bunimovich-Mendrazitsky S, Kronik N, Vainstein V, Optimization of interferon-alpha and imatinib combination therapy for CML: A modeling approach, Adv Theory Simul 2(1):1800081, 2018] introduced a combined treatment of CML based on imatinib therapy and immunotherapy. Immunotherapy based on Interferon alpha-2a (IFN-[Formula: see text]) affects stem and mature cancer cell mortality, and leads to outcome improvements in the combined therapy. The qualitative character of our results shows that additional therapy for the complete cure of CML patients is required. This additional treatment is tumor infiltrating lymphocytes (TIL) along with a combination imatinib and IFN-[Formula: see text] treatment. The model examines the interaction between CML cancer cells and effector cells, using an ODE system. Stability analysis of the model defines conditions when imatinib treatment might lead to the eradication of CML with IFN-[Formula: see text] and TIL. Three equilibria are investigated for the proposed model. Stability conditions for equilibria are formulated in terms of the linear matrix inequalities (LMIs).
Collapse
Affiliation(s)
| | - LEONID SHAIKHET
- Department of Mathematics, Ariel University, Ariel 40700, Israel
| |
Collapse
|
12
|
Stability Analysis of Delayed Tumor-Antigen-ActivatedImmune Response in Combined BCG and IL-2Immunotherapy of Bladder Cancer. Processes (Basel) 2020. [DOI: 10.3390/pr8121564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
We use a system biology approach to translate the interaction of Bacillus Calmette-Gurin (BCG) + interleukin 2 (IL-2) for the treatment of bladder cancer into a mathematical model. The main goal of this research is to predict the outcome of BCG + IL-2 treatment combinations. We examined whether the delay effect caused by the proliferation of tumor antigen-specific effector cells after the immune system destroys BCG-infected urothelium cells after BCG and IL-2 immunotherapy influences success in bladder cancer treatment. To do this, we introduce a system of differential equations where the variables are the main participants in the immune response after BCG installations to fight cancer: the number of tumor cells, BCG cells, immune cells, and cytokines involved in the tumor-immune response. The relevant parameters describing the dynamics of the system are taken from a variety of biological, clinical literature and estimated using the mathematical models. We examine the local stability analysis of non-negative equilibrium states of the model. In theory, treatment could improve system stability, and we analyze the stability of all equilibria using the method of Lyapunov functionals construction and the method of linear matrix inequalities (LMIs). Our results prove that the period for the proliferation of tumor antigen-specific effector cells does not influence to the success of the non-responsive patients after an intensified combined BCG + IL-2 treatment.
Collapse
|
13
|
Osojnik A, Gaffney EA, Davies M, Yates JWT, Byrne HM. Identifying and characterising the impact of excitability in a mathematical model of tumour-immune interactions. J Theor Biol 2020; 501:110250. [PMID: 32199856 DOI: 10.1016/j.jtbi.2020.110250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 02/24/2020] [Accepted: 03/17/2020] [Indexed: 02/07/2023]
Abstract
We study a five-compartment mathematical model originally proposed by Kuznetsov et al. (1994) to investigate the effect of nonlinear interactions between tumour and immune cells in the tumour microenvironment, whereby immune cells may induce tumour cell death, and tumour cells may inactivate immune cells. Exploiting a separation of timescales in the model, we use the method of matched asymptotics to derive a new two-dimensional, long-timescale, approximation of the full model, which differs from the quasi-steady-state approximation introduced by Kuznetsov et al. (1994), but is validated against numerical solutions of the full model. Through a phase-plane analysis, we show that our reduced model is excitable, a feature not traditionally associated with tumour-immune dynamics. Through a systematic parameter sensitivity analysis, we demonstrate that excitability generates complex bifurcating dynamics in the model. These are consistent with a variety of clinically observed phenomena, and suggest that excitability may underpin tumour-immune interactions. The model exhibits the three stages of immunoediting - elimination, equilibrium, and escape, via stable steady states with different tumour cell concentrations. Such heterogeneity in tumour cell numbers can stem from variability in initial conditions and/or model parameters that control the properties of the immune system and its response to the tumour. We identify different biophysical parameter targets that could be manipulated with immunotherapy in order to control tumour size, and we find that preferred strategies may differ between patients depending on the strength of their immune systems, as determined by patient-specific values of associated model parameters.
Collapse
Affiliation(s)
- Ana Osojnik
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Road, Oxford, OX2 6GG, UK.
| | - Eamonn A Gaffney
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Road, Oxford, OX2 6GG, UK
| | - Michael Davies
- DMPK, Early Oncology, Oncology R&D, AstraZeneca, Chesterford Research Park, Little Chesterford, Cambridge, CB10 1XL, UK
| | - James W T Yates
- DMPK, Early Oncology, Oncology R&D, AstraZeneca, Chesterford Research Park, Little Chesterford, Cambridge, CB10 1XL, UK
| | - Helen M Byrne
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Road, Oxford, OX2 6GG, UK
| |
Collapse
|
14
|
Agur Z, Elishmereni M, Foryś U, Kogan Y. Accelerating the Development of Personalized Cancer Immunotherapy by Integrating Molecular Patients' Profiles with Dynamic Mathematical Models. Clin Pharmacol Ther 2020; 108:515-527. [PMID: 32535891 DOI: 10.1002/cpt.1942] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 06/03/2020] [Indexed: 01/08/2023]
Abstract
We review the evolution, achievements, and limitations of the current paradigm shift in medicine, from the "one-size-fits-all" model to "Precision Medicine." Precision, or personalized, medicine-tailoring the medical treatment to the personal characteristics of each patient-engages advanced statistical methods to evaluate the relationships between static patient profiling (e.g., genomic and proteomic), and a simple clinically motivated output (e.g., yes/no responder). Today, precision medicine technologies that have facilitated groundbreaking advances in oncology, notably in cancer immunotherapy, are approaching the limits of their potential, mainly due to the scarcity of methods for integrating genomic, proteomic and clinical patient information. A different approach to treatment personalization involves methodologies focusing on the dynamic interactions in the patient-disease-drug system, as portrayed in mathematical modeling. Achievements of this scientific approach, in the form of algorithms for predicting personal disease dynamics in individual patients under immunotherapeutic drugs, are reviewed as well. The contribution of the dynamic approaches to precision medicine is limited, at present, due to insufficient applicability and validation. Yet, the time is ripe for amalgamating together these two approaches, for maximizing their joint potential to personalize and improve cancer immunotherapy. We suggest the roadmap toward achieving this goal, technologically, and urge clinicians, pharmacologists, and computational biologists to join forces along the pharmaco-clinical track of this development.
Collapse
Affiliation(s)
- Zvia Agur
- Institute for Medical Biomathematics (IMBM), Bene Ataroth, Israel
| | | | - Urszula Foryś
- Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Yuri Kogan
- Institute for Medical Biomathematics (IMBM), Bene Ataroth, Israel
| |
Collapse
|
15
|
Garcia V, Bonhoeffer S, Fu F. Cancer-induced immunosuppression can enable effectiveness of immunotherapy through bistability generation: A mathematical and computational examination. J Theor Biol 2020; 492:110185. [PMID: 32035826 PMCID: PMC7079339 DOI: 10.1016/j.jtbi.2020.110185] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 01/09/2020] [Accepted: 02/03/2020] [Indexed: 12/22/2022]
Abstract
The presence of an immunological barrier in cancer- immune system interaction (CISI) is consistent with the bistability patterns in that system. In CISI models, bistability patterns are consistent with immunosuppressive effects dominating immunoproliferative effects. Bistability could be harnessed to devise effective combination immunotherapy approaches.
Cancer immunotherapies rely on how interactions between cancer and immune system cells are constituted. The more essential to the emergence of the dynamical behavior of cancer growth these interactions are, the more effectively they may be used as mechanisms for interventions. Mathematical modeling can help unearth such connections, and help explain how they shape the dynamics of cancer growth. Here, we explored whether there exist simple, consistent properties of cancer-immune system interaction (CISI) models that might be harnessed to devise effective immunotherapy approaches. We did this for a family of three related models of increasing complexity. To this end, we developed a base model of CISI, which captures some essential features of the more complex models built on it. We find that the base model and its derivates can plausibly reproduce biological behavior that is consistent with the notion of an immunological barrier. This behavior is also in accord with situations in which the suppressive effects exerted by cancer cells on immune cells dominate their proliferative effects. Under these circumstances, the model family may display a pattern of bistability, where two distinct, stable states (a cancer-free, and a full-grown cancer state) are possible. Increasing the effectiveness of immune-caused cancer cell killing may remove the basis for bistability, and abruptly tip the dynamics of the system into a cancer-free state. Additionally, in combination with the administration of immune effector cells, modifications in cancer cell killing may be harnessed for immunotherapy without the need for resolving the bistability. We use these ideas to test immunotherapeutic interventions in silico in a stochastic version of the base model. This bistability-reliant approach to cancer interventions might offer advantages over those that comprise gradual declines in cancer cell numbers.
Collapse
Affiliation(s)
- Victor Garcia
- Institute of Applied Simulation, Zurich University of Applied Sciences, Einsiedlerstrasse 31a, 8820 Wädenswil, Switzerland; ETH Zurich, Universitätstrasse 16, 8092 Zürich, Switzerland; Institute for Social and Preventive Medicine, University of Bern, Finkenhubelweg 11, 3012 Bern, Switzerland; Department of Biology, Stanford University, 371 Serra Mall, Stanford CA 94305, USA.
| | | | - Feng Fu
- Department of Mathematics, Dartmouth College, 27 N. Main Street, 6188 Kemeny Hall, Hanover, NH 03755-3551, USA; ETH Zurich, Universitätstrasse 16, 8092 Zürich, Switzerland
| |
Collapse
|
16
|
Less is more: reducing the number of administered chimeric antigen receptor T cells in a mouse model using a mathematically guided approach. Cancer Immunol Immunother 2020; 69:1165-1175. [DOI: 10.1007/s00262-020-02516-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 02/12/2020] [Indexed: 12/13/2022]
|
17
|
Tsur N, Kogan Y, Rehm M, Agur Z. Response of patients with melanoma to immune checkpoint blockade – insights gleaned from analysis of a new mathematical mechanistic model. J Theor Biol 2020; 485:110033. [DOI: 10.1016/j.jtbi.2019.110033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 07/05/2019] [Accepted: 09/26/2019] [Indexed: 12/30/2022]
|
18
|
KHAJANCHI SUBHAS, BANERJEE SANDIP. A STRATEGY OF OPTIMAL EFFICACY OF T11 TARGET STRUCTURE IN THE TREATMENT OF BRAIN TUMOR. J BIOL SYST 2019. [DOI: 10.1142/s0218339019500104] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
We report a mathematical model depicting gliomas and immune system interactions by considering the role of immunotherapeutic drug T11 target structure (T11TS). The mathematical model comprises a system of coupled nonlinear ordinary differential equations involving glioma cells, macrophages, activated cytotoxic T-lymphocytes (CTLs), immunosuppressive cytokine transforming growth factor-[Formula: see text] (TGF-[Formula: see text]), immunostimulatory cytokine interferon-[Formula: see text] (IFN-[Formula: see text]) and the concentrations of immunotherapeutic agent T11TS. For the better understanding of the circumstances under which the gliomas can be eradicated from a patient, we use optimal control strategy. We design the objective functional by considering the biomedical goal, which minimizes the glioma burden and maximizes the macrophages and activated CTLs. The existence and the characterization for the optimal control are established. The uniqueness of the quadratic optimal control problem is also analyzed. We demonstrate numerically that the optimal treatment strategies using T11TS reduce the glioma burden and increase the cell count of activated CTLs and macrophages.
Collapse
Affiliation(s)
- SUBHAS KHAJANCHI
- Department of Mathematics, Presidency University, Kolkata 700073, West Bengal, India
| | - SANDIP BANERJEE
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee 247667, Uttarakhand, India
| |
Collapse
|
19
|
Peskov K, Azarov I, Chu L, Voronova V, Kosinsky Y, Helmlinger G. Quantitative Mechanistic Modeling in Support of Pharmacological Therapeutics Development in Immuno-Oncology. Front Immunol 2019; 10:924. [PMID: 31134058 PMCID: PMC6524731 DOI: 10.3389/fimmu.2019.00924] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/10/2019] [Indexed: 12/15/2022] Open
Abstract
Following the approval, in recent years, of the first immune checkpoint inhibitor, there has been an explosion in the development of immuno-modulating pharmacological modalities for the treatment of various cancers. From the discovery phase to late-stage clinical testing and regulatory approval, challenges in the development of immuno-oncology (IO) drugs are multi-fold and complex. In the preclinical setting, the multiplicity of potential drug targets around immune checkpoints, the growing list of immuno-modulatory molecular and cellular forces in the tumor microenvironment-with additional opportunities for IO drug targets, the emergence of exploratory biomarkers, and the unleashed potential of modality combinations all have necessitated the development of quantitative, mechanistically-oriented systems models which incorporate key biology and patho-physiology aspects of immuno-oncology and the pharmacokinetics of IO-modulating agents. In the clinical setting, the qualification of surrogate biomarkers predictive of IO treatment efficacy or outcome, and the corresponding optimization of IO trial design have become major challenges. This mini-review focuses on the evolution and state-of-the-art of quantitative systems models describing the tumor vs. immune system interplay, and their merging with quantitative pharmacology models of IO-modulating agents, as companion tools to support the addressing of these challenges.
Collapse
Affiliation(s)
- Kirill Peskov
- M&S Decisions, Moscow, Russia.,Computational Oncology Group, I.M. Sechenov First Moscow State Medical University of the Russian Ministry of Health, Moscow, Russia
| | | | - Lulu Chu
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Boston, MA, United States
| | | | | | - Gabriel Helmlinger
- Quantitative Clinical Pharmacology, Early Clinical Development, IMED Biotech Unit, AstraZeneca Pharmaceuticals, Boston, MA, United States
| |
Collapse
|
20
|
Cova TFGG, Bento DJ, Nunes SCC. Computational Approaches in Theranostics: Mining and Predicting Cancer Data. Pharmaceutics 2019; 11:E119. [PMID: 30871264 PMCID: PMC6471740 DOI: 10.3390/pharmaceutics11030119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 02/02/2023] Open
Abstract
The ability to understand the complexity of cancer-related data has been prompted by the applications of (1) computer and data sciences, including data mining, predictive analytics, machine learning, and artificial intelligence, and (2) advances in imaging technology and probe development. Computational modelling and simulation are systematic and cost-effective tools able to identify important temporal/spatial patterns (and relationships), characterize distinct molecular features of cancer states, and address other relevant aspects, including tumor detection and heterogeneity, progression and metastasis, and drug resistance. These approaches have provided invaluable insights for improving the experimental design of therapeutic delivery systems and for increasing the translational value of the results obtained from early and preclinical studies. The big question is: Could cancer theranostics be determined and controlled in silico? This review describes the recent progress in the development of computational models and methods used to facilitate research on the molecular basis of cancer and on the respective diagnosis and optimized treatment, with particular emphasis on the design and optimization of theranostic systems. The current role of computational approaches is providing innovative, incremental, and complementary data-driven solutions for the prediction, simplification, and characterization of cancer and intrinsic mechanisms, and to promote new data-intensive, accurate diagnostics and therapeutics.
Collapse
Affiliation(s)
- Tânia F G G Cova
- Coimbra Chemistry Centre, Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Daniel J Bento
- Coimbra Chemistry Centre, Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal.
| | - Sandra C C Nunes
- Coimbra Chemistry Centre, Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
21
|
Influence of multiple delays in brain tumor and immune system interaction with T11 target structure as a potent stimulator. Math Biosci 2018; 302:116-130. [DOI: 10.1016/j.mbs.2018.06.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 04/05/2018] [Accepted: 06/06/2018] [Indexed: 11/20/2022]
|
22
|
A structural methodology for modeling immune-tumor interactions including pro- and anti-tumor factors for clinical applications. Math Biosci 2018; 304:48-61. [PMID: 30055212 DOI: 10.1016/j.mbs.2018.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/10/2018] [Accepted: 07/17/2018] [Indexed: 12/17/2022]
Abstract
The immune system turns out to have both stimulatory and inhibitory factors influencing on tumor growth. In recent years, the pro-tumor role of immunity factors such as regulatory T cells and TGF-β cytokines has specially been considered in mathematical modeling of tumor-immune interactions. This paper presents a novel structural methodology for reviewing these models and classifies them into five subgroups on the basis of immune factors included. By using our experimental data due to immunotherapy experimentation in mice, these five modeling groups are evaluated and scored. The results show that a model with a small number of variables and coefficients performs efficiently in predicting the tumor-immune system interactions. Though immunology theorems suggest to employ a larger number of variables and coefficients, more complicated models are here shown to be inefficient due to redundant parallel pathways. So, these models are trapped in local minima and restricted in prediction capability. This paper investigates the mathematical models that were previously developed and proposes variables and pathways that are essential for modeling tumor-immune. Using these variables and pathways, a minimal structure for modeling tumor-immune interactions is proposed for future studies.
Collapse
|
23
|
Konstorum A, Vella AT, Adler AJ, Laubenbacher RC. Addressing current challenges in cancer immunotherapy with mathematical and computational modelling. J R Soc Interface 2018; 14:rsif.2017.0150. [PMID: 28659410 DOI: 10.1098/rsif.2017.0150] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 05/31/2017] [Indexed: 02/06/2023] Open
Abstract
The goal of cancer immunotherapy is to boost a patient's immune response to a tumour. Yet, the design of an effective immunotherapy is complicated by various factors, including a potentially immunosuppressive tumour microenvironment, immune-modulating effects of conventional treatments and therapy-related toxicities. These complexities can be incorporated into mathematical and computational models of cancer immunotherapy that can then be used to aid in rational therapy design. In this review, we survey modelling approaches under the umbrella of the major challenges facing immunotherapy development, which encompass tumour classification, optimal treatment scheduling and combination therapy design. Although overlapping, each challenge has presented unique opportunities for modellers to make contributions using analytical and numerical analysis of model outcomes, as well as optimization algorithms. We discuss several examples of models that have grown in complexity as more biological information has become available, showcasing how model development is a dynamic process interlinked with the rapid advances in tumour-immune biology. We conclude the review with recommendations for modellers both with respect to methodology and biological direction that might help keep modellers at the forefront of cancer immunotherapy development.
Collapse
Affiliation(s)
- Anna Konstorum
- Center for Quantitative Medicine, UConn Health, Farmington, CT, USA
| | | | - Adam J Adler
- Department of Immunology, UConn Health, Farmington, CT, USA
| | - Reinhard C Laubenbacher
- Center for Quantitative Medicine, UConn Health, Farmington, CT, USA .,Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| |
Collapse
|
24
|
Morales V, Soto-Ortiz L. Modeling Macrophage Polarization and Its Effect on Cancer Treatment Success. ACTA ACUST UNITED AC 2018; 8:36-80. [PMID: 35847834 PMCID: PMC9286492 DOI: 10.4236/oji.2018.82004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Positive feedback loops drive immune cell polarization toward a pro-tumor phenotype that accentuates immunosuppression and tumor angiogenesis. This phenotypic switch leads to the escape of cancer cells from immune destruction. These positive feedback loops are generated by cytokines such as TGF-β, Interleukin-10 and Interleukin-4, which are responsible for the polarization of monocytes and M1 macrophages into pro-tumor M2 macrophages, and the polarization of naive helper T cells intopro-tumor Th2 cells. In this article, we present a deterministic ordinary differential equation (ODE) model that includes key cellular interactions and cytokine signaling pathways that lead to immune cell polarization in the tumor microenvironment. The model was used to simulate various cancer treatments in silico. We identified combination therapies that consist of M1 macrophages or Th1 helper cells, coupled with an anti-angiogenic treatment, that are robust with respect to immune response strength, initial tumor size and treatment resistance. We also identified IL-4 and IL-10 as the targets that should be neutralized in order to make these combination treatments robust with respect to immune cell polarization. The model simulations confirmed a hypothesis based on published experimental evidence that a polarization into the M1 and Th1 phenotypes to increase the M1-to-M2 and Th1-to-Th2 ratios plays a significant role in treatment success. Our results highlight the importance of immune cell reprogramming as a viable strategy to eradicate a highly vascularized tumor when the strength of the immune response is characteristically weak and cell polarization to the pro-tumor phenotype has occurred.
Collapse
Affiliation(s)
- Valentin Morales
- Department of Engineering and Technologies, East Los Angeles College, Monterey Park, USA
| | - Luis Soto-Ortiz
- Department of Mathematics, East Los Angeles College, Monterey Park, USA
| |
Collapse
|
25
|
Khajanchi S. Uniform Persistence and Global Stability for a Brain Tumor and Immune System Interaction. ACTA ACUST UNITED AC 2017. [DOI: 10.1142/s1793048017500114] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
This paper describes the synergistic interaction between the growth of malignant gliomas and the immune system interactions using a system of coupled ordinary differential equations (ODEs). The proposed mathematical model comprises the interaction of glioma cells, macrophages, activated Cytotoxic T-Lymphocytes (CTLs), the immunosuppressive factor TGF-[Formula: see text] and the immuno-stimulatory factor IFN-[Formula: see text]. The dynamical behavior of the proposed system both analytically and numerically is investigated from the point of view of stability. By constructing Lyapunov functions, the global behavior of the glioma-free and the interior equilibrium point have been analyzed under some assumptions. Finally, we perform numerical simulations in order to illustrate our analytical findings by varying the system parameters.
Collapse
Affiliation(s)
- Subhas Khajanchi
- Department of Mathematics, Bankura University, Bankura 722155, West Bengal, India
| |
Collapse
|
26
|
Khajanchi S, Banerjee S. Quantifying the role of immunotherapeutic drug T11 target structure in progression of malignant gliomas: Mathematical modeling and dynamical perspective. Math Biosci 2017; 289:69-77. [DOI: 10.1016/j.mbs.2017.04.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/07/2017] [Accepted: 04/26/2017] [Indexed: 10/19/2022]
|
27
|
Mathematical Models for Immunology: Current State of the Art and Future Research Directions. Bull Math Biol 2016; 78:2091-2134. [PMID: 27714570 PMCID: PMC5069344 DOI: 10.1007/s11538-016-0214-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 09/26/2016] [Indexed: 01/01/2023]
Abstract
The advances in genetics and biochemistry that have taken place over the last 10 years led to significant advances in experimental and clinical immunology. In turn, this has led to the development of new mathematical models to investigate qualitatively and quantitatively various open questions in immunology. In this study we present a review of some research areas in mathematical immunology that evolved over the last 10 years. To this end, we take a step-by-step approach in discussing a range of models derived to study the dynamics of both the innate and immune responses at the molecular, cellular and tissue scales. To emphasise the use of mathematics in modelling in this area, we also review some of the mathematical tools used to investigate these models. Finally, we discuss some future trends in both experimental immunology and mathematical immunology for the upcoming years.
Collapse
|
28
|
Agur Z, Halevi-Tobias K, Kogan Y, Shlagman O. Employing dynamical computational models for personalizing cancer immunotherapy. Expert Opin Biol Ther 2016; 16:1373-1385. [PMID: 27564141 DOI: 10.1080/14712598.2016.1223622] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Recently, cancer immunotherapy has shown considerable success, but due to the complexity of the immune-cancer interactions, clinical outcomes vary largely between patients. A possible approach to overcome this difficulty may be to develop new methodologies for personal predictions of therapy outcomes, by the integration of patient data with dynamical mathematical models of the drug-affected pathophysiological processes. AREAS COVERED This review unfolds the story of mathematical modeling in cancer immunotherapy, and examines the feasibility of using these models for immunotherapy personalization. The reviewed studies suggest that response to immunotherapy can be improved by patient-specific regimens, which can be worked out by personalized mathematical models. The studies further indicate that personalized models can be constructed and validated relatively early in treatment. EXPERT OPINION The suggested methodology has the potential to raise the overall efficacy of the developed immunotherapy. If implemented already during drug development it may increase the prospects of the technology being approved for clinical use. However, schedule personalization, per se, does not comply with the current, 'one size fits all,' paradigm of clinical trials. It is worthwhile considering adjustment of the current paradigm to involve personally tailored immunotherapy regimens.
Collapse
Affiliation(s)
- Zvia Agur
- a Institute for Medical BioMathematics (IMBM) , Bene Ataroth , Israel
| | | | - Yuri Kogan
- a Institute for Medical BioMathematics (IMBM) , Bene Ataroth , Israel
| | - Ofer Shlagman
- a Institute for Medical BioMathematics (IMBM) , Bene Ataroth , Israel
| |
Collapse
|
29
|
Modeling the Treatment of Glioblastoma Multiforme and Cancer Stem Cells with Ordinary Differential Equations. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2016; 2016:1239861. [PMID: 27022405 PMCID: PMC4745194 DOI: 10.1155/2016/1239861] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 11/12/2015] [Accepted: 12/22/2015] [Indexed: 11/24/2022]
Abstract
Despite improvements in cancer therapy and treatments, tumor recurrence is a common event in cancer patients. One explanation of recurrence is that cancer therapy focuses on treatment of tumor cells and does not eradicate cancer stem cells (CSCs). CSCs are postulated to behave similar to normal stem cells in that their role is to maintain homeostasis. That is, when the population of tumor cells is reduced or depleted by treatment, CSCs will repopulate the tumor, causing recurrence. In this paper, we study the application of the CSC Hypothesis to the treatment of glioblastoma multiforme by immunotherapy. We extend the work of Kogan et al. (2008) to incorporate the dynamics of CSCs, prove the existence of a recurrence state, and provide an analysis of possible cancerous states and their dependence on treatment levels.
Collapse
|
30
|
Foryś U, Bodnar M, Kogan Y. Asymptotic dynamics of some t-periodic one-dimensional model with application to prostate cancer immunotherapy. J Math Biol 2016; 73:867-83. [PMID: 26897354 PMCID: PMC5018042 DOI: 10.1007/s00285-016-0978-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 12/02/2015] [Indexed: 01/21/2023]
Abstract
In the case of some specific cancers, immunotherapy is one of the possible treatments that can be considered. Our study is based on a mathematical model of patient-specific immunotherapy proposed in Kronik et al. (PLoS One 5(12):e15,482, 2010). This model was validated for clinical trials presented in Michael et al. (Clin Cancer Res 11(12):4469–4478, 2005). It consists of seven ordinary differential equations and its asymptotic dynamics can be described by some t-periodic one-dimensional dynamical system. In this paper we propose a generalised version of this t-periodic system and study the dynamics of the proposed model. We show that there are three possible types of the model behaviour: the solution either converges to zero, or diverges to infinity, or it is periodic. Moreover, the periodic solution is unique, and it divides the phase space into two sub-regions. The general results are applied to the PC specific case, which allow to derive conditions guaranteeing successful as well as unsuccessful treatment. The results indicate that a single vaccination is not sufficient to cure the cancer.
Collapse
Affiliation(s)
- U Foryś
- Faculty of Mathematics, Informatics and Mechanics, Institute of Applied Mathematics and Mechanics, Warsaw University, Warsaw, Poland
| | - M Bodnar
- Faculty of Mathematics, Informatics and Mechanics, Institute of Applied Mathematics and Mechanics, Warsaw University, Warsaw, Poland.
| | - Y Kogan
- Institute for Medical BioMathematics, Bene Ataroth, Israel
| |
Collapse
|
31
|
Poleszczuk JT, Luddy KA, Prokopiou S, Robertson-Tessi M, Moros EG, Fishman M, Djeu JY, Finkelstein SE, Enderling H. Abscopal Benefits of Localized Radiotherapy Depend on Activated T-cell Trafficking and Distribution between Metastatic Lesions. Cancer Res 2016; 76:1009-18. [PMID: 26833128 DOI: 10.1158/0008-5472.can-15-1423] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 12/03/2015] [Indexed: 11/16/2022]
Abstract
It remains unclear how localized radiotherapy for cancer metastases can occasionally elicit a systemic antitumor effect, known as the abscopal effect, but historically, it has been speculated to reflect the generation of a host immunotherapeutic response. The ability to purposefully and reliably induce abscopal effects in metastatic tumors could meet many unmet clinical needs. Here, we describe a mathematical model that incorporates physiologic information about T-cell trafficking to estimate the distribution of focal therapy-activated T cells between metastatic lesions. We integrated a dynamic model of tumor-immune interactions with systemic T-cell trafficking patterns to simulate the development of metastases. In virtual case studies, we found that the dissemination of activated T cells among multiple metastatic sites is complex and not intuitively predictable. Furthermore, we show that not all metastatic sites participate in systemic immune surveillance equally, and therefore the success in triggering the abscopal effect depends, at least in part, on which metastatic site is selected for localized therapy. Moreover, simulations revealed that seeding new metastatic sites may accelerate the growth of the primary tumor, because T-cell responses are partially diverted to the developing metastases, but the removal of the primary tumor can also favor the rapid growth of preexisting metastatic lesions. Collectively, our work provides the framework to prospectively identify anatomically defined focal therapy targets that are most likely to trigger an immune-mediated abscopal response and therefore may inform personalized treatment strategies in patients with metastatic disease.
Collapse
Affiliation(s)
- Jan T Poleszczuk
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida.
| | - Kimberly A Luddy
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Sotiris Prokopiou
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mark Robertson-Tessi
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Eduardo G Moros
- Department of Cancer Imaging and Metabolism, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida. Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mayer Fishman
- Department of GU Oncology MMG, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Julie Y Djeu
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Heiko Enderling
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| |
Collapse
|
32
|
|
33
|
Castillo-Montiel E, Chimal-Eguía JC, Tello JI, Piñon-Zaráte G, Herrera-Enríquez M, Castell-Rodríguez AE. Enhancing dendritic cell immunotherapy for melanoma using a simple mathematical model. Theor Biol Med Model 2015; 12:11. [PMID: 26054860 PMCID: PMC4469008 DOI: 10.1186/s12976-015-0007-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Accepted: 05/25/2015] [Indexed: 11/13/2022] Open
Abstract
Background The immunotherapy using dendritic cells (DCs) against different varieties of cancer is an approach that has been previously explored which induces a specific immune response. This work presents a mathematical model of DCs immunotherapy for melanoma in mice based on work by Experimental Immunotherapy Laboratory of the Medicine Faculty in the Universidad Autonoma de Mexico (UNAM). Method The model is a five delay differential equation (DDEs) which represents a simplified view of the immunotherapy mechanisms. The mathematical model takes into account the interactions between tumor cells, dendritic cells, naive cytotoxic T lymphocytes cells (inactivated cytotoxic cells), effector cells (cytotoxic T activated cytotoxic cells) and transforming growth factor β cytokine (TGF−β). The model is validated comparing the computer simulation results with biological trial results of the immunotherapy developed by the research group of UNAM. Results The results of the growth of tumor cells obtained by the control immunotherapy simulation show a similar amount of tumor cell population than the biological data of the control immunotherapy. Moreover, comparing the increase of tumor cells obtained from the immunotherapy simulation and the biological data of the immunotherapy applied by the UNAM researchers obtained errors of approximately 10 %. This allowed us to use the model as a framework to test hypothetical treatments. The numerical simulations suggest that by using more doses of DCs and changing the infusion time, the tumor growth decays compared with the current immunotherapy. In addition, a local sensitivity analysis is performed; the results show that the delay in time “ τ”, the maximal growth rate of tumor “r” and the maximal efficiency of tumor cytotoxic cells rate “aT” are the most sensitive model parameters. Conclusion By using this mathematical model it is possible to simulate the growth of the tumor cells with or without immunotherapy using the infusion protocol of the UNAM researchers, to obtain a good approximation of the biological trials data. It is worth mentioning that by manipulating the different parameters of the model the effectiveness of the immunotherapy may increase. This last suggests that different protocols could be implemented by the Immunotherapy Laboratory of UNAM in order to improve their results. Electronic supplementary material The online version of this article (doi:10.1186/s12976-015-0007-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- E Castillo-Montiel
- Laboratorio de Modelación y Simulación, Centro de Investigación en Computación, Instituto Politécnico Nacional, Av. Juan de Dios Bátiz, Esq. Miguel Othón de Mendizábal, Del. Gustavo A. Madero, México City, 07738, México.
| | - J C Chimal-Eguía
- Laboratorio de Modelación y Simulación, Centro de Investigación en Computación, Instituto Politécnico Nacional, Av. Juan de Dios Bátiz, Esq. Miguel Othón de Mendizábal, Del. Gustavo A. Madero, México City, 07738, México.
| | - J Ignacio Tello
- Departamento de Matemática Aplicada a las Tecnologías de la Información y las Telecomunicaciones, E.T.S.I. Sistemas Informáticos, Universidad Politécnica de Madrid, Ctral. de Valencia. Km. 7, Madrid, 28031, Spain.
| | - G Piñon-Zaráte
- Laboratorio de Inmunoterapia e Ingeniería de Tejidos, Departamento de Biológia Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Edificio A, Sexto Piso, Ciudad Universitaria, Av. Universidad No. 3000, México City, 04510, México.
| | - M Herrera-Enríquez
- Laboratorio de Inmunoterapia e Ingeniería de Tejidos, Departamento de Biológia Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Edificio A, Sexto Piso, Ciudad Universitaria, Av. Universidad No. 3000, México City, 04510, México.
| | - A E Castell-Rodríguez
- Laboratorio de Inmunoterapia e Ingeniería de Tejidos, Departamento de Biológia Celular y Tisular, Facultad de Medicina, Universidad Nacional Autónoma de México, Edificio A, Sexto Piso, Ciudad Universitaria, Av. Universidad No. 3000, México City, 04510, México.
| |
Collapse
|
34
|
Banerjee S, Khajanchi S, Chaudhuri S. A mathematical model to elucidate brain tumor abrogation by immunotherapy with T11 target structure. PLoS One 2015; 10:e0123611. [PMID: 25955428 PMCID: PMC4425651 DOI: 10.1371/journal.pone.0123611] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/19/2015] [Indexed: 11/17/2022] Open
Abstract
T11 Target structure (T11TS), a membrane glycoprotein isolated from sheep erythrocytes, reverses the immune suppressed state of brain tumor induced animals by boosting the functional status of the immune cells. This study aims at aiding in the design of more efficacious brain tumor therapies with T11 target structure. We propose a mathematical model for brain tumor (glioma) and the immune system interactions, which aims in designing efficacious brain tumor therapy. The model encompasses considerations of the interactive dynamics of glioma cells, macrophages, cytotoxic T-lymphocytes (CD8+ T-cells), TGF-β, IFN-γ and the T11TS. The system undergoes sensitivity analysis, that determines which state variables are sensitive to the given parameters and the parameters are estimated from the published data. Computer simulations were used for model verification and validation, which highlight the importance of T11 target structure in brain tumor therapy.
Collapse
Affiliation(s)
- Sandip Banerjee
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee - 247667, Uttaranchal, India
| | - Subhas Khajanchi
- Department of Mathematics, Indian Institute of Technology Roorkee, Roorkee - 247667, Uttaranchal, India
| | - Swapna Chaudhuri
- Department of Laboratory Medicine, School of Tropical Medicine, Kolkata-700073, West Bengal, India
| |
Collapse
|
35
|
Bunimovich-Mendrazitsky S, Halachmi S, Kronik N. Improving Bacillus Calmette-Guérin (BCG) immunotherapy for bladder cancer by adding interleukin 2 (IL-2): a mathematical model. MATHEMATICAL MEDICINE AND BIOLOGY-A JOURNAL OF THE IMA 2015; 33:159-88. [PMID: 25888550 DOI: 10.1093/imammb/dqv007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 03/05/2015] [Indexed: 01/28/2023]
Abstract
One of the treatments offered to non-invasive bladder cancer patients is BCG instillations, using a well-established, time-honoured protocol. Some of the patients, however, do not respond to this protocol. To examine possible changes in the protocol, we provide a platform for in silico testing of alternative protocols for BCG instillations and combinations with IL-2, to be used by urologists in planning new treatment strategies for subpopulations of bladder cancer patients who may benefit from a personalized protocol. We use a systems biology approach to describe the BCG-tumour-immune interplay and translate it into a set of mathematical differential equations. The variables of the equation set are the number of tumour cells, bacteria cells, immune cells, and cytokines participating in the tumour-immune response. Relevant parameters that describe the system's dynamics are taken from a variety of independent literature, unrelated to the clinical trial results assessed by the model predictions. Model simulations use a clinically relevant range of initial tumour sizes (tumour volume) and tumour growth rates (tumour grade), representative of a virtual population of fifty patients. Our model successfully retrieved previous clinical results for BCG induction treatment and BCG maintenance therapy with a complete response (CR) rate of 82%. Furthermore, we designed alternative maintenance protocols, using IL-2 combinations with BCG, which improved success rates up to 86% and 100% of the patients, albeit without considering possible side effects. We have shown our simulation platform to be reliable by demonstrating its ability to retrieve published clinical trial results. We used this platform to predict the outcome of treatment combinations. Our results suggest that the subpopulation of non-responsive patients may benefit from an intensified combined BCG IL-2 maintenance treatment.
Collapse
Affiliation(s)
| | - Sarel Halachmi
- Department of Urology, Bnai Zion Medical Center, Faculty of Medicine, Technion, Haifa, Israel
| | - Natalie Kronik
- Quantitative Oncology and Medicine Association, Rte de l'Etoile 37, 202, Gorgier, Switzerland
| |
Collapse
|
36
|
Goriely A, Geers MGD, Holzapfel GA, Jayamohan J, Jérusalem A, Sivaloganathan S, Squier W, van Dommelen JAW, Waters S, Kuhl E. Mechanics of the brain: perspectives, challenges, and opportunities. Biomech Model Mechanobiol 2015; 14:931-65. [PMID: 25716305 PMCID: PMC4562999 DOI: 10.1007/s10237-015-0662-4] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/14/2015] [Indexed: 12/24/2022]
Abstract
The human brain is the continuous subject of extensive investigation aimed at understanding its behavior and function. Despite a clear evidence that mechanical factors play an important role in regulating brain activity, current research efforts focus mainly on the biochemical or electrophysiological activity of the brain. Here, we show that classical mechanical concepts including deformations, stretch, strain, strain rate, pressure, and stress play a crucial role in modulating both brain form and brain function. This opinion piece synthesizes expertise in applied mathematics, solid and fluid mechanics, biomechanics, experimentation, material sciences, neuropathology, and neurosurgery to address today’s open questions at the forefront of neuromechanics. We critically review the current literature and discuss challenges related to neurodevelopment, cerebral edema, lissencephaly, polymicrogyria, hydrocephaly, craniectomy, spinal cord injury, tumor growth, traumatic brain injury, and shaken baby syndrome. The multi-disciplinary analysis of these various phenomena and pathologies presents new opportunities and suggests that mechanical modeling is a central tool to bridge the scales by synthesizing information from the molecular via the cellular and tissue all the way to the organ level.
Collapse
Affiliation(s)
- Alain Goriely
- Mathematical Institute, University of Oxford, Oxford, OX2 6GG, UK,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Iarosz KC, Borges FS, Batista AM, Baptista MS, Siqueira RAN, Viana RL, Lopes SR. Mathematical model of brain tumour with glia-neuron interactions and chemotherapy treatment. J Theor Biol 2015; 368:113-21. [PMID: 25596516 DOI: 10.1016/j.jtbi.2015.01.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Revised: 12/09/2014] [Accepted: 01/07/2015] [Indexed: 01/23/2023]
Abstract
In recent years, it became clear that a better understanding of the interactions among the main elements involved in the cancer network is necessary for the treatment of cancer and the suppression of cancer growth. In this work we propose a system of coupled differential equations that model brain tumour under treatment by chemotherapy, which considers interactions among the glial cells, the glioma, the neurons, and the chemotherapeutic agents. We study the conditions for the glioma growth to be eliminated, and identify values of the parameters for which the inhibition of the glioma growth is obtained with a minimal loss of healthy cells.
Collapse
Affiliation(s)
- Kelly C Iarosz
- Institute for Complex Systems and Mathematical Biology, University of Aberdeen, AB24 3UE Aberdeen, UK.
| | - Fernando S Borges
- Pós-Graduação em Ciências/Física, Universidade Estadual de Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Antonio M Batista
- Institute for Complex Systems and Mathematical Biology, University of Aberdeen, AB24 3UE Aberdeen, UK; Pós-Graduação em Ciências/Física, Universidade Estadual de Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil; Departamento de Matemática e Estatística, Universidade Estadual de Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Murilo S Baptista
- Institute for Complex Systems and Mathematical Biology, University of Aberdeen, AB24 3UE Aberdeen, UK
| | - Regiane A N Siqueira
- Pós-Graduação em Ciências/Física, Universidade Estadual de Ponta Grossa, 84030-900 Ponta Grossa, PR, Brazil
| | - Ricardo L Viana
- Departamento de Física, Universidade Federal do Paraná, 81531-990 Curitiba, PR, Brazil
| | - Sergio R Lopes
- Departamento de Física, Universidade Federal do Paraná, 81531-990 Curitiba, PR, Brazil
| |
Collapse
|
38
|
Immunobiology and immunotherapeutic targeting of glioma stem cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 853:139-66. [PMID: 25895711 DOI: 10.1007/978-3-319-16537-0_8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
For decades human brain tumors have confounded our efforts to effectively manage and treat patients. In adults, glioblastoma multiforme is the most common malignant brain tumor with a patient survival of just over 14 months. In children, brain tumors are the leading cause of solid tumor cancer death and gliomas account for one-fifth of all childhood cancers. Despite advances in conventional treatments such as surgical resection, radiotherapy, and systemic chemotherapy, the incidence and mortality rates for gliomas have essentially stayed the same. Furthermore, research efforts into novel therapeutics that initially appeared promising have yet to show a marked benefit. A shocking and somewhat disturbing view is that investigators and clinicians may have been targeting the wrong cells, resulting in the appearance of the removal or eradication of patient gliomas only to have brain cancer recurrence. Here we review research progress in immunotherapy as it pertains to glioma treatment and how it can and is being adapted to target glioma stem cells (GSCs) as a means of dealing with this potential paradigm.
Collapse
|
39
|
Viger L, Denis F, Rosalie M, Letellier C. A cancer model for the angiogenic switch. J Theor Biol 2014; 360:21-33. [DOI: 10.1016/j.jtbi.2014.06.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 06/04/2014] [Accepted: 06/17/2014] [Indexed: 11/28/2022]
|
40
|
Pellegatta S, Eoli M, Frigerio S, Antozzi C, Bruzzone MG, Cantini G, Nava S, Anghileri E, Cuppini L, Cuccarini V, Ciusani E, Dossena M, Pollo B, Mantegazza R, Parati EA, Finocchiaro G. The natural killer cell response and tumor debulking are associated with prolonged survival in recurrent glioblastoma patients receiving dendritic cells loaded with autologous tumor lysates. Oncoimmunology 2014; 2:e23401. [PMID: 23802079 PMCID: PMC3661164 DOI: 10.4161/onci.23401] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 12/19/2012] [Accepted: 12/21/2012] [Indexed: 12/27/2022] Open
Abstract
Recurrent glioblastomas (GBs) are highly aggressive tumors associated with a 6–8 mo survival rate. In this study, we evaluated the possible benefits of an immunotherapeutic strategy based on mature dendritic cells (DCs) loaded with autologous tumor-cell lysates in 15 patients affected by recurrent GB. The median progression-free survival (PFS) of this patient cohort was 4.4 mo, and the median overall survival (OS) was 8.0 mo. Patients with small tumors at the time of the first vaccination (< 20 cm3; n = 8) had significantly longer PFS and OS than the other patients (6.0 vs. 3.0 mo, p = 0.01; and 16.5 vs. 7.0 mo, p = 0.003, respectively). CD8+ T cells, CD56+ natural killer (NK) cells and other immune parameters, such as the levels of transforming growth factor β, vascular endothelial growth factor, interleukin-12 and interferon γ (IFNγ), were measured in the peripheral blood and serum of patients before and after immunization, which enabled us to obtain a vaccination/baseline ratio (V/B ratio). An increased V/B ratio for NK cells, but not CD8+ T cells, was significantly associated with prolonged PFS and OS. Patients exhibiting NK-cell responses were characterized by high levels of circulating IFNγ and E4BP4, an NK-cell transcription factor. Furthermore, the NK cell V/B ratio was inversely correlated with the TGFβ2 and VEGF V/B ratios. These results suggest that tumor-loaded DCs may increase the survival rate of patients with recurrent GB after effective tumor debulking, and emphasize the role of the NK-cell response in this therapeutic setting.
Collapse
Affiliation(s)
- Serena Pellegatta
- Unit of Molecular Neuro-Oncology; Fondazione I.R.C.C.S. Istituto Neurologico C. Besta; Milan, Italy ; Department of Experimental Oncology; European Institute of Oncology - Campus IFOM-IEO; Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
dePillis LG, Eladdadi A, Radunskaya AE. Modeling cancer-immune responses to therapy. J Pharmacokinet Pharmacodyn 2014; 41:461-78. [DOI: 10.1007/s10928-014-9386-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/17/2014] [Indexed: 12/26/2022]
|
42
|
Ribba B, Holford NH, Magni P, Trocóniz I, Gueorguieva I, Girard P, Sarr C, Elishmereni M, Kloft C, Friberg LE. A review of mixed-effects models of tumor growth and effects of anticancer drug treatment used in population analysis. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e113. [PMID: 24806032 PMCID: PMC4050233 DOI: 10.1038/psp.2014.12] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 03/14/2014] [Indexed: 12/12/2022]
Abstract
Population modeling of tumor size dynamics has recently emerged as an important tool in pharmacometric research. A series of new mixed-effects models have been reported recently, and we present herein a synthetic view of models with published mathematical equations aimed at describing the dynamics of tumor size in cancer patients following anticancer drug treatment. This selection of models will constitute the basis for the Drug Disease Model Resources (DDMoRe) repository for models on oncology.
Collapse
Affiliation(s)
- B Ribba
- INRIA, Project-Team NUMED, École Normale Supérieure de Lyon, Lyon, France
| | - N H Holford
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| | - P Magni
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, Pavia, Italy
| | - I Trocóniz
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, Pamplona, Spain
| | - I Gueorguieva
- Global PK/PD Department, Lilly Research Laboratories, Surrey, UK
| | - P Girard
- Merck Institute for Pharmacometrics, EPFL, Lausanne, Switzerland
| | - C Sarr
- Advanced Quantitative Sciences Department, Novartis Pharma AG, Basel, Switzerland
| | | | - C Kloft
- Department of Clinical Pharmacy and Biochemistry, Institute of Pharmacy, Freie Universitaet Berlin, Berlin, Germany
| | - L E Friberg
- Department of Pharmaceutical Biosciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
Dudek RM, Chuang Y, Leonard JN. Engineered cell-based therapies: a vanguard of design-driven medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 844:369-91. [PMID: 25480651 DOI: 10.1007/978-1-4939-2095-2_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Engineered cell-based therapies are uniquely capable of performing sophisticated therapeutic functions in vivo, and this strategy is yielding promising clinical benefits for treating cancer. In this review, we discuss key opportunities and challenges for engineering customized cellular functions using cell-based therapy for cancer as a representative case study. We examine the historical development of chimeric antigen receptor (CAR) therapies as an illustration of the engineering design cycle. We also consider the potential roles that the complementary disciplines of systems biology and synthetic biology may play in realizing safe and effective treatments for a broad range of patients and diseases. In particular, we discuss how systems biology may facilitate both fundamental research and clinical translation, and we describe how the emerging field of synthetic biology is providing novel modalities for building customized cellular functions to overcome existing clinical barriers. Together, these approaches provide a powerful set of conceptual and experimental tools for transforming information into understanding, and for translating understanding into novel therapeutics to establish a new framework for design-driven medicine.
Collapse
Affiliation(s)
- Rachel M Dudek
- Northwestern University, 2145 Sheridan Road, Technological Institute, Rm. E136, Evanston, IL, 60208-3120, USA,
| | | | | |
Collapse
|
44
|
Gallasch R, Efremova M, Charoentong P, Hackl H, Trajanoski Z. Mathematical models for translational and clinical oncology. J Clin Bioinforma 2013; 3:23. [PMID: 24195863 PMCID: PMC3828625 DOI: 10.1186/2043-9113-3-23] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/04/2013] [Indexed: 01/22/2023] Open
Abstract
In the context of translational and clinical oncology, mathematical models can provide novel insights into tumor-related processes and can support clinical oncologists in the design of the treatment regime, dosage, schedule, toxicity and drug-sensitivity. In this review we present an overview of mathematical models in this field beginning with carcinogenesis and proceeding to the different cancer treatments. By doing so we intended to highlight recent developments and emphasize the power of such theoretical work.We first highlight mathematical models for translational oncology comprising epidemiologic and statistical models, mechanistic models for carcinogenesis and tumor growth, as well as evolutionary dynamics models which can help to describe and overcome a major problem in the clinic: therapy resistance. Next we review models for clinical oncology with a special emphasis on therapy including chemotherapy, targeted therapy, radiotherapy, immunotherapy and interaction of cancer cells with the immune system.As evident from the published studies, mathematical modeling and computational simulation provided valuable insights into the molecular mechanisms of cancer, and can help to improve diagnosis and prognosis of the disease, and pinpoint novel therapeutic targets.
Collapse
Affiliation(s)
| | | | | | | | - Zlatko Trajanoski
- Biocenter, Division of Bioinformatics, Innsbruck Medical University, Innrain 80, 6020 Innsbruck, Austria.
| |
Collapse
|
45
|
Badhiwala J, Decker WK, Berens ME, Bhardwaj RD. Clinical trials in cellular immunotherapy for brain/CNS tumors. Expert Rev Neurother 2013; 13:405-24. [PMID: 23545055 DOI: 10.1586/ern.13.23] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
High-grade gliomas are the most common type of primary malignant brain/CNS tumor. There have been only modest advances in surgical techniques, radiotherapy and chemotherapy for high-grade gliomas over the past several decades. None of these have provided a major improvement in survival for patients. Recently, immunotherapy has been explored for the treatment of high-grade gliomas. Immunotherapy capitalizes on the specificity of the host immune system to selectively target tumor cells for destruction, while sparing normal brain parenchyma, thus making it a particularly attractive option. This article provides a comprehensive review of published clinical trials evaluating cellular immunotherapy in primary brain/CNS tumors.
Collapse
Affiliation(s)
- Jetan Badhiwala
- Michael G DeGroote School of Medicine, McMaster University, 1280 Main Street W, Hamilton, ON, L8S 4K1, Canada
| | | | | | | |
Collapse
|
46
|
Petrosiute A, Auletta JJ, Lazarus HM. Achieving graft-versus-tumor effect in brain tumor patients: from autologous progenitor cell transplant to active immunotherapy. Immunotherapy 2013. [PMID: 23194364 DOI: 10.2217/imt.12.96] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Success in treating aggressive brain tumors like glioblastoma multiforme and medulloblastoma remains challenging, in part because these malignancies overcome CNS immune surveillance. New insights into brain tumor immunology have led to a rational development of immunotherapeutic strategies, including cytotoxic Tlymphocyte therapies and dendritic cell vaccines. However, these therapies are most effective when applied in a setting of minimal residual disease, so require prior use of standard cytotoxic therapies or cytoreduction by surgery. Myeloablative chemotherapy with autologous hematopoietic cell transplantation (autoHCT) can offer a platform upon which different cellular therapies can be effectively instituted. Specifically, this approach provides an inherent 'chemical debulking' through high-dose chemotherapy and a graft-versus-tumor effect through an autologous T-cell replete graft. Furthermore, autoHCT may be beneficial in 'resetting' the body's immune system, potentially 'breaking' tumor tolerance, and in providing a 'boost' of immune effector cells (NK cells or cytotoxic T lymphocytes), which could augment desired anti-tumor effects. As literature on the use of autoHCT in brain tumors is scarce, aspects of immunotherapies applied in non-CNS malignancies are reviewed as potential therapies that could be used in conjunction with autoHCT to eradicate brain tumors.
Collapse
Affiliation(s)
- Agne Petrosiute
- Department of Pediatrics, Hematology/Oncology, Rainbow Babies & Children's Hospital, Case Western Reserve University, 11100 Euclid Avenue, Mailstop 6054, Cleveland, OH 44106, USA.
| | | | | |
Collapse
|
47
|
Parra-Guillen ZP, Berraondo P, Grenier E, Ribba B, Troconiz IF. Mathematical model approach to describe tumour response in mice after vaccine administration and its applicability to immune-stimulatory cytokine-based strategies. AAPS JOURNAL 2013; 15:797-807. [PMID: 23605806 DOI: 10.1208/s12248-013-9483-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Accepted: 03/26/2013] [Indexed: 01/21/2023]
Abstract
Immunotherapy is a growing therapeutic strategy in oncology based on the stimulation of innate and adaptive immune systems to induce the death of tumour cells. In this paper, we have developed a population semi-mechanistic model able to characterize the mechanisms implied in tumour growth dynamic after the administration of CyaA-E7, a vaccine able to target antigen to dendritic cells, thus triggering a potent immune response. The mathematical model developed presented the following main components: (1) tumour progression in the animals without treatment was described with a linear model, (2) vaccine effects were modelled assuming that vaccine triggers a non-instantaneous immune response inducing cell death. Delayed response was described with a series of two transit compartments, (3) a resistance effect decreasing vaccine efficiency was also incorporated through a regulator compartment dependent upon tumour size, and (4) a mixture model at the level of the elimination of the induced signal vaccine (k 2) to model tumour relapse after treatment, observed in a small percentage of animals (15.6%). The proposed model structure was successfully applied to describe antitumor effect of IL-12, suggesting its applicability to different immune-stimulatory therapies. In addition, a simulation exercise to evaluate in silico the impact on tumour size of possible combination therapies has been shown. This type of mathematical approaches may be helpful to maximize the information obtained from experiments in mice, reducing the number of animals and the cost of developing new antitumor immunotherapies.
Collapse
Affiliation(s)
- Zinnia P Parra-Guillen
- Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Navarra, C/Irunlarrea 1, 31008, Pamplona, Navarra, Spain
| | | | | | | | | |
Collapse
|
48
|
Letellier C, Denis F, Aguirre L. What can be learned from a chaotic cancer model? J Theor Biol 2013; 322:7-16. [DOI: 10.1016/j.jtbi.2013.01.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Revised: 11/20/2012] [Accepted: 01/05/2013] [Indexed: 02/02/2023]
|
49
|
Kogan Y, Agur Z, Elishmereni M. A mathematical model for the immunotherapeutic control of the Th1/Th2 imbalance in melanoma. ACTA ACUST UNITED AC 2013. [DOI: 10.3934/dcdsb.2013.18.1017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
50
|
Multifaceted Kinetics of Immuno-Evasion from Tumor Dormancy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 734:111-43. [DOI: 10.1007/978-1-4614-1445-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|