1
|
Hao S, Ge P, Su W, Wang Y, Abd El-Aty AM, Tan M. Steady-State Delivery and Chemical Modification of Food Nutrients to Improve Cancer Intervention Ability. Foods 2024; 13:1363. [PMID: 38731734 PMCID: PMC11083276 DOI: 10.3390/foods13091363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Cancer is a crucial global health problem, and prevention is an important strategy to reduce the burden of the disease. Daily diet is the key modifiable risk factor for cancer, and an increasing body of evidence suggests that specific nutrients in foods may have a preventive effect against cancer. This review summarizes the current evidence on the role of nutrients from foods in cancer intervention. It discusses the potential mechanisms of action of various dietary components, including phytochemicals, vitamins, minerals, and fiber. The findings of epidemiological and clinical studies on their association with cancer risk are highlighted. The foods are rich in bioactive compounds such as carotenoids, flavonoids, and ω-3 fatty acids, which have been proven to have anticancer properties. The effects of steady-state delivery and chemical modification of these food's bioactive components on anticancer and intervention are summarized. Future research should focus on identifying the specific bioactive compounds in foods responsible for their intervention effects and exploring the potential synergistic effects of combining different nutrients in foods. Dietary interventions that incorporate multiple nutrients and whole foods may hold promise for reducing the risk of cancer and improving overall health.
Collapse
Affiliation(s)
- Sijia Hao
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China; (S.H.); (P.G.); (W.S.); (Y.W.)
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, China
| | - Peng Ge
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China; (S.H.); (P.G.); (W.S.); (Y.W.)
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, China
| | - Wentao Su
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China; (S.H.); (P.G.); (W.S.); (Y.W.)
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, China
| | - Yuxiao Wang
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China; (S.H.); (P.G.); (W.S.); (Y.W.)
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, China
| | - A. M. Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum 25240, Turkey
| | - Mingqian Tan
- State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, China; (S.H.); (P.G.); (W.S.); (Y.W.)
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
- National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, China
- Dalian Key Laboratory for Precision Nutrition, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
2
|
Wang Z, Cho H, Choyke P, Levy D, Sato N. A Mathematical Model of TCR-T Cell Therapy for Cervical Cancer. Bull Math Biol 2024; 86:57. [PMID: 38625492 DOI: 10.1007/s11538-024-01261-9] [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: 09/22/2023] [Accepted: 01/11/2024] [Indexed: 04/17/2024]
Abstract
Engineered T cell receptor (TCR)-expressing T (TCR-T) cells are intended to drive strong anti-tumor responses upon recognition of the specific cancer antigen, resulting in rapid expansion in the number of TCR-T cells and enhanced cytotoxic functions, causing cancer cell death. However, although TCR-T cell therapy against cancers has shown promising results, it remains difficult to predict which patients will benefit from such therapy. We develop a mathematical model to identify mechanisms associated with an insufficient response in a mouse cancer model. We consider a dynamical system that follows the population of cancer cells, effector TCR-T cells, regulatory T cells (Tregs), and "non-cancer-killing" TCR-T cells. We demonstrate that the majority of TCR-T cells within the tumor are "non-cancer-killing" TCR-T cells, such as exhausted cells, which contribute little or no direct cytotoxicity in the tumor microenvironment (TME). We also establish two important factors influencing tumor regression: the reversal of the immunosuppressive TME following depletion of Tregs, and the increased number of effector TCR-T cells with antitumor activity. Using mathematical modeling, we show that certain parameters, such as increasing the cytotoxicity of effector TCR-T cells and modifying the number of TCR-T cells, play important roles in determining outcomes.
Collapse
Affiliation(s)
- Zuping Wang
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA
| | - Heyrim Cho
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - Peter Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Doron Levy
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA.
| | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
3
|
Valega-Mackenzie W, Rodriguez Messan M, Yogurtcu ON, Nukala U, Sauna ZE, Yang H. Dose optimization of an adjuvanted peptide-based personalized neoantigen melanoma vaccine. PLoS Comput Biol 2024; 20:e1011247. [PMID: 38427689 PMCID: PMC10936818 DOI: 10.1371/journal.pcbi.1011247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 03/13/2024] [Accepted: 01/03/2024] [Indexed: 03/03/2024] Open
Abstract
The advancements in next-generation sequencing have made it possible to effectively detect somatic mutations, which has led to the development of personalized neoantigen cancer vaccines that are tailored to the unique variants found in a patient's cancer. These vaccines can provide significant clinical benefit by leveraging the patient's immune response to eliminate malignant cells. However, determining the optimal vaccine dose for each patient is a challenge due to the heterogeneity of tumors. To address this challenge, we formulate a mathematical dose optimization problem based on a previous mathematical model that encompasses the immune response cascade produced by the vaccine in a patient. We propose an optimization approach to identify the optimal personalized vaccine doses, considering a fixed vaccination schedule, while simultaneously minimizing the overall number of tumor and activated T cells. To validate our approach, we perform in silico experiments on six real-world clinical trial patients with advanced melanoma. We compare the results of applying an optimal vaccine dose to those of a suboptimal dose (the dose used in the clinical trial and its deviations). Our simulations reveal that an optimal vaccine regimen of higher initial doses and lower final doses may lead to a reduction in tumor size for certain patients. Our mathematical dose optimization offers a promising approach to determining an optimal vaccine dose for each patient and improving clinical outcomes.
Collapse
Affiliation(s)
- Wencel Valega-Mackenzie
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Marisabel Rodriguez Messan
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Osman N. Yogurtcu
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Ujwani Nukala
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Zuben E. Sauna
- Office of Therapeutic Products, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Hong Yang
- Office of Biostatistics and Pharmacovigilance, Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| |
Collapse
|
4
|
Pei Y, Han S, Li C, Lei J, Wen F. Data-based modeling of breast cancer and optimal therapy. J Theor Biol 2023; 573:111593. [PMID: 37544589 DOI: 10.1016/j.jtbi.2023.111593] [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: 04/02/2023] [Revised: 07/19/2023] [Accepted: 08/02/2023] [Indexed: 08/08/2023]
Abstract
Excessive accumulation of β-catenin proteins is a vital driver in the development of breast cancer. Many clinical assessments incorporating immunotherapy with targeted mRNA of β-catenin are costly endeavor. This paper develops novel mathematical models for different treatments by invoking available clinical data to calibrate models, along with the selection and evaluation of therapy strategies in a faster manner with lower cost. Firstly, in order to explore the interactions between cancer cells and the immune system within the tumor microenvironment, we construct different types of breast cancer treatment models based on RNA interference technique and immune checkpoint inhibitors, which have been proved to be an effective combined therapy in pre-clinical trials associated with the inhibition of β-catenin proteins to enhance intrinsic anti-tumor immune response. Secondly, various techniques including MCMC are adopted to estimate multiple parameters and thus simulations in agreement with experimental results sustain the validity of our models. Furthermore, the gradient descent method and particle swarm algorithm are designed to optimize therapy schemes to inhibit the growth of tumor and lower the treatment cost. Considering the mechanisms of drug resistance in vivo, simulations exhibit that therapies are ineffective resulting in cancer relapse in the prolonged time. For this reason, parametric sensitivity analysis sheds light on the choice of new treatments which indicate that, in addition to inhibiting β-catenin proteins and improving self-immunity, the injection of dendritic cells promoting immunity may provide a novel vision for the future of cancer treatment. Overall, our study provides witness of principle from a mathematical perspective to guide clinical trials and the selection of treatment regimens.
Collapse
Affiliation(s)
- Yongzhen Pei
- School of Mathematical Sciences, Tiangong University, Tianjin 300387, China.
| | - Siqi Han
- School of Mathematical Sciences, Tiangong University, Tianjin 300387, China.
| | - Changguo Li
- Department of Basic Science, Army Military Transportation University, Tianjin 300161, China.
| | - Jinzhi Lei
- School of Mathematical Sciences, Tiangong University, Tianjin 300387, China.
| | - Fengxi Wen
- School of Mathematical Sciences, Tiangong University, Tianjin 300387, China.
| |
Collapse
|
5
|
Effects of intravenous administration of polysaccharide purified from fermented barley on tumor metastasis inhibition via immunostimulating activities. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
6
|
Hormuth DA, Farhat M, Christenson C, Curl B, Chad Quarles C, Chung C, Yankeelov TE. Opportunities for improving brain cancer treatment outcomes through imaging-based mathematical modeling of the delivery of radiotherapy and immunotherapy. Adv Drug Deliv Rev 2022; 187:114367. [PMID: 35654212 PMCID: PMC11165420 DOI: 10.1016/j.addr.2022.114367] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 11/01/2022]
Abstract
Immunotherapy has become a fourth pillar in the treatment of brain tumors and, when combined with radiation therapy, may improve patient outcomes and reduce the neurotoxicity. As with other combination therapies, the identification of a treatment schedule that maximizes the synergistic effect of radiation- and immune-therapy is a fundamental challenge. Mechanism-based mathematical modeling is one promising approach to systematically investigate therapeutic combinations to maximize positive outcomes within a rigorous framework. However, successful clinical translation of model-generated combinations of treatment requires patient-specific data to allow the models to be meaningfully initialized and parameterized. Quantitative imaging techniques have emerged as a promising source of high quality, spatially and temporally resolved data for the development and validation of mathematical models. In this review, we will present approaches to personalize mechanism-based modeling frameworks with patient data, and then discuss how these techniques could be leveraged to improve brain cancer outcomes through patient-specific modeling and optimization of treatment strategies.
Collapse
Affiliation(s)
- David A Hormuth
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA.
| | - Maguy Farhat
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Chase Christenson
- Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Brandon Curl
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - C Chad Quarles
- Barrow Neuroimaging Innovation Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Caroline Chung
- Departments of Radiation Oncology, MD Anderson Cancer Center, Houston, TX 77230, USA
| | - Thomas E Yankeelov
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Diagnostic Medicine, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Oncology, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Livestrong Cancer Institutes, The University of Texas at Austin, Austin, TX 78712, USA; Departments of Imaging Physics, MD Anderson Cancer Center, Houston, TX 77230, USA
| |
Collapse
|
7
|
Son SU, Park HY, Suh HJ, Shin KS. Evaluation of antitumor metastasis via immunostimulating activities of pectic polysaccharides isolated from radish leaves. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
8
|
Chaudhury A, Zhu X, Chu L, Goliaei A, June CH, Kearns JD, Stein AM. Chimeric Antigen Receptor T Cell Therapies: A Review of Cellular Kinetic-Pharmacodynamic Modeling Approaches. J Clin Pharmacol 2021; 60 Suppl 1:S147-S159. [PMID: 33205434 DOI: 10.1002/jcph.1691] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Chimeric antigen receptor T cell (CAR-T cell) therapies have shown significant efficacy in CD19+ leukemias and lymphomas. There remain many challenges and questions for improving next-generation CAR-T cell therapies, and mathematical modeling of CAR-T cells may play a role in supporting further development. In this review, we introduce a mathematical modeling taxonomy for a set of relatively simple cellular kinetic-pharmacodynamic models that describe the in vivo dynamics of CAR-T cell and their interactions with cancer cells. We then discuss potential extensions of this model to include target binding, tumor distribution, cytokine-release syndrome, immunophenotype differentiation, and genotypic heterogeneity.
Collapse
Affiliation(s)
- Anwesha Chaudhury
- Pharmacometrics, Novartis Institutes of BioMedical Research, Cambridge, Massachusetts, USA
| | - Xu Zhu
- PK Sciences Oncology, Novartis Institutes of BioMedical Research, Cambridge, Massachusetts, USA
| | - Lulu Chu
- PK Sciences Modeling & Simulation, Novartis Institutes of BioMedical Research, Cambridge, Massachusetts, USA
| | - Ardeshir Goliaei
- PK Sciences Modeling & Simulation, Novartis Institutes of BioMedical Research, Cambridge, Massachusetts, USA
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeffrey D Kearns
- PK Sciences Modeling & Simulation, Novartis Institutes of BioMedical Research, Cambridge, Massachusetts, USA
| | - Andrew M Stein
- Pharmacometrics, Novartis Institutes of BioMedical Research, Cambridge, Massachusetts, USA
| |
Collapse
|
9
|
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
|
10
|
Idrees M, Sohail A. Bio-algorithms for the modeling and simulation of cancer cells and the immune response. BIO-ALGORITHMS AND MED-SYSTEMS 2021. [DOI: 10.1515/bams-2020-0054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Abstract
There have been significant developments in clinical, experimental, and theoretical approaches to understand the biomechanics of tumor cells and immune cells. Cytotoxic T lymphocytes (CTLs) are regarded as a major antitumor mechanism of immune cells. Mathematical modeling of tumor growth is an important and useful tool to observe and understand clinical phenomena analytically. This work develops a novel two-variable mathematical model to describe the interaction of tumor cells and CTLs. The designed model is providing an integrated framework to investigate the complexity of tumor progression and answer clinical questions that cannot always be reached with experimental tools. The parameters of the model are estimated from experimental study and stability analysis of the model is performed through nullclines. A global sensitivity analysis is also performed to check the uncertainty of the parameters. The results of numerical simulations of the model support the importance of the CTLs and demonstrate that CTLs can eliminate small tumors. The proposed model provides efficacious information to study and demonstrate the complex dynamics of breast cancer.
Collapse
Affiliation(s)
- Muhammad Idrees
- Department of Mathematics , COMSATS University Islamabad , Lahore , Pakistan
| | - Ayesha Sohail
- Department of Mathematics , COMSATS University Islamabad , Lahore , Pakistan
| |
Collapse
|
11
|
Abstract
Modern cancer immunotherapy has revolutionised oncology and carries the potential to radically change the approach to cancer treatment. However, numerous questions remain to be answered to understand immunotherapy response better and further improve the benefit for future cancer patients. Computational models are promising tools that can contribute to accelerated immunotherapy research by providing new clues and hypotheses that could be tested in future trials, based on preceding simulations in addition to the empirical rationale. In this topical review, we briefly summarise the history of cancer immunotherapy, including computational modelling of traditional cancer immunotherapy, and comprehensively review computational models of modern cancer immunotherapy, such as immune checkpoint inhibitors (as monotherapy and combination treatment), co-stimulatory agonistic antibodies, bispecific antibodies, and chimeric antigen receptor T cells. The modelling approaches are classified into one of the following categories: data-driven top-down vs mechanistic bottom-up, simplistic vs detailed, continuous vs discrete, and hybrid. Several common modelling approaches are summarised, such as pharmacokinetic/pharmacodynamic models, Lotka-Volterra models, evolutionary game theory models, quantitative systems pharmacology models, spatio-temporal models, agent-based models, and logic-based models. Pros and cons of each modelling approach are critically discussed, particularly with the focus on the potential for successful translation into immuno-oncology research and routine clinical practice. Specific attention is paid to calibration and validation of each model, which is a necessary prerequisite for any successful model, and at the same time, one of the main obstacles. Lastly, we provide guidelines and suggestions for the future development of the field.
Collapse
Affiliation(s)
- Damijan Valentinuzzi
- Jožef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia. Faculty of Mathematics and Physics, University of Ljubljana, Jadranska ulica 19, 1111 Ljubljana, Slovenia
| | | |
Collapse
|
12
|
Sové RJ, Jafarnejad M, Zhao C, Wang H, Ma H, Popel AS. QSP-IO: A Quantitative Systems Pharmacology Toolbox for Mechanistic Multiscale Modeling for Immuno-Oncology Applications. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 9:484-497. [PMID: 32618119 PMCID: PMC7499194 DOI: 10.1002/psp4.12546] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 07/17/2020] [Indexed: 12/25/2022]
Abstract
Immunotherapy has shown great potential in the treatment of cancer; however, only a fraction of patients respond to treatment, and many experience autoimmune‐related side effects. The pharmaceutical industry has relied on mathematical models to study the behavior of candidate drugs and more recently, complex, whole‐body, quantitative systems pharmacology (QSP) models have become increasingly popular for discovery and development. QSP modeling has the potential to discover novel predictive biomarkers as well as test the efficacy of treatment plans and combination therapies through virtual clinical trials. In this work, we present a QSP modeling platform for immuno‐oncology (IO) that incorporates detailed mechanisms for important immune interactions. This modular platform allows for the construction of QSP models of IO with varying degrees of complexity based on the research questions. Finally, we demonstrate the use of the platform through two example applications of immune checkpoint therapy.
Collapse
Affiliation(s)
- Richard J Sové
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Chen Zhao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hanwen Wang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Huilin Ma
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
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: 8] [Impact Index Per Article: 2.0] [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
|
14
|
Metaheuristics and Pontryagin's minimum principle for optimal therapeutic protocols in cancer immunotherapy: a case study and methods comparison. J Math Biol 2020; 81:691-723. [PMID: 32712711 DOI: 10.1007/s00285-020-01525-7] [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: 07/25/2019] [Revised: 06/04/2020] [Indexed: 12/24/2022]
Abstract
In this paper, the performance appropriateness of population-based metaheuristics for immunotherapy protocols is investigated on a comparative basis while the goal is to stimulate the immune system to defend against cancer. For this purpose, genetic algorithm and particle swarm optimization are employed and compared with modern method of Pontryagin's minimum principle (PMP). To this end, a well-known mathematical model of cell-based cancer immunotherapy is described and examined to formulate the optimal control problem in which the objective is the annihilation of tumour cells by using the minimum amount of cultured immune cells. In this regard, the main aims are: (i) to introduce a single-objective optimization problem and to design the considered metaheuristics in order to appropriately deal with it; (ii) to use the PMP in order to obtain the necessary conditions for optimality, i.e. the governing boundary value problem; (iii) to measure the results obtained by using the proposed metaheuristics against those results obtained by using an indirect approach called forward-backward sweep method; and finally (iv) to produce a set of optimal treatment strategies by formulating the problem in a bi-objective form and demonstrating its advantages over single-objective optimization problem. A set of obtained results conforms the performance capabilities of the considered metaheuristics.
Collapse
|
15
|
Albrecht M, Lucarelli P, Kulms D, Sauter T. Computational models of melanoma. Theor Biol Med Model 2020; 17:8. [PMID: 32410672 PMCID: PMC7222475 DOI: 10.1186/s12976-020-00126-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 04/29/2020] [Indexed: 02/08/2023] Open
Abstract
Genes, proteins, or cells influence each other and consequently create patterns, which can be increasingly better observed by experimental biology and medicine. Thereby, descriptive methods of statistics and bioinformatics sharpen and structure our perception. However, additionally considering the interconnectivity between biological elements promises a deeper and more coherent understanding of melanoma. For instance, integrative network-based tools and well-grounded inductive in silico research reveal disease mechanisms, stratify patients, and support treatment individualization. This review gives an overview of different modeling techniques beyond statistics, shows how different strategies align with the respective medical biology, and identifies possible areas of new computational melanoma research.
Collapse
Affiliation(s)
- Marco Albrecht
- Systems Biology Group, Life Science Research Unit, University of Luxembourg, 6, avenue du Swing, Belval, 4367 Luxembourg
| | - Philippe Lucarelli
- Systems Biology Group, Life Science Research Unit, University of Luxembourg, 6, avenue du Swing, Belval, 4367 Luxembourg
| | - Dagmar Kulms
- Experimental Dermatology, Department of Dermatology, Dresden University of Technology, Fetscherstraße 105, Dresden, 01307 Germany
| | - Thomas Sauter
- Systems Biology Group, Life Science Research Unit, University of Luxembourg, 6, avenue du Swing, Belval, 4367 Luxembourg
| |
Collapse
|
16
|
Coulibaly A, Bettendorf A, Kostina E, Figueiredo AS, Velásquez SY, Bock HG, Thiel M, Lindner HA, Barbarossa MV. Interleukin-15 Signaling in HIF-1α Regulation in Natural Killer Cells, Insights Through Mathematical Models. Front Immunol 2019; 10:2401. [PMID: 31681292 PMCID: PMC6805776 DOI: 10.3389/fimmu.2019.02401] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/25/2019] [Indexed: 12/17/2022] Open
Abstract
Natural killer (NK) cells belong to the first line of host defense against infection and cancer. Cytokines, including interleukin-15 (IL-15), critically regulate NK cell activity, resulting in recognition and direct killing of transformed and infected target cells. NK cells have to adapt and respond in inflamed and often hypoxic areas. Cellular stabilization and accumulation of the transcription factor hypoxia-inducible factor-1α (HIF-1α) is a key mechanism of the cellular hypoxia response. At the same time, HIF-1α plays a critical role in both innate and adaptive immunity. While the HIF-1α hydroxylation and degradation pathway has been recently described with the help of mathematical methods, less is known concerning the mechanistic mathematical description of processes regulating the levels of HIF-1α mRNA and protein. In this work we combine mathematical modeling with experimental laboratory analysis and examine the dynamic relationship between HIF-1α mRNA, HIF-1α protein, and IL-15-mediated upstream signaling events in NK cells from human blood. We propose a system of non-linear ordinary differential equations with positive and negative feedback loops for describing the complex interplay of HIF-1α regulators. The experimental design is optimized with the help of mathematical methods, and numerical optimization techniques yield reliable parameter estimates. The mathematical model allows for the investigation and prediction of HIF-1α stabilization under different inflammatory conditions and provides a better understanding of mechanisms mediating cellular enrichment of HIF-1α. Thanks to the combination of in vitro experimental data and in silico predictions we identified the mammalian target of rapamycin (mTOR), the nuclear factor-κB (NF-κB), and the signal transducer and activator of transcription 3 (STAT3) as central regulators of HIF-1α accumulation. We hypothesize that the regulatory pathway proposed here for NK cells can be extended to other types of immune cells. Understanding the molecular mechanisms involved in the dynamic regulation of the HIF-1α pathway in immune cells is of central importance to the immune cell function and could be a promising strategy in the design of treatments for human inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Anna Coulibaly
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Anja Bettendorf
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | - Ekaterina Kostina
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany.,Institute for Applied Mathematics, Heidelberg University, Heidelberg, Germany
| | - Ana Sofia Figueiredo
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Sonia Y Velásquez
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Hans-Georg Bock
- Interdisciplinary Center for Scientific Computing, Heidelberg University, Heidelberg, Germany
| | - Manfred Thiel
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Holger A Lindner
- Department of Anesthesiology and Surgical Intensive Care Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | | |
Collapse
|
17
|
Milberg O, Gong C, Jafarnejad M, Bartelink IH, Wang B, Vicini P, Narwal R, Roskos L, Popel AS. A QSP Model for Predicting Clinical Responses to Monotherapy, Combination and Sequential Therapy Following CTLA-4, PD-1, and PD-L1 Checkpoint Blockade. Sci Rep 2019; 9:11286. [PMID: 31375756 PMCID: PMC6677731 DOI: 10.1038/s41598-019-47802-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/24/2019] [Indexed: 01/12/2023] Open
Abstract
Over the past decade, several immunotherapies have been approved for the treatment of melanoma. The most prominent of these are the immune checkpoint inhibitors, which are antibodies that block the inhibitory effects on the immune system by checkpoint receptors, such as CTLA-4, PD-1 and PD-L1. Preclinically, blocking these receptors has led to increased activation and proliferation of effector cells following stimulation and antigen recognition, and subsequently, more effective elimination of cancer cells. Translation from preclinical to clinical outcomes in solid tumors has shown the existence of a wide diversity of individual patient responses, linked to several patient-specific parameters. We developed a quantitative systems pharmacology (QSP) model that looks at the mentioned checkpoint blockade therapies administered as mono-, combo- and sequential therapies, to show how different combinations of specific patient parameters defined within physiological ranges distinguish different types of virtual patient responders to these therapies for melanoma. Further validation by fitting and subsequent simulations of virtual clinical trials mimicking actual patient trials demonstrated that the model can capture a wide variety of tumor dynamics that are observed in the clinic and can predict median clinical responses. Our aim here is to present a QSP model for combination immunotherapy specific to melanoma.
Collapse
Affiliation(s)
- Oleg Milberg
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | - Chang Gong
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mohammad Jafarnejad
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Imke H Bartelink
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD), MedImmune, South San Francisco, California, USA.,Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bing Wang
- Clinical Pharmacology, Pharmacometrics and DMPK (CPD), MedImmune, South San Francisco, California, USA
| | - Paolo Vicini
- Clinical Pharmacology, Pharmacometrics and DMPK, MedImmune, Cambridge, United Kingdom
| | | | | | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
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: 28] [Impact Index Per Article: 5.6] [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
|
19
|
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: 3.2] [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
|
20
|
Mahlbacher GE, Reihmer KC, Frieboes HB. Mathematical modeling of tumor-immune cell interactions. J Theor Biol 2019; 469:47-60. [PMID: 30836073 DOI: 10.1016/j.jtbi.2019.03.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 02/14/2019] [Accepted: 03/01/2019] [Indexed: 12/22/2022]
Abstract
The anti-tumor activity of the immune system is increasingly recognized as critical for the mounting of a prolonged and effective response to cancer growth and invasion, and for preventing recurrence following resection or treatment. As the knowledge of tumor-immune cell interactions has advanced, experimental investigation has been complemented by mathematical modeling with the goal to quantify and predict these interactions. This succinct review offers an overview of recent tumor-immune continuum modeling approaches, highlighting spatial models. The focus is on work published in the past decade, incorporating one or more immune cell types and evaluating immune cell effects on tumor progression. Due to their relevance to cancer, the following immune cells and their combinations are described: macrophages, Cytotoxic T Lymphocytes, Natural Killer cells, dendritic cells, T regulatory cells, and CD4+ T helper cells. Although important insight has been gained from a mathematical modeling perspective, the development of models incorporating patient-specific data remains an important goal yet to be realized for potential clinical benefit.
Collapse
Affiliation(s)
| | - Kara C Reihmer
- Department of Bioengineering, University of Louisville, KY, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, KY, USA; Department of Pharmacology & Toxicology, University of Louisville, KY, USA.
| |
Collapse
|
21
|
Abstract
Cytokines that control the immune response were shown to have efficacy in preclinical murine cancer models. Interferon (IFN)-α is approved for treatment of hairy cell leukemia, and interleukin (IL)-2 for the treatment of advanced melanoma and metastatic renal cancer. In addition, IL-12, IL-15, IL-21, and granulocyte macrophage colony-stimulating factor (GM-CSF) have been evaluated in clinical trials. However, the cytokines as monotherapy have not fulfilled their early promise because cytokines administered parenterally do not achieve sufficient concentrations in the tumor, are often associated with severe toxicities, and induce humoral or cellular checkpoints. To circumvent these impediments, cytokines are being investigated clinically in combination therapy with checkpoint inhibitors, anticancer monoclonal antibodies to increase the antibody-dependent cellular cytotoxicity (ADCC) of these antibodies, antibody cytokine fusion proteins, and anti-CD40 to facilitate tumor-specific immune responses.
Collapse
Affiliation(s)
- Thomas A Waldmann
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Clinical Center, Bethesda, Maryland 20892-1374
| |
Collapse
|
22
|
Blazquez-Navarro A, Schachtner T, Stervbo U, Sefrin A, Stein M, Westhoff TH, Reinke P, Klipp E, Babel N, Neumann AU, Or-Guil M. Differential T cell response against BK virus regulatory and structural antigens: A viral dynamics modelling approach. PLoS Comput Biol 2018; 14:e1005998. [PMID: 29746472 PMCID: PMC5944912 DOI: 10.1371/journal.pcbi.1005998] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 01/24/2018] [Indexed: 12/26/2022] Open
Abstract
BK virus (BKV) associated nephropathy affects 1-10% of kidney transplant recipients, leading to graft failure in about 50% of cases. Immune responses against different BKV antigens have been shown to have a prognostic value for disease development. Data currently suggest that the structural antigens and regulatory antigens of BKV might each trigger a different mode of action of the immune response. To study the influence of different modes of action of the cellular immune response on BKV clearance dynamics, we have analysed the kinetics of BKV plasma load and anti-BKV T cell response (Elispot) in six patients with BKV associated nephropathy using ODE modelling. The results show that only a small number of hypotheses on the mode of action are compatible with the empirical data. The hypothesis with the highest empirical support is that structural antigens trigger blocking of virus production from infected cells, whereas regulatory antigens trigger an acceleration of death of infected cells. These differential modes of action could be important for our understanding of BKV resolution, as according to the hypothesis, only regulatory antigens would trigger a fast and continuous clearance of the viral load. Other hypotheses showed a lower degree of empirical support, but could potentially explain the clearing mechanisms of individual patients. Our results highlight the heterogeneity of the dynamics, including the delay between immune response against structural versus regulatory antigens, and its relevance for BKV clearance. Our modelling approach is the first that studies the process of BKV clearance by bringing together viral and immune kinetics and can provide a framework for personalised hypotheses generation on the interrelations between cellular immunity and viral dynamics.
Collapse
Affiliation(s)
- Arturo Blazquez-Navarro
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
- Systems Immunology Lab, Department of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Schachtner
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care, Charité-Universitätsmedizin, Berlin, Germany
| | - Ulrik Stervbo
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
- Medical Clinic I, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Anett Sefrin
- Department of Nephrology and Internal Intensive Care, Charité-Universitätsmedizin, Berlin, Germany
| | - Maik Stein
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
| | - Timm H Westhoff
- Medical Clinic I, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Petra Reinke
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care, Charité-Universitätsmedizin, Berlin, Germany
| | - Edda Klipp
- Theoretical Biophysics Group, Department of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nina Babel
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
- Medical Clinic I, Marien Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Avidan U Neumann
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin, Berlin, Germany
- Institute of Environmental Medicine, UNIKA-T, Helmholtz Zentrum München, Augsburg, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Michal Or-Guil
- Systems Immunology Lab, Department of Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
23
|
Ribba B, Boetsch C, Nayak T, Grimm HP, Charo J, Evers S, Klein C, Tessier J, Charoin JE, Phipps A, Pisa P, Teichgräber V. Prediction of the Optimal Dosing Regimen Using a Mathematical Model of Tumor Uptake for Immunocytokine-Based Cancer Immunotherapy. Clin Cancer Res 2018; 24:3325-3333. [PMID: 29463551 DOI: 10.1158/1078-0432.ccr-17-2953] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/05/2017] [Accepted: 02/14/2018] [Indexed: 11/16/2022]
Abstract
Purpose: Optimal dosing is critical for immunocytokine-based cancer immunotherapy to maximize efficacy and minimize toxicity. Cergutuzumab amunaleukin (CEA-IL2v) is a novel CEA-targeted immunocytokine. We set out to develop a mathematical model to predict intratumoral CEA-IL2v concentrations following various systemic dosing intensities.Experimental Design: Sequential measurements of CEA-IL2v plasma concentrations in 74 patients with solid tumors were applied in a series of differential equations to devise a model that also incorporates the peripheral concentrations of IL2 receptor-positive cell populations (i.e., CD8+, CD4+, NK, and B cells), which affect tumor bioavailability of CEA-IL2v. Imaging data from a subset of 14 patients were subsequently utilized to additionally predict antibody uptake in tumor tissues.Results: We created a pharmacokinetic/pharmacodynamic mathematical model that incorporates the expansion of IL2R-positive target cells at multiple dose levels and different schedules of CEA-IL2v. Model-based prediction of drug levels correlated with the concentration of IL2R-positive cells in the peripheral blood of patients. The pharmacokinetic model was further refined and extended by adding a model of antibody uptake, which is based on drug dose and the biological properties of the tumor. In silico predictions of our model correlated with imaging data and demonstrated that a dose-dense schedule comprising escalating doses and shortened intervals of drug administration can improve intratumoral drug uptake and overcome consumption of CEA-IL2v by the expanding population of IL2R-positive cells.Conclusions: The model presented here allows simulation of individualized treatment plans for optimal dosing and scheduling of immunocytokines for anticancer immunotherapy. Clin Cancer Res; 24(14); 3325-33. ©2018 AACRSee related commentary by Ruiz-Cerdá et al., p. 3236.
Collapse
Affiliation(s)
- Benjamin Ribba
- Pharmaceutical Sciences, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Basel, Switzerland.
| | - Christophe Boetsch
- Pharmaceutical Sciences, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Tapan Nayak
- Translational Imaging Science Oncology, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Hans Peter Grimm
- Pharmaceutical Sciences, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Jehad Charo
- Translational Medicine Oncology, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Stefan Evers
- Translational Medicine Oncology, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Christian Klein
- Discovery Oncology, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Jean Tessier
- Translational Imaging Science Oncology, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Jean Eric Charoin
- Pharmaceutical Sciences, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Basel, Switzerland
| | - Alex Phipps
- Pharmaceutical Sciences, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Welwyn, England
| | - Pavel Pisa
- Translational Medicine Oncology, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Zurich, Switzerland
| | - Volker Teichgräber
- Translational Medicine Oncology, Roche Pharmaceutical Research & Early Development, Roche Innovation Center, Zurich, Switzerland
| |
Collapse
|
24
|
Sharifi M, Jamshidi A, Sarvestani NN. An Adaptive Robust Control Strategy in a Cancer Tumor-Immune System under Uncertainties. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2018; 16:865-873. [PMID: 29994095 DOI: 10.1109/tcbb.2018.2803175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
We propose an adaptive robust control for a second order nonlinear model of the interaction between cancer and immune cells of the body to control the growth of cancer and maintain the number of immune cells in an appropriate level. Most of the control approaches are based on minimizing the drug dosage based on an optimal control structure. However, in many cases, measuring the exact quantity of the model parameters is not possible. This is due to limitation in measuring devices, variational and undetermined characteristics of micro-environmental factors. It is of great importance to present a control strategy that can deal with these unknown factors in a nonlinear model.
Collapse
|
25
|
Heidari N, Abroun S, Bertacchini J, Vosoughi T, Rahim F, Saki N. Significance of Inactivated Genes in Leukemia: Pathogenesis and Prognosis. CELL JOURNAL 2017; 19:9-26. [PMID: 28580304 PMCID: PMC5448318 DOI: 10.22074/cellj.2017.4908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/14/2017] [Indexed: 11/04/2022]
Abstract
Epigenetic and genetic alterations are two mechanisms participating in leukemia, which can inactivate genes involved in leukemia pathogenesis or progression. The purpose of this review was to introduce various inactivated genes and evaluate their possible role in leukemia pathogenesis and prognosis. By searching the mesh words "Gene, Silencing AND Leukemia" in PubMed website, relevant English articles dealt with human subjects as of 2000 were included in this study. Gene inactivation in leukemia is largely mediated by promoter's hypermethylation of gene involving in cellular functions such as cell cycle, apoptosis, and gene transcription. Inactivated genes, such as ASPP1, TP53, IKZF1 and P15, may correlate with poor prognosis in acute lymphoid leukemia (ALL), chronic lymphoid leukemia (CLL), chronic myelogenous leukemia (CML) and acute myeloid leukemia (AML), respectively. Gene inactivation may play a considerable role in leukemia pathogenesis and prognosis, which can be considered as complementary diagnostic tests to differentiate different leukemia types, determine leukemia prognosis, and also detect response to therapy. In general, this review showed some genes inactivated only in leukemia (with differences between B-ALL, T-ALL, CLL, AML and CML). These differences could be of interest as an additional tool to better categorize leukemia types. Furthermore; based on inactivated genes, a diverse classification of Leukemias could represent a powerful method to address a targeted therapy of the patients, in order to minimize side effects of conventional therapies and to enhance new drug strategies.
Collapse
Affiliation(s)
- Nazanin Heidari
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Abroun
- Department of Hematology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Jessika Bertacchini
- Signal Transduction Unit, Department of Surgery, Medicine, Dentistry and Morphology, University of Modena and Reggio Emilia, Modena, Italy
| | - Tina Vosoughi
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Fakher Rahim
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Health Research Institute, Thalassemia and Hemoglobinopathy Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
26
|
Ashcroft P, Smith CE, Garrod M, Galla T. Effects of population growth on the success of invading mutants. J Theor Biol 2017; 420:232-240. [DOI: 10.1016/j.jtbi.2017.03.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/28/2017] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
|
27
|
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: 2.5] [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
|
28
|
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: 87] [Impact Index Per Article: 10.9] [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
|
29
|
|
30
|
Abstract
Mathematical modelling approaches have become increasingly abundant in cancer research. The complexity of cancer is well suited to quantitative approaches as it provides challenges and opportunities for new developments. In turn, mathematical modelling contributes to cancer research by helping to elucidate mechanisms and by providing quantitative predictions that can be validated. The recent expansion of quantitative models addresses many questions regarding tumour initiation, progression and metastases as well as intra-tumour heterogeneity, treatment responses and resistance. Mathematical models can complement experimental and clinical studies, but also challenge current paradigms, redefine our understanding of mechanisms driving tumorigenesis and shape future research in cancer biology.
Collapse
Affiliation(s)
- Philipp M Altrock
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Department of Biostatistics, Harvard T.H. Chan School of Public Health, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
- Program for Evolutionary Dynamics, Harvard University, 1 Brattle Square, Suite 6, Cambridge, Massachusetts 02138, USA
| | - Lin L Liu
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Department of Biostatistics, Harvard T.H. Chan School of Public Health, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Franziska Michor
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Department of Biostatistics, Harvard T.H. Chan School of Public Health, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| |
Collapse
|
31
|
Lee L, Gupta M, Sahasranaman S. Immune Checkpoint inhibitors: An introduction to the next-generation cancer immunotherapy. J Clin Pharmacol 2015; 56:157-69. [PMID: 26183909 DOI: 10.1002/jcph.591] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 07/13/2015] [Indexed: 12/31/2022]
Abstract
Activating the immune system to eliminate cancer cells and produce clinically relevant responses has been a long-standing goal of cancer research. Most promising therapeutic approaches to activating antitumor immunity include immune checkpoint inhibitors. Immune checkpoints are numerous inhibitory pathways hardwired in the immune system. They are critical for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues to minimize collateral tissue damage. Tumors regulate certain immune checkpoint pathways as a major mechanism of immune resistance. Because immune checkpoints are initiated by ligand-receptor interactions, blockade by antibodies provides a rational therapeutic approach. Although targeted therapies are clinically successful, they are often short-lived due to rapid development of resistance. Immunotherapies offer one notable advantage. Enhancing the cell-mediated immune response against tumor cells leads to generation of a long-term memory lymphocyte population patrolling the body to attack growth of any new tumor cells, thereby sustaining the therapeutic effects. Furthermore, early clinical results suggest that combination immunotherapies offer even more potent antitumor activity. This review is intended to provide an introduction to immune checkpoint inhibitors and discusses the scientific overview of cancer immunotherapy, mechanisms of the inhibitors, clinical pharmacology considerations, advances in combination therapies, and challenges in drug development.
Collapse
Affiliation(s)
- Lucy Lee
- Clinical Pharmacology, Immunomedics Inc., Morris Plains, NJ, USA
| | - Manish Gupta
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, NJ, USA
| | | |
Collapse
|
32
|
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: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 09/17/2014] [Indexed: 12/26/2022]
|
33
|
Tabatabai MA, Eby WM, Singh KP, Bae S. T model of growth and its application in systems of tumor-immune dynamics. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2013; 10:925-938. [PMID: 23906156 PMCID: PMC4476034 DOI: 10.3934/mbe.2013.10.925] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
In this paper we introduce a new growth model called T growth model. This model is capable of representing sigmoidal growth as well as biphasic growth. This dual capability is achieved without introducing additional parameters. The T model is useful in modeling cellular proliferation or regression of cancer cells, stem cells, bacterial growth and drug dose-response relationships. We recommend usage of the T growth model for the growth of tumors as part of any system of differential equations. Use of this model within a system will allow more flexibility in representing the natural rate of tumor growth. For illustration, we examine some systems of tumor-immune interaction in which the T growth rate is applied. We also apply the model to a set of tumor growth data.
Collapse
Affiliation(s)
| | - Wayne M. Eby
- Department of Mathematical Sciences, Cameron University, Lawton, OK 73505, USA
| | - Karan P. Singh
- School of Medicine, University of Alabama at Birmingham, Birmingham AL 35294, USA
| | - Sejong Bae
- School of Medicine, University of Alabama at Birmingham, Birmingham AL 35294, USA
| |
Collapse
|
34
|
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: 2.1] [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
|
35
|
DePillis L, Gallegos A, Radunskaya A. A model of dendritic cell therapy for melanoma. Front Oncol 2013; 3:56. [PMID: 23516248 PMCID: PMC3601335 DOI: 10.3389/fonc.2013.00056] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 03/02/2013] [Indexed: 11/25/2022] Open
Abstract
Dendritic cells are a promising immunotherapy tool for boosting an individual's antigen-specific immune response to cancer. We develop a mathematical model using differential and delay-differential equations to describe the interactions between dendritic cells, effector-immune cells, and tumor cells. We account for the trafficking of immune cells between lymph, blood, and tumor compartments. Our model reflects experimental results both for dendritic cell trafficking and for immune suppression of tumor growth in mice. In addition, in silico experiments suggest more effective immunotherapy treatment protocols can be achieved by modifying dose location and schedule. A sensitivity analysis of the model reveals which patient-specific parameters have the greatest impact on treatment efficacy.
Collapse
Affiliation(s)
- Lisette DePillis
- Department of Mathematics, Harvey Mudd CollegeClaremont, CA, USA
| | - Angela Gallegos
- Department of Mathematics, Loyola Marymount UniversityLos Angeles, CA, USA
| | - Ami Radunskaya
- Department of Mathematics, Pomona College, Claremont CollegesClaremont, CA, USA
| |
Collapse
|
36
|
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]
|
37
|
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]
|
38
|
Watzl C, Sternberg-Simon M, Urlaub D, Mehr R. Understanding natural killer cell regulation by mathematical approaches. Front Immunol 2012; 3:359. [PMID: 23264774 PMCID: PMC3525018 DOI: 10.3389/fimmu.2012.00359] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Accepted: 11/10/2012] [Indexed: 11/13/2022] Open
Abstract
The activity of natural killer (NK) cells is regulated by various processes including education/licensing, priming, integration of positive and negative signals through an array of activating and inhibitory receptors, and the development of memory-like functionality. These processes are often very complex due to the large number of different receptors and signaling pathways involved. Understanding these complex mechanisms is therefore a challenge, but is critical for understanding NK cell regulation. Mathematical approaches can facilitate the analysis and understanding of complex systems. Therefore, they may be instrumental for studies in NK cell biology. Here we provide a review of the different mathematical approaches to the analysis of NK cell signal integration, activation, proliferation, and the acquisition of inhibitory receptors. These studies show how mathematical methods can aid the analysis of NK cell regulation.
Collapse
Affiliation(s)
- Carsten Watzl
- IfADo - Leibniz Institute for Occupational Research Dortmund, Germany
| | | | | | | |
Collapse
|
39
|
Caravagna G, Barbuti R, d'Onofrio A. Fine-tuning anti-tumor immunotherapies via stochastic simulations. BMC Bioinformatics 2012; 13 Suppl 4:S8. [PMID: 22536975 PMCID: PMC3303725 DOI: 10.1186/1471-2105-13-s4-s8] [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] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Anti-tumor therapies aim at reducing to zero the number of tumor cells in a host within their end or, at least, aim at leaving the patient with a sufficiently small number of tumor cells so that the residual tumor can be eradicated by the immune system. Besides severe side-effects, a key problem of such therapies is finding a suitable scheduling of their administration to the patients. In this paper we study the effect of varying therapy-related parameters on the final outcome of the interplay between a tumor and the immune system. RESULTS This work generalizes our previous study on hybrid models of such an interplay where interleukins are modeled as a continuous variable, and the tumor and the immune system as a discrete-state continuous-time stochastic process. The hybrid model we use is obtained by modifying the corresponding deterministic model, originally proposed by Kirschner and Panetta. We consider Adoptive Cellular Immunotherapies and Interleukin-based therapies, as well as their combination. By asymptotic and transitory analyses of the corresponding deterministic model we find conditions guaranteeing tumor eradication, and we tune the parameters of the hybrid model accordingly. We then perform stochastic simulations of the hybrid model under various therapeutic settings: constant, piece-wise constant or impulsive infusion and daily or weekly delivery schedules. CONCLUSIONS Results suggest that, in some cases, the delivery schedule may deeply impact on the therapy-induced tumor eradication time. Indeed, our model suggests that Interleukin-based therapies may not be effective for every patient, and that the piece-wise constant is the most effective delivery to stimulate the immune-response. For Adoptive Cellular Immunotherapies a metronomic delivery seems more effective, as it happens for other anti-angiogenesis therapies and chemotherapies, and the impulsive delivery seems more effective than the piece-wise constant. The expected synergistic effects have been observed when the therapies are combined.
Collapse
Affiliation(s)
- Giulio Caravagna
- Institute for Informatics and Telematics, National Research Council, Pisa, Italy
| | | | | |
Collapse
|
40
|
Wilson S, Levy D. A mathematical model of the enhancement of tumor vaccine efficacy by immunotherapy. Bull Math Biol 2012; 74:1485-500. [PMID: 22438084 DOI: 10.1007/s11538-012-9722-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Accepted: 03/01/2012] [Indexed: 11/28/2022]
Abstract
TGF-β is an immunoregulatory protein that contributes to inadequate antitumor immune responses in cancer patients. Recent experimental data suggests that TGF-β inhibition alone, provides few clinical benefits, yet it can significantly amplify the anti-tumor immune response when combined with a tumor vaccine. We develop a mathematical model in order to gain insight into the cooperative interaction between anti-TGF-β and vaccine treatments. The mathematical model follows the dynamics of the tumor size, TGF-β concentration, activated cytotoxic effector cells, and regulatory T cells. Using numerical simulations and stability analysis, we study the following scenarios: a control case of no treatment, anti-TGF-β treatment, vaccine treatment, and combined anti-TGF-β vaccine treatments. We show that our model is capable of capturing the observed experimental results, and hence can be potentially used in designing future experiments involving this approach to immunotherapy.
Collapse
Affiliation(s)
- Shelby Wilson
- Department of Mathematics and Center for Scientific Computation and Mathematical Modeling (CSCAMM), University of Maryland, College Park, MD 20742, USA.
| | | |
Collapse
|
41
|
Reconsidering the Paradigm of Cancer Immunotherapy by Computationally Aided Real-time Personalization. Cancer Res 2012; 72:2218-27. [DOI: 10.1158/0008-5472.can-11-4166] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
42
|
Elishmereni M, Kheifetz Y, Søndergaard H, Overgaard RV, Agur Z. An integrated disease/pharmacokinetic/pharmacodynamic model suggests improved interleukin-21 regimens validated prospectively for mouse solid cancers. PLoS Comput Biol 2011; 7:e1002206. [PMID: 22022259 PMCID: PMC3182868 DOI: 10.1371/journal.pcbi.1002206] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2011] [Accepted: 08/08/2011] [Indexed: 11/20/2022] Open
Abstract
Interleukin (IL)-21 is an attractive antitumor agent with potent immunomodulatory functions. Yet thus far, the cytokine has yielded only partial responses in solid cancer patients, and conditions for beneficial IL-21 immunotherapy remain elusive. The current work aims to identify clinically-relevant IL-21 regimens with enhanced efficacy, based on mathematical modeling of long-term antitumor responses. For this purpose, pharmacokinetic (PK) and pharmacodynamic (PD) data were acquired from a preclinical study applying systemic IL-21 therapy in murine solid cancers. We developed an integrated disease/PK/PD model for the IL-21 anticancer response, and calibrated it using selected “training” data. The accuracy of the model was verified retrospectively under diverse IL-21 treatment settings, by comparing its predictions to independent “validation” data in melanoma and renal cell carcinoma-challenged mice (R2>0.90). Simulations of the verified model surfaced important therapeutic insights: (1) Fractionating the standard daily regimen (50 µg/dose) into a twice daily schedule (25 µg/dose) is advantageous, yielding a significantly lower tumor mass (45% decrease); (2) A low-dose (12 µg/day) regimen exerts a response similar to that obtained under the 50 µg/day treatment, suggestive of an equally efficacious dose with potentially reduced toxicity. Subsequent experiments in melanoma-bearing mice corroborated both of these predictions with high precision (R2>0.89), thus validating the model also prospectively in vivo. Thus, the confirmed PK/PD model rationalizes IL-21 therapy, and pinpoints improved clinically-feasible treatment schedules. Our analysis demonstrates the value of employing mathematical modeling and in silico-guided design of solid tumor immunotherapy in the clinic. Among the many potential drugs explored within the scope of cancer immunotherapy are selected cytokines which possess promising immune-boosting properties. Yet, the natural involvement of these proteins in multiple, often contradicting biological processes can complicate their use in the clinic. The cytokine interleukin (IL)-21 is no exception: while its strength as an anticancer agent has been established in several animal studies, response rates in melanoma and renal cell carcinoma patients remain low. To help guide the design of effective IL-21 therapy, we have developed a mathematical model that bridges between the complex biology of IL-21 and its optimal clinical use. Our model integrates data from preclinical studies under diverse IL-21 treatment settings, and was validated by extensive experiments in tumor-bearing mice. Model simulations predicted that beneficial, clinically practical IL-21 therapy should be composed of low-dose schedules, and/or schedules in which several partial doses are administered rather than a single complete dose. These findings were subsequently confirmed in mice with melanoma. Thus, future testing of these strategies in solid cancer patients can be a promising starting point for improving IL-21 therapy. Our model can thus provide a computational platform for rationalizing IL-21 regimens and streamlining its clinical development.
Collapse
Affiliation(s)
| | - Yuri Kheifetz
- Institute for Medical Biomathematics (IMBM), Bene-Ataroth, Israel
| | | | | | - Zvia Agur
- Institute for Medical Biomathematics (IMBM), Bene-Ataroth, Israel
- Optimata Ltd., Ramat-Gan, Israel
- * E-mail:
| |
Collapse
|
43
|
A mathematical model of combined bacillus Calmette-Guerin (BCG) and interleukin (IL)-2 immunotherapy of superficial bladder cancer. J Theor Biol 2011; 277:27-40. [DOI: 10.1016/j.jtbi.2011.02.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Revised: 02/08/2011] [Accepted: 02/08/2011] [Indexed: 01/01/2023]
|
44
|
Owen MR, Stamper IJ, Muthana M, Richardson GW, Dobson J, Lewis CE, Byrne HM. Mathematical modeling predicts synergistic antitumor effects of combining a macrophage-based, hypoxia-targeted gene therapy with chemotherapy. Cancer Res 2011; 71:2826-37. [PMID: 21363914 DOI: 10.1158/0008-5472.can-10-2834] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Tumor hypoxia is associated with low rates of cell proliferation and poor drug delivery, limiting the efficacy of many conventional therapies such as chemotherapy. Because many macrophages accumulate in hypoxic regions of tumors, one way to target tumor cells in these regions could be to use genetically engineered macrophages that express therapeutic genes when exposed to hypoxia. Systemic delivery of such therapeutic macrophages may also be enhanced by preloading them with nanomagnets and applying a magnetic field to the tumor site. Here, we use a new mathematical model to compare the effects of conventional cyclophosphamide therapy with those induced when macrophages are used to deliver hypoxia-inducible cytochrome P450 to locally activate cyclophosphamide. Our mathematical model describes the spatiotemporal dynamics of vascular tumor growth and treats cells as distinct entities. Model simulations predict that combining conventional and macrophage-based therapies would be synergistic, producing greater antitumor effects than the additive effects of each form of therapy. We find that timing is crucial in this combined approach with efficacy being greatest when the macrophage-based, hypoxia-targeted therapy is administered shortly before or concurrently with chemotherapy. Last, we show that therapy with genetically engineered macrophages is markedly enhanced by using the magnetic approach described above, and that this enhancement depends mainly on the strength of the applied field, rather than its direction. This insight may be important in the treatment of nonsuperficial tumors, where generating a specific orientation of a magnetic field may prove difficult. In conclusion, we demonstrate that mathematical modeling can be used to design and maximize the efficacy of combined therapeutic approaches in cancer.
Collapse
Affiliation(s)
- Markus R Owen
- Centre for Mathematical Medicine and Biology, School of Mathematical Sciences, University of Nottingham, Nottingham, UK.
| | | | | | | | | | | | | |
Collapse
|
45
|
Theoretical modeling techniques and their impact on tumor immunology. Clin Dev Immunol 2010; 2010:271794. [PMID: 21234354 PMCID: PMC3018070 DOI: 10.1155/2010/271794] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 10/10/2010] [Accepted: 10/11/2010] [Indexed: 01/19/2023]
Abstract
Currently, cancer is one of the leading causes of death in industrial nations. While conventional cancer treatment usually results in the patient suffering from severe side effects, immunotherapy is a promising alternative. Nevertheless, some questions remain unanswered with regard to using immunotherapy to treat cancer hindering it from being widely established. To help rectify this deficit in knowledge, experimental data, accumulated from a huge number of different studies, can be integrated into theoretical models of the tumor-immune system interaction. Many complex mechanisms in immunology and oncology cannot be measured in experiments, but can be analyzed by mathematical simulations. Using theoretical modeling techniques, general principles of tumor-immune system interactions can be explored and clinical treatment schedules optimized to lower both tumor burden and side effects. In this paper, we aim to explain the main mathematical and computational modeling techniques used in tumor immunology to experimental researchers and clinicians. In addition, we review relevant published work and provide an overview of its impact to the field.
Collapse
|
46
|
Kronik N, Kogan Y, Elishmereni M, Halevi-Tobias K, Vuk-Pavlović S, Agur Z. Predicting outcomes of prostate cancer immunotherapy by personalized mathematical models. PLoS One 2010; 5:e15482. [PMID: 21151630 PMCID: PMC2999571 DOI: 10.1371/journal.pone.0015482] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2010] [Accepted: 09/23/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Therapeutic vaccination against disseminated prostate cancer (PCa) is partially effective in some PCa patients. We hypothesized that the efficacy of treatment will be enhanced by individualized vaccination regimens tailored by simple mathematical models. METHODOLOGY/PRINCIPAL FINDINGS We developed a general mathematical model encompassing the basic interactions of a vaccine, immune system and PCa cells, and validated it by the results of a clinical trial testing an allogeneic PCa whole-cell vaccine. For model validation in the absence of any other pertinent marker, we used the clinically measured changes in prostate-specific antigen (PSA) levels as a correlate of tumor burden. Up to 26 PSA levels measured per patient were divided into each patient's training set and his validation set. The training set, used for model personalization, contained the patient's initial sequence of PSA levels; the validation set contained his subsequent PSA data points. Personalized models were simulated to predict changes in tumor burden and PSA levels and predictions were compared to the validation set. The model accurately predicted PSA levels over the entire measured period in 12 of the 15 vaccination-responsive patients (the coefficient of determination between the predicted and observed PSA values was R(2) = 0.972). The model could not account for the inconsistent changes in PSA levels in 3 of the 15 responsive patients at the end of treatment. Each validated personalized model was simulated under many hypothetical immunotherapy protocols to suggest alternative vaccination regimens. Personalized regimens predicted to enhance the effects of therapy differed among the patients. CONCLUSIONS/SIGNIFICANCE Using a few initial measurements, we constructed robust patient-specific models of PCa immunotherapy, which were retrospectively validated by clinical trial results. Our results emphasize the potential value and feasibility of individualized model-suggested immunotherapy protocols.
Collapse
Affiliation(s)
- Natalie Kronik
- Institute for Medical BioMathematics, Bene Ataroth, Israel
| | - Yuri Kogan
- Institute for Medical BioMathematics, Bene Ataroth, Israel
| | | | | | | | - Zvia Agur
- Institute for Medical BioMathematics, Bene Ataroth, Israel
| |
Collapse
|
47
|
Gilert A, Machluf M. Nano to micro delivery systems: targeting angiogenesis in brain tumors. JOURNAL OF ANGIOGENESIS RESEARCH 2010; 2:20. [PMID: 20932320 PMCID: PMC2964525 DOI: 10.1186/2040-2384-2-20] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 10/08/2010] [Indexed: 01/09/2023]
Abstract
Treating brain tumors using inhibitors of angiogenesis is extensively researched and tested in clinical trials. Although anti-angiogenic treatment holds a great potential for treating primary and secondary brain tumors, no clinical treatment is currently approved for brain tumor patients. One of the main hurdles in treating brain tumors is the blood brain barrier - a protective barrier of the brain, which prevents drugs from entering the brain parenchyma. As most therapeutics are excluded from the brain there is an urgent need to develop delivery platforms which will bypass such hurdles and enable the delivery of anti-angiogenic drugs into the tumor bed. Such delivery systems should be able to control release the drug or a combination of drugs at a therapeutic level for the desired time. In this mini-review we will discuss the latest improvements in nano and micro drug delivery platforms that were designed to deliver inhibitors of angiogenesis to the brain.
Collapse
Affiliation(s)
- Ariel Gilert
- Faculty of Biotechnology and Food Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| | | |
Collapse
|
48
|
Caravagna G, d’Onofrio A, Milazzo P, Barbuti R. Tumour suppression by immune system through stochastic oscillations. J Theor Biol 2010; 265:336-45. [DOI: 10.1016/j.jtbi.2010.05.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 05/05/2010] [Accepted: 05/08/2010] [Indexed: 10/19/2022]
|
49
|
Agur Z. From the evolution of toxin resistance to virtual clinical trials: the role of mathematical models in oncology. Future Oncol 2010; 6:917-27. [DOI: 10.2217/fon.10.61] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Sustainable delivery of quality healthcare at affordable cost is a major challenge, especially in oncology. Through interdisciplinary collaboration, researchers can provide new insights into familiar concepts and radically change the ways in which biopharmaceutical and medical studies are conducted and translated into clinical practice. One interdisciplinary approach is ‘Virtual R&D’, that is, biomedical research and development aided by mathematical models of the human body. This approach has facilitated the development of the ‘virtual patient’, a clinically validated modeling system that accurately predicts efficacy and toxicity of various oncology drug combinations in individuals and in populations. The use of virtual patients in clinical research will dramatically shorten the period of development of new drugs. Such models can also be used to personalize treatment regimens, substantially reducing the risk of clinical failure. This strategy can improve the quality of healthcare and reduce costs associated with clinical development.
Collapse
Affiliation(s)
- Zvia Agur
- Institute for Medical BioMathematics (IMBM), PO Box 282, Hateena Street, 10 Bene Ataroth, 60991, Israel
- Optimata Ltd, Aba Hillel Street, 7 Ramat-Gan 52522, Israel
| |
Collapse
|
50
|
Eftimie R, Bramson JL, Earn DJD. Interactions between the immune system and cancer: a brief review of non-spatial mathematical models. Bull Math Biol 2010; 73:2-32. [PMID: 20225137 DOI: 10.1007/s11538-010-9526-3] [Citation(s) in RCA: 174] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Accepted: 02/18/2010] [Indexed: 12/14/2022]
Abstract
We briefly review spatially homogeneous mechanistic mathematical models describing the interactions between a malignant tumor and the immune system. We begin with the simplest (single equation) models for tumor growth and proceed to consider greater immunological detail (and correspondingly more equations) in steps. This approach allows us to clarify the necessity for expanding the complexity of models in order to capture the biological mechanisms we wish to understand. We conclude by discussing some unsolved problems in the mathematical modeling of cancer-immune system interactions.
Collapse
Affiliation(s)
- Raluca Eftimie
- Department of Mathematics and Statistic, McMaster University, Hamilton, ON, Canada, L8S 4K1.
| | | | | |
Collapse
|