51
|
Integrative Approaches to Cancer Immunotherapy. Trends Cancer 2020; 5:400-410. [PMID: 31311655 DOI: 10.1016/j.trecan.2019.05.010] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 05/17/2019] [Accepted: 05/30/2019] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy aims to arm patients with cancer-fighting immunity. Many new cancer-specific immunotherapeutic drugs have gained approval in the past several years, demonstrating immunotherapy's efficacy and promise as an anticancer modality. Despite these successes, several outstanding questions remain for cancer immunotherapy, including how to make immunotherapy more efficacious in a broader range of cancer types and patients, and how to predict which patients will respond or not respond to therapy. We present a case for integrative systems approaches that will answer these questions. This involves applying mechanistic and statistical modeling, establishing consistent and widely adopted experimental tools to generate systems-level data, and creating sustained mechanisms of support. If implemented, these approaches will lead to major advances in cancer treatment.
Collapse
|
52
|
Yu JS, Bagheri N. Agent-Based Models Predict Emergent Behavior of Heterogeneous Cell Populations in Dynamic Microenvironments. Front Bioeng Biotechnol 2020; 8:249. [PMID: 32596213 PMCID: PMC7301008 DOI: 10.3389/fbioe.2020.00249] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/10/2020] [Indexed: 01/18/2023] Open
Abstract
Computational models are most impactful when they explain and characterize biological phenomena that are non-intuitive, unexpected, or difficult to study experimentally. Countless equation-based models have been built for these purposes, but we have yet to realize the extent to which rules-based models offer an intuitive framework that encourages computational and experimental collaboration. We develop ARCADE, a multi-scale agent-based model to interrogate emergent behavior of heterogeneous cell agents within dynamic microenvironments and demonstrate how complexity of intracellular metabolism and signaling modules impacts emergent dynamics. We perform in silico case studies on context, competition, and heterogeneity to demonstrate the utility of our model for gaining computational and experimental insight. Notably, there exist (i) differences in emergent behavior between colony and tissue contexts, (ii) linear, non-linear, and multimodal consequences of parameter variation on competition in simulated co-cultures, and (iii) variable impact of cell and population heterogeneity on emergent outcomes. Our extensible framework is easily modified to explore numerous biological systems, from tumor microenvironments to microbiomes.
Collapse
Affiliation(s)
- Jessica S Yu
- Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States
| | - Neda Bagheri
- Chemical and Biological Engineering, Northwestern University, Evanston, IL, United States.,Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL, United States.,Center for Synthetic Biology, Northwestern University, Evanston, IL, United States.,Biology, University of Washington, Seattle, WA, United States.,Chemical Engineering, University of Washington, Seattle, WA, United States
| |
Collapse
|
53
|
Stalidzans E, Zanin M, Tieri P, Castiglione F, Polster A, Scheiner S, Pahle J, Stres B, List M, Baumbach J, Lautizi M, Van Steen K, Schmidt HH. Mechanistic Modeling and Multiscale Applications for Precision Medicine: Theory and Practice. NETWORK AND SYSTEMS MEDICINE 2020. [DOI: 10.1089/nsm.2020.0002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Egils Stalidzans
- Computational Systems Biology Group, University of Latvia, Riga, Latvia
- Latvian Biomedical Reasearch and Study Centre, Riga, Latvia
| | - Massimiliano Zanin
- Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Pozuelo de Alarcón, Spain
| | - Paolo Tieri
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | - Filippo Castiglione
- CNR National Research Council, IAC Institute for Applied Computing, Rome, Italy
| | | | - Stefan Scheiner
- Institute for Mechanics of Materials and Structures, Vienna University of Technology, Vienna, Austria
| | - Jürgen Pahle
- BioQuant, Heidelberg University, Heidelberg, Germany
| | - Blaž Stres
- Department of Animal Science, University of Ljubljana, Ljubljana, Slovenia
- Faculty of Civil and Geodetic Engineering, University of Ljubljana, Ljubljana, Slovenia
- Department of Automation, Biocybernetics and Robotics, Jozef Stefan Institute, Ljubljana, Slovenia
| | - Markus List
- Big Data in BioMedicine Research Group, Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Jan Baumbach
- Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Manuela Lautizi
- Computational Systems Medicine Research Group, Chair of Experimental Bioinformatics, TUM School of Weihenstephan, Technical University of Munich, Freising, Germany
| | - Kristel Van Steen
- BIO-Systems Genetics, GIGA-R, University of Liège, Liège, Belgium
- BIO3—Systems Medicine, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Harald H.H.W. Schmidt
- Department of Pharmacology and Personalised Medicine, Faculty of Health, Medicine and Life Science, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
54
|
Fullstone G, Guttà C, Beyer A, Rehm M. The FLAME-accelerated signalling tool (FaST) for facile parallelisation of flexible agent-based models of cell signalling. NPJ Syst Biol Appl 2020; 6:10. [PMID: 32313030 PMCID: PMC7170865 DOI: 10.1038/s41540-020-0128-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/17/2020] [Indexed: 11/18/2022] Open
Abstract
Agent-based modelling is particularly adept at modelling complex features of cell signalling pathways, where heterogeneity, stochastic and spatial effects are important, thus increasing our understanding of decision processes in biology in such scenarios. However, agent-based modelling often is computationally prohibitive to implement. Parallel computing, either on central processing units (CPUs) or graphical processing units (GPUs), can provide a means to improve computational feasibility of agent-based applications but generally requires specialist coding knowledge and extensive optimisation. In this paper, we address these challenges through the development and implementation of the FLAME-accelerated signalling tool (FaST), a software that permits easy creation and parallelisation of agent-based models of cell signalling, on CPUs or GPUs. FaST incorporates validated new agent-based methods, for accurate modelling of reaction kinetics and, as proof of concept, successfully converted an ordinary differential equation (ODE) model of apoptosis execution into an agent-based model. We finally parallelised this model through FaST on CPUs and GPUs resulting in an increase in performance of 5.8× (16 CPUs) and 53.9×, respectively. The FaST takes advantage of the communicating X-machine approach used by FLAME and FLAME GPU to allow easy alteration or addition of functionality to parallel applications, but still includes inherent parallelisation optimisation. The FaST, therefore, represents a new and innovative tool to easily create and parallelise bespoke, robust, agent-based models of cell signalling.
Collapse
Affiliation(s)
- Gavin Fullstone
- Institute for Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany. .,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Nobelstrasse 15, 70569, Stuttgart, Germany.
| | - Cristiano Guttà
- Institute for Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Amatus Beyer
- Institute for Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Markus Rehm
- Institute for Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany. .,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Nobelstrasse 15, 70569, Stuttgart, Germany.
| |
Collapse
|
55
|
Alsassa S, Lefèvre T, Laugier V, Stindel E, Ansart S. Modeling Early Stages of Bone and Joint Infections Dynamics in Humans: A Multi-Agent, Multi-System Based Model. Front Mol Biosci 2020; 7:26. [PMID: 32226790 PMCID: PMC7080862 DOI: 10.3389/fmolb.2020.00026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 02/07/2020] [Indexed: 11/13/2022] Open
Abstract
Diagnosis and management of bone and joint infections (BJI) is a challenging task. The high intra and inter patient's variability in terms of clinical presentation makes it impossible to rely on a systematic description or classical statistical analysis for its diagnosis. Advances can be achieved through a better understanding of the system behavior that results from the interactions between the components at a micro-scale level, which is difficult to mastered using traditional methods. Multiple studies from the literature report factors and interactions that affect the dynamics of the BJI system. The objectives of this study were (i) to perform a systematic review to identify relevant interactions between agents (cells, pathogens) and parameters values that characterize agents and interactions, and (ii) to develop a two dimensional computational model of the BJI system based on the results of the systematic review. The model would simulate the behavior resulting from the interactions on the cellular and molecular levels to explore the BJI dynamics, using an agent-based modeling approach. The BJI system's response to different microbial inoculum levels was simulated. The model succeeded in mimicking the dynamics of bacteria, the innate immune cells, and the bone mass during the first stage of infection and for different inoculum levels in a consistent manner. The simulation displayed the destruction in bone tissue as a result of the alteration in bone remodeling process during the infection. The model was used to generate different patterns of system behaviors that could be analyzed in further steps. Simulations results suggested evidence for the existence of latent infections. Finally, we presented a way to analyze and synthesize massive simulated data in a concise and comprehensive manner based on the semi-supervised identification of ordinary differential equations (ODE) systems. It allows to use the known framework for temporal and structural ODE analyses and therefore summarize the whole simulated system dynamical behavior. This first model is intended to be validated by in vivo or in vitro data and expected to generate hypotheses to be challenged by real data. Step by step, it can be modified and complexified based on the test/validation iteration cycles.
Collapse
Affiliation(s)
- Salma Alsassa
- Laboratory of Medical Information Processing (LaTIM - UMR 1101 INSERM), IBRS, Université de Bretagne Occidentale, Department of Medicine, Brest, France
- Tekliko SARL, Paris, France
| | - Thomas Lefèvre
- Iris UMR 8156 CNRS - U997 Inserm - EHESS - UP 13, Paris, France
- AP-HP, Jean Verdier Teaching Hospital, Department of Legal and Social Medicine, Bondy, France
| | | | - Eric Stindel
- Laboratory of Medical Information Processing (LaTIM - UMR 1101 INSERM), IBRS, Université de Bretagne Occidentale, Department of Medicine, Brest, France
- La Cavale Blanche University Hospital, Infection Diseases Unit, Brest, France
| | - Séverine Ansart
- Laboratory of Medical Information Processing (LaTIM - UMR 1101 INSERM), IBRS, Université de Bretagne Occidentale, Department of Medicine, Brest, France
- La Cavale Blanche University Hospital, Infection Diseases Unit, Brest, France
| |
Collapse
|
56
|
Letort G, Montagud A, Stoll G, Heiland R, Barillot E, Macklin P, Zinovyev A, Calzone L. PhysiBoSS: a multi-scale agent-based modelling framework integrating physical dimension and cell signalling. Bioinformatics 2020; 35:1188-1196. [PMID: 30169736 PMCID: PMC6449758 DOI: 10.1093/bioinformatics/bty766] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 07/28/2018] [Accepted: 08/30/2018] [Indexed: 01/22/2023] Open
Abstract
MOTIVATION Due to the complexity and heterogeneity of multicellular biological systems, mathematical models that take into account cell signalling, cell population behaviour and the extracellular environment are particularly helpful. We present PhysiBoSS, an open source software which combines intracellular signalling using Boolean modelling (MaBoSS) and multicellular behaviour using agent-based modelling (PhysiCell). RESULTS PhysiBoSS provides a flexible and computationally efficient framework to explore the effect of environmental and genetic alterations of individual cells at the population level, bridging the critical gap from single-cell genotype to single-cell phenotype and emergent multicellular behaviour. PhysiBoSS thus becomes very useful when studying heterogeneous population response to treatment, mutation effects, different modes of invasion or isomorphic morphogenesis events. To concretely illustrate a potential use of PhysiBoSS, we studied heterogeneous cell fate decisions in response to TNF treatment. We explored the effect of different treatments and the behaviour of several resistant mutants. We highlighted the importance of spatial information on the population dynamics by considering the effect of competition for resources like oxygen. AVAILABILITY AND IMPLEMENTATION PhysiBoSS is freely available on GitHub (https://github.com/sysbio-curie/PhysiBoSS), with a Docker image (https://hub.docker.com/r/gletort/physiboss/). It is distributed as open source under the BSD 3-clause license. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Gaelle Letort
- Institut Curie, PSL Research University, Paris, France.,INSERM, U900, Paris, France.,CBIO-Centre for Computational Biology, MINES ParisTech, PSL Research University, Paris, France
| | - Arnau Montagud
- Institut Curie, PSL Research University, Paris, France.,INSERM, U900, Paris, France.,CBIO-Centre for Computational Biology, MINES ParisTech, PSL Research University, Paris, France
| | - Gautier Stoll
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France.,Gustave Roussy Cancer Campus, Villejuif, France.,INSERM, U1138, Paris, France.,Equipe 11 Labellisée par la Ligue Nationale Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
| | - Randy Heiland
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Paris, France.,INSERM, U900, Paris, France.,CBIO-Centre for Computational Biology, MINES ParisTech, PSL Research University, Paris, France
| | - Paul Macklin
- Intelligent Systems Engineering, Indiana University, Bloomington, IN, USA
| | - Andrei Zinovyev
- Institut Curie, PSL Research University, Paris, France.,INSERM, U900, Paris, France.,CBIO-Centre for Computational Biology, MINES ParisTech, PSL Research University, Paris, France
| | - Laurence Calzone
- Institut Curie, PSL Research University, Paris, France.,INSERM, U900, Paris, France.,CBIO-Centre for Computational Biology, MINES ParisTech, PSL Research University, Paris, France
| |
Collapse
|
57
|
Verma M, Bassaganya-Riera J, Leber A, Tubau-Juni N, Hoops S, Abedi V, Chen X, Hontecillas R. High-resolution computational modeling of immune responses in the gut. Gigascience 2020; 8:5513894. [PMID: 31185494 PMCID: PMC6559340 DOI: 10.1093/gigascience/giz062] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 01/19/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023] Open
Abstract
Background Helicobacter pylori causes gastric cancer in 1–2% of cases but is also beneficial for protection against allergies and gastroesophageal diseases. An estimated 85% of H. pylori–colonized individuals experience no detrimental effects. To study the mechanisms promoting host tolerance to the bacterium in the gastrointestinal mucosa and systemic regulatory effects, we investigated the dynamics of immunoregulatory mechanisms triggered by H. pylori using a high-performance computing–driven ENteric Immunity SImulator multiscale model. Immune responses were simulated by integrating an agent-based model, ordinary, and partial differential equations. Results The outputs were analyzed using 2 sequential stages: the first used a partial rank correlation coefficient regression–based and the second a metamodel-based global sensitivity analysis. The influential parameters screened from the first stage were selected to be varied for the second stage. The outputs from both stages were combined as a training dataset to build a spatiotemporal metamodel. The Sobol indices measured time-varying impact of input parameters during initiation, peak, and chronic phases of infection. The study identified epithelial cell proliferation and epithelial cell death as key parameters that control infection outcomes. In silico validation showed that colonization with H. pylori decreased with a decrease in epithelial cell proliferation, which was linked to regulatory macrophages and tolerogenic dendritic cells. Conclusions The hybrid model of H. pylori infection identified epithelial cell proliferation as a key factor for successful colonization of the gastric niche and highlighted the role of tolerogenic dendritic cells and regulatory macrophages in modulating the host responses and shaping infection outcomes.
Collapse
Affiliation(s)
- Meghna Verma
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, 1015 Life Science Circle, Blacksburg, VA 24061, USA.,Graduate Program in Translational Biology, Medicine and Health, Virginia Tech, Blacksburg, 1 Riverside Circle, Roanoke, VA 24016, USA
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, 1015 Life Science Circle, Blacksburg, VA 24061, USA
| | - Andrew Leber
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, 1015 Life Science Circle, Blacksburg, VA 24061, USA
| | - Nuria Tubau-Juni
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, 1015 Life Science Circle, Blacksburg, VA 24061, USA
| | - Stefan Hoops
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, 1015 Life Science Circle, Blacksburg, VA 24061, USA
| | - Vida Abedi
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, 1015 Life Science Circle, Blacksburg, VA 24061, USA
| | - Xi Chen
- Grado Department of Industrial and Systems Engineering, Virginia Tech, 250 Perry St, Blacksburg, VA 24061, USA
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory, Biocomplexity Institute of Virginia Tech, 1015 Life Science Circle, Blacksburg, VA 24061, USA
| |
Collapse
|
58
|
Macfarlane FR, Chaplain MAJ, Eftimie R. Quantitative Predictive Modelling Approaches to Understanding Rheumatoid Arthritis: A Brief Review. Cells 2019; 9:E74. [PMID: 31892234 PMCID: PMC7016994 DOI: 10.3390/cells9010074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis is a chronic autoimmune disease that is a major public health challenge. The disease is characterised by inflammation of synovial joints and cartilage erosion, which lead to chronic pain, poor life quality and, in some cases, mortality. Understanding the biological mechanisms behind the progression of the disease, as well as developing new methods for quantitative predictions of disease progression in the presence/absence of various therapies is important for the success of therapeutic approaches. The aim of this study is to review various quantitative predictive modelling approaches for understanding rheumatoid arthritis. To this end, we start by briefly discussing the biology of this disease and some current treatment approaches, as well as emphasising some of the open problems in the field. Then, we review various mathematical mechanistic models derived to address some of these open problems. We discuss models that investigate the biological mechanisms behind the progression of the disease, as well as pharmacokinetic and pharmacodynamic models for various drug therapies. Furthermore, we highlight models aimed at optimising the costs of the treatments while taking into consideration the evolution of the disease and potential complications.
Collapse
Affiliation(s)
- Fiona R. Macfarlane
- School of Mathematics and Statistics, University of St Andrews, St Andrews KY16 9RJ, UK;
| | - Mark A. J. Chaplain
- School of Mathematics and Statistics, University of St Andrews, St Andrews KY16 9RJ, UK;
| | - Raluca Eftimie
- Department of Mathematics, University of Dundee, Dundee DD1 4HN, UK;
| |
Collapse
|
59
|
Rikard SM, Athey TL, Nelson AR, Christiansen SLM, Lee JJ, Holmes JW, Peirce SM, Saucerman JJ. Multiscale Coupling of an Agent-Based Model of Tissue Fibrosis and a Logic-Based Model of Intracellular Signaling. Front Physiol 2019; 10:1481. [PMID: 31920691 PMCID: PMC6928129 DOI: 10.3389/fphys.2019.01481] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 11/18/2019] [Indexed: 12/14/2022] Open
Abstract
Wound healing and fibrosis following myocardial infarction (MI) is a dynamic process involving many cell types, extracellular matrix (ECM), and inflammatory cues. As both incidence and survival rates for MI increase, management of post-MI recovery and associated complications are an increasingly important focus. Complexity of the wound healing process and the need for improved therapeutics necessitate a better understanding of the biochemical cues that drive fibrosis. To study the progression of cardiac fibrosis across spatial and temporal scales, we developed a novel hybrid multiscale model that couples a logic-based differential equation (LDE) model of the fibroblast intracellular signaling network with an agent-based model (ABM) of multi-cellular tissue remodeling. The ABM computes information about cytokine and growth factor levels in the environment including TGFβ, TNFα, IL-1β, and IL-6, which are passed as inputs to the LDE model. The LDE model then computes the network signaling state of individual cardiac fibroblasts within the ABM. Based on the current network state, fibroblasts make decisions regarding cytokine secretion and deposition and degradation of collagen. Simulated fibroblasts respond dynamically to rapidly changing extracellular environments and contribute to spatial heterogeneity in model predicted fibrosis, which is governed by many parameters including cell density, cell migration speeds, and cytokine levels. Verification tests confirmed that predictions of the coupled model and network model alone were consistent in response to constant cytokine inputs and furthermore, a subset of coupled model predictions were validated with in vitro experiments with human cardiac fibroblasts. This multiscale framework for cardiac fibrosis will allow for systematic screening of the effects of molecular perturbations in fibroblast signaling on tissue-scale extracellular matrix composition and organization.
Collapse
Affiliation(s)
- S Michaela Rikard
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Thomas L Athey
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States
| | - Anders R Nelson
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States
| | - Steven L M Christiansen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Jia-Jye Lee
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Jeffrey W Holmes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States.,Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Shayn M Peirce
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| | - Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States.,Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
60
|
Chowkwale M, Mahler GJ, Huang P, Murray BT. A multiscale in silico model of endothelial to mesenchymal transformation in a tumor microenvironment. J Theor Biol 2019; 480:229-240. [PMID: 31430445 DOI: 10.1016/j.jtbi.2019.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 08/01/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
Endothelial to mesenchymal transformation (EndMT) is a process in which endothelial cells gain a mesenchymal-like phenotype in response to mechanobiological signals that results in the remodeling or repair of underlying tissue. While initially associated with embryonic development, this process has since been shown to occur in adult tissue remodeling including wound healing, fibrosis, and cancer. In an attempt to understand the role of EndMT in cancer progression and metastasis, we present a multiscale, three-dimensional, in silico model. The model couples tissue level phenomena such as extracellular matrix remodeling, cellular level phenomena such as migration and proliferation, and chemical transport in the tumor microenvironment to mimic in vitro tissue models of the cancer microenvironment. The model is used to study the presence of EndMT-derived activated fibroblasts (EDAFs) and varying substrate stiffness on tumor cell migration and proliferation. The simulations accurately model the behavior of tumor cells under given conditions. The presence of EDAFs and/or an increase in substrate stiffness resulted in an increase in tumor cell activity. This model lays the foundation of further studies of EDAFs in a tumor microenvironment on a cellular and subcellular physiological level.
Collapse
Affiliation(s)
- M Chowkwale
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA
| | - G J Mahler
- Department of Biomedical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA
| | - P Huang
- Department of Mechanical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA
| | - B T Murray
- Department of Mechanical Engineering, Binghamton University, PO Box 6000, Binghamton, NY 13902, USA.
| |
Collapse
|
61
|
Vodovotz Y, An G. Agent-based models of inflammation in translational systems biology: A decade later. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2019; 11:e1460. [PMID: 31260168 PMCID: PMC8140858 DOI: 10.1002/wsbm.1460] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 06/14/2019] [Accepted: 06/15/2019] [Indexed: 12/11/2022]
Abstract
Agent-based modeling is a rule-based, discrete-event, and spatially explicit computational modeling method that employs computational objects that instantiate the rules and interactions among the individual components ("agents") of system. Agent-based modeling is well suited to translating into a computational model the knowledge generated from basic science research, particularly with respect to translating across scales the mechanisms of cellular behavior into aggregated cell population dynamics manifesting at the tissue and organ level. This capacity has made agent-based modeling an integral method in translational systems biology (TSB), an approach that uses multiscale dynamic computational modeling to explicitly represent disease processes in a clinically relevant fashion. The initial work in the early 2000s using agent-based models (ABMs) in TSB focused on examining acute inflammation and its intersection with wound healing; the decade since has seen vast growth in both the application of agent-based modeling to a wide array of disease processes as well as methodological advancements in the use and analysis of ABM. This report presents an update on an earlier review of ABMs in TSB and presents examples of exciting progress in the modeling of various organs and diseases that involve inflammation. This review also describes developments that integrate the use of ABMs with cutting-edge technologies such as high-performance computing, machine learning, and artificial intelligence, with a view toward the future integration of these methodologies. This article is categorized under: Translational, Genomic, and Systems Medicine > Translational Medicine Models of Systems Properties and Processes > Mechanistic Models Models of Systems Properties and Processes > Organ, Tissue, and Physiological Models Models of Systems Properties and Processes > Organismal Models.
Collapse
Affiliation(s)
- Yoram Vodovotz
- Department of Surgery, Immunology, Computational & Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Gary An
- Department of Surgery, University of Vermont, Burlington, Vermont
| |
Collapse
|
62
|
Harris LA, Beik S, Ozawa PMM, Jimenez L, Weaver AM. Modeling heterogeneous tumor growth dynamics and cell-cell interactions at single-cell and cell-population resolution. CURRENT OPINION IN SYSTEMS BIOLOGY 2019; 17:24-34. [PMID: 32642602 PMCID: PMC7343346 DOI: 10.1016/j.coisb.2019.09.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cancer is a complex, dynamic disease that despite recent advances remains mostly incurable. Inter- and intratumoral heterogeneity are generally considered major drivers of therapy resistance, metastasis, and treatment failure. Recent advances in high-throughput experimentation have produced a wealth of data on tumor heterogeneity and researchers are increasingly turning to mathematical modeling to aid in the interpretation of these complex datasets. In this mini-review, we discuss three important classes of approaches for modeling cellular dynamics within heterogeneous tumors: agent-based models, population dynamics, and multiscale models. An important new focus, for which we provide an example, is the role of intratumoral cell-cell interactions.
Collapse
Affiliation(s)
- Leonard A. Harris
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Samantha Beik
- Cancer Biology Graduate Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Patricia M. M. Ozawa
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Lizandra Jimenez
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Alissa M. Weaver
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
63
|
Garg A, Yuen S, Seekhao N, Yu G, Karwowski JAC, Powell M, Sakata JT, Mongeau L, JaJa J, Li-Jessen NYK. Towards a Physiological Scale of Vocal Fold Agent-Based Models of Surgical Injury and Repair: Sensitivity Analysis, Calibration and Verification. APPLIED SCIENCES (BASEL, SWITZERLAND) 2019; 9:2974. [PMID: 31372307 PMCID: PMC6675024 DOI: 10.3390/app9152974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Agent based models (ABM) were developed to numerically simulate the biological response to surgical vocal fold injury and repair at the physiological level. This study aimed to improve the representation of existing ABM through a combination of empirical and computational experiments. Empirical data of vocal fold cell populations including neutrophils, macrophages and fibroblasts were obtained using flow cytometry up to four weeks following surgical injury. Random Forests were used as a sensitivity analysis method to identify model parameters that were most influential to ABM outputs. Statistical Parameter Optimization Tool for Python was used to calibrate those parameter values to match the ABM-simulation data with the corresponding empirical data from Day 1 to Day 5 following surgery. Model performance was evaluated by verifying if the empirical data fell within the 95% confidence intervals of ABM outputs of cell quantities at Day 7, Week 2 and Week 4. For Day 7, all empirical data were within the ABM output ranges. The trends of ABM-simulated cell populations were also qualitatively comparable to those of the empirical data beyond Day 7. Exact values, however, fell outside of the 95% statistical confidence intervals. Parameters related to fibroblast proliferation were indicative to the ABM-simulation of fibroblast dynamics in final stages of wound healing.
Collapse
Affiliation(s)
- Aman Garg
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Samson Yuen
- School of Communication Sciences and Disorders, McGill University, Montreal, QC H3A 1G1, Canada
| | - Nuttiiya Seekhao
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD 20742, USA
| | - Grace Yu
- School of Communication Sciences and Disorders, McGill University, Montreal, QC H3A 1G1, Canada
| | | | - Michael Powell
- Virginia Tech Carilion Research Institute, Roanoke, VA 24016, USA
| | - Jon T. Sakata
- Department of Biology, McGill University, Montreal, QC H3A 1G1, Canada
| | - Luc Mongeau
- Department of Mechanical Engineering, McGill University, Montreal, QC H3A 0G4, Canada
| | - Joseph JaJa
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD 20742, USA
| | - Nicole Y. K. Li-Jessen
- Department of Biological and Biomedical Engineering, McGill University, Montreal, QC H3A 0G4, Canada
- School of Communication Sciences and Disorders, McGill University, Montreal, QC H3A 1G1, Canada
- Department of Otolaryngology–Head and Neck Surgery, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
64
|
Kumar M, Ji B, Zengler K, Nielsen J. Modelling approaches for studying the microbiome. Nat Microbiol 2019; 4:1253-1267. [PMID: 31337891 DOI: 10.1038/s41564-019-0491-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 05/21/2019] [Indexed: 02/08/2023]
Abstract
Advances in metagenome sequencing of the human microbiome have provided a plethora of new insights and revealed a close association of this complex ecosystem with a range of human diseases. However, there is little knowledge about how the different members of the microbial community interact with each other and with the host, and we lack basic mechanistic understanding of these interactions related to health and disease. Mathematical modelling has been demonstrated to be highly advantageous for gaining insights into the dynamics and interactions of complex systems and in recent years, several modelling approaches have been proposed to enhance our understanding of the microbiome. Here, we review the latest developments and current approaches, and highlight how different modelling strategies have been applied to unravel the highly dynamic nature of the human microbiome. Furthermore, we discuss present limitations of different modelling strategies and provide a perspective of how modelling can advance understanding and offer new treatment routes to impact human health.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden.,Department of Pediatrics, University of California, San Diego, CA, USA
| | - Boyang Ji
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Karsten Zengler
- Department of Pediatrics, University of California, San Diego, CA, USA.,Department of Bioengineering, University of California, San Diego, CA, USA.,Center for Microbiome Innovation, University of California, San Diego, CA, USA
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden. .,Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark.
| |
Collapse
|
65
|
Burrowes KS, Iravani A, Kang W. Integrated lung tissue mechanics one piece at a time: Computational modeling across the scales of biology. Clin Biomech (Bristol, Avon) 2019; 66:20-31. [PMID: 29352607 DOI: 10.1016/j.clinbiomech.2018.01.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/05/2017] [Accepted: 01/09/2018] [Indexed: 02/07/2023]
Abstract
The lung is a delicately balanced and highly integrated mechanical system. Lung tissue is continuously exposed to the environment via the air we breathe, making it susceptible to damage. As a consequence, respiratory diseases present a huge burden on society and their prevalence continues to rise. Emergent function is produced not only by the sum of the function of its individual components but also by the complex feedback and interactions occurring across the biological scales - from genes to proteins, cells, tissue and whole organ - and back again. Computational modeling provides the necessary framework for pulling apart and putting back together the pieces of the body and organ systems so that we can fully understand how they function in both health and disease. In this review, we discuss models of lung tissue mechanics spanning from the protein level (the extracellular matrix) through to the level of cells, tissue and whole organ, many of which have been developed in isolation. This is a vital step in the process but to understand the emergent behavior of the lung, we must work towards integrating these component parts and accounting for feedback across the scales, such as mechanotransduction. These interactions will be key to unlocking the mechanisms occurring in disease and in seeking new pharmacological targets and improving personalized healthcare.
Collapse
Affiliation(s)
- Kelly S Burrowes
- Department of Chemical and Materials Engineering, University of Auckland, 2-6 Park Avenue, Auckland 1023, New Zealand; Auckland Bioengineering Institute, University of Auckland, 70 Symonds Street, Auckland 1010, New Zealand.
| | - Amin Iravani
- Department of Chemical and Materials Engineering, University of Auckland, 2-6 Park Avenue, Auckland 1023, New Zealand.
| | - Wendy Kang
- Auckland Bioengineering Institute, University of Auckland, 70 Symonds Street, Auckland 1010, New Zealand.
| |
Collapse
|
66
|
Petersen BK, Yang J, Grathwohl WS, Cockrell C, Santiago C, An G, Faissol DM. Deep Reinforcement Learning and Simulation as a Path Toward Precision Medicine. J Comput Biol 2019; 26:597-604. [PMID: 30681362 DOI: 10.1089/cmb.2018.0168] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Traditionally, precision medicine involves classifying patients to identify subpopulations that respond favorably to specific therapeutics. We pose precision medicine as a dynamic feedback control problem, where treatment administered to a patient is guided by measurements taken during the course of treatment. We consider sepsis, a life-threatening condition in which dysregulation of the immune system causes tissue damage. We leverage an existing simulation of the innate immune response to infection and apply deep reinforcement learning (DRL) to discover an adaptive personalized treatment policy that specifies effective multicytokine therapy to simulated sepsis patients based on systemic measurements. The learned policy achieves a dramatic reduction in mortality rate over a set of 500 simulated patients relative to standalone antibiotic therapy. Advantages of our approach are threefold: (1) the use of simulation allows exploring therapeutic strategies beyond clinical practice and available data, (2) advances in DRL accommodate learning complex therapeutic strategies for complex biological systems, and (3) optimized treatments respond to a patient's individual disease progression over time, therefore, capturing both differences across patients and the inherent randomness of disease progression within a single patient. We hope that this work motivates both considering adaptive personalized multicytokine mediation therapy for sepsis and exploiting simulation with DRL for precision medicine more broadly.
Collapse
Affiliation(s)
- Brenden K Petersen
- 1 Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, California
| | - Jiachen Yang
- 1 Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, California
| | - Will S Grathwohl
- 1 Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, California
| | - Chase Cockrell
- 2 Department of Surgery, University of Vermont, Burlington, Vermont
| | - Claudio Santiago
- 1 Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, California
| | - Gary An
- 2 Department of Surgery, University of Vermont, Burlington, Vermont
| | - Daniel M Faissol
- 1 Computational Engineering Division, Lawrence Livermore National Laboratory, Livermore, California
| |
Collapse
|
67
|
Verma BK, Subramaniam P, Vadigepalli R. Model-based virtual patient analysis of human liver regeneration predicts critical perioperative factors controlling the dynamic mode of response to resection. BMC SYSTEMS BIOLOGY 2019; 13:9. [PMID: 30651095 PMCID: PMC6335689 DOI: 10.1186/s12918-019-0678-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 01/02/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND Liver has the unique ability to regenerate following injury, with a wide range of variability of the regenerative response across individuals. Existing computational models of the liver regeneration are largely tuned based on rodent data and hence it is not clear how well these models capture the dynamics of human liver regeneration. Recent availability of human liver volumetry time series data has enabled new opportunities to tune the computational models for human-relevant time scales, and to predict factors that can significantly alter the dynamics of liver regeneration following a resection. METHODS We utilized a mathematical model that integrates signaling mechanisms and cellular functional state transitions. We tuned the model parameters to match the time scale of human liver regeneration using an elastic net based regularization approach for identifying optimal parameter values. We initially examined the effect of each parameter individually on the response mode (normal, suppressed, failure) and extent of recovery to identify critical parameters. We employed phase plane analysis to compute the threshold of resection. We mapped the distribution of the response modes and threshold of resection in a virtual patient cohort generated in silico via simultaneous variations in two most critical parameters. RESULTS Analysis of the responses to resection with individual parameter variations showed that the response mode and extent of recovery following resection were most sensitive to variations in two perioperative factors, metabolic load and cell death post partial hepatectomy. Phase plane analysis identified two steady states corresponding to recovery and failure, with a threshold of resection separating the two basins of attraction. The size of the basin of attraction for the recovery mode varied as a function of metabolic load and cell death sensitivity, leading to a change in the multiplicity of the system in response to changes in these two parameters. CONCLUSIONS Our results suggest that the response mode and threshold of failure are critically dependent on the metabolic load and cell death sensitivity parameters that are likely to be patient-specific. Interventions that modulate these critical perioperative factors may be helpful to drive the liver regenerative response process towards a complete recovery mode.
Collapse
Affiliation(s)
- Babita K Verma
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Chemical Engineering, Indian Institute of Technology-Madras, Chennai, India
| | | | - Rajanikanth Vadigepalli
- Daniel Baugh Institute for Functional Genomics/Computational Biology, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
68
|
Norton KA, Gong C, Jamalian S, Popel AS. Multiscale Agent-Based and Hybrid Modeling of the Tumor Immune Microenvironment. Processes (Basel) 2019; 7:37. [PMID: 30701168 PMCID: PMC6349239 DOI: 10.3390/pr7010037] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Multiscale systems biology and systems pharmacology are powerful methodologies that are playing increasingly important roles in understanding the fundamental mechanisms of biological phenomena and in clinical applications. In this review, we summarize the state of the art in the applications of agent-based models (ABM) and hybrid modeling to the tumor immune microenvironment and cancer immune response, including immunotherapy. Heterogeneity is a hallmark of cancer; tumor heterogeneity at the molecular, cellular, and tissue scales is a major determinant of metastasis, drug resistance, and low response rate to molecular targeted therapies and immunotherapies. Agent-based modeling is an effective methodology to obtain and understand quantitative characteristics of these processes and to propose clinical solutions aimed at overcoming the current obstacles in cancer treatment. We review models focusing on intra-tumor heterogeneity, particularly on interactions between cancer cells and stromal cells, including immune cells, the role of tumor-associated vasculature in the immune response, immune-related tumor mechanobiology, and cancer immunotherapy. We discuss the role of digital pathology in parameterizing and validating spatial computational models and potential applications to therapeutics.
Collapse
Affiliation(s)
- Kerri-Ann Norton
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Computer Science Program, Department of Science, Mathematics, and Computing, Bard College, Annandale-on-Hudson, NY 12504, USA
| | - Chang Gong
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Samira Jamalian
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
69
|
Multicellular Models Bridging Intracellular Signaling and Gene Transcription to Population Dynamics. Processes (Basel) 2018. [DOI: 10.3390/pr6110217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cell signaling and gene transcription occur at faster time scales compared to cellular death, division, and evolution. Bridging these multiscale events in a model is computationally challenging. We introduce a framework for the systematic development of multiscale cell population models. Using message passing interface (MPI) parallelism, the framework creates a population model from a single-cell biochemical network model. It launches parallel simulations on a single-cell model and treats each stand-alone parallel process as a cell object. MPI mediates cell-to-cell and cell-to-environment communications in a server-client fashion. In the framework, model-specific higher level rules link the intracellular molecular events to cellular functions, such as death, division, or phenotype change. Cell death is implemented by terminating a parallel process, while cell division is carried out by creating a new process (daughter cell) from an existing one (mother cell). We first demonstrate these capabilities by creating two simple example models. In one model, we consider a relatively simple scenario where cells can evolve independently. In the other model, we consider interdependency among the cells, where cellular communication determines their collective behavior and evolution under a temporally evolving growth condition. We then demonstrate the framework’s capability by simulating a full-scale model of bacterial quorum sensing, where the dynamics of a population of bacterial cells is dictated by the intercellular communications in a time-evolving growth environment.
Collapse
|
70
|
Abstract
Abstract
Crowdsourcing is a very effective technique for outsourcing work to a vast network usually comprising anonymous people. In this study, we review the application of crowdsourcing to modeling systems originating from systems biology. We consider a variety of verified approaches, including well-known projects such as EyeWire, FoldIt, and DREAM Challenges, as well as novel projects conducted at the European Center for Bioinformatics and Genomics. The latter projects utilized crowdsourced serious games to design models of dynamic biological systems, and it was demonstrated that these models could be used successfully to involve players without domain knowledge. We conclude the review of these systems by providing 10 guidelines to facilitate the efficient use of crowdsourcing.
Collapse
|
71
|
Fitzpatrick BG. Issues in Reproducible Simulation Research. Bull Math Biol 2018; 81:1-6. [PMID: 30191471 DOI: 10.1007/s11538-018-0496-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/29/2018] [Indexed: 11/25/2022]
Abstract
In recent years, serious concerns have arisen about reproducibility in science. Estimates of the cost of irreproducible preclinical studies range from 28 billion USD per year in the USA alone (Freedman et al. in PLoS Biol 13(6):e1002165, 2015) to over 200 billion USD per year worldwide (Chalmers and Glasziou in Lancet 374:86-89, 2009). The situation in the social sciences is not very different: Reproducibility in psychological research, for example, has been estimated to be below 50% as well (Open Science Collaboration in Science 349:6251, 2015). Less well studied is the issue of reproducibility of simulation research. A few replication studies of agent-based models, however, suggest the problem for computational modeling may be more severe than for laboratory experiments (Willensky and Rand in JASSS 10(4):2, 2007; Donkin et al. in Environ Model Softw 92:142-151, 2017; Bajracharya and Duboz in: Proceedings of the symposium on theory of modeling and simulation-DEVS integrative M&S symposium, pp 6-11, 2013). In this perspective, we discuss problems of reproducibility in agent-based simulations of life and social science problems, drawing on best practices research in computer science and in wet-lab experiment design and execution to suggest some ways to improve simulation research practice.
Collapse
Affiliation(s)
- B G Fitzpatrick
- Tempest Technologies and Department of Mathematics, Loyola Marymount University, Los Angeles, CA, USA.
| |
Collapse
|
72
|
Deng J, Jhandey A, Zhu X, Yang Z, Yik KFP, Zuo Z, Lam TN. In silico drug absorption tract: An agent-based biomimetic model for human oral drug absorption. PLoS One 2018; 13:e0203361. [PMID: 30169515 PMCID: PMC6118387 DOI: 10.1371/journal.pone.0203361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 08/20/2018] [Indexed: 11/26/2022] Open
Abstract
Background An agent-based modeling approach has been suggested as an alternative to traditional, equation-based modeling methods for describing oral drug absorption. It enables researchers to gain a better understanding of the pharmacokinetic (PK) mechanisms of a drug. This project demonstrates that a biomimetic agent-based model can adequately describe the absorption and disposition kinetics both of midazolam and clonazepam. Methods An agent-based biomimetic model, in silico drug absorption tract (ISDAT), was built to mimic oral drug absorption in humans. The model consisted of distinct spaces, membranes, and metabolic enzymes, and it was altogether representative of human physiology relating to oral drug absorption. Simulated experiments were run with the model, and the results were compared to the referent data from clinical equivalence trials. Acceptable similarity was verified by pre-specified criteria, which included 1) qualitative visual matching between the clinical and simulated concentration-time profiles, 2) quantitative similarity indices, namely, weighted root mean squared error (RMSE), and weighted mean absolute percentage error (MAPE) and 3) descriptive similarity which requires less than 25% difference between key PK parameters calculated by the clinical and the simulated concentration-time profiles. The model and its parameters were iteratively refined until all similarity criteria were met. Furthermore, simulated PK experiments were conducted to predict bioavailability (F). For better visualization, a graphical user interface for the model was developed and a video is available in Supporting Information. Results Simulation results satisfied all three levels of similarity criteria for both drugs. The weighted RMSE was 0.51 and 0.92, and the weighted MAPE was 5.99% and 8.43% for midazolam and clonazepam, respectively. Calculated PK parameter values, including area under the curve (AUC), peak plasma drug concentration (Cmax), time to reach Cmax (Tmax), terminal elimination rate constant (Kel), terminal elimination half life (T1/2), apparent oral clearance (CL/F), and apparent volume of distribution (V/F), were reasonable compared to the referent values. The predicted absolute oral bioavailability (F) was 44% for midazolam (literature reported value, 31–72%) and 93% (literature reported value, ≥ 90%) for clonazepam. Conclusion The ISDAT met all the pre-specified similarity criteria for both midazolam and clonazepam, and demonstrated its ability to describe absorption kinetics of both drugs. Therefore, the validated ISDAT can be a promising platform for further research into the use of similar in silico models for drug absorption kinetics.
Collapse
Affiliation(s)
- Jianyuan Deng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Anika Jhandey
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Xiao Zhu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Zhibo Yang
- Department of Computer Science, Stony Brook University, Stony Brook, NY, United States of America
| | - Kin Fu Patrick Yik
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tai Ning Lam
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
- * E-mail:
| |
Collapse
|
73
|
Ferreira GBS, Scheutz M, Boyd SK. Mate choice strategies in a spatially-explicit model environment. PLoS One 2018; 13:e0202680. [PMID: 30138426 PMCID: PMC6107201 DOI: 10.1371/journal.pone.0202680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 08/07/2018] [Indexed: 11/18/2022] Open
Abstract
Decisions about the choice of a mate can greatly impact both individual fitness and selection processes. We developed a novel agent-based model to investigate two common mate choice rules that may be used by female gray treefrogs (Hyla versicolor). In this model environment, female agents using the minimum-threshold strategy found higher quality mates and traveled shorter distances on average, compared with female agents using the best-of-n strategy. Females using the minimum-threshold strategy, however, incur significant lost opportunity costs, depending on the male population quality average. The best-of-n strategy leads to significant female:female competition that limits their ability to find high quality mates. Thus, when the sex ratio is 0.8, best-of-5 and best-of-2 strategies yield mates of nearly identical quality. Although the distance traveled by females in the mating task varied depending on male spatial distribution in the environment, this did not interact with female choice for the best-of-n or minimum-threshold strategies. By incorporating empirical data from the frogs in this temporally- and spatially-explicit model, we thus show the emergence of novel interactions of common decision-making rules with realistic environmental variables.
Collapse
Affiliation(s)
| | - Matthias Scheutz
- Department of Computer Science, Tufts University, Medford, MA, United States of America
| | - Sunny K. Boyd
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, United States of America
| |
Collapse
|
74
|
Abstract
Liver resection is an important clinical intervention to treat liver disease. Following liver resection, patients exhibit a wide range of outcomes including normal recovery, suppressed recovery, or liver failure, depending on the regenerative capacity of the remnant liver. The objective of this work is to study the distinct patient outcomes post hepatectomy and determine the processes that are accountable for liver failure. Our model based approach shows that cell death is one of the important processes but not the sole controlling process responsible for liver failure. Additionally, our simulations showed wide variation in the timescale of liver failure that is consistent with the clinically observed timescales of post hepatectomy liver failure scenarios. Liver failure can take place either instantaneously or after a certain delay. We analyzed a virtual patient cohort and concluded that remnant liver fraction is a key regulator of the timescale of liver failure, with higher remnant liver fraction leading to longer time delay prior to failure. Our results suggest that, for a given remnant liver fraction, modulating a combination of cell death controlling parameters and metabolic load may help shift the clinical outcome away from post hepatectomy liver failure towards normal recovery.
Collapse
|
75
|
Abstract
PURPOSE OF REVIEW There is confusion in all of healthcare, including oculofacial surgery, as to what is 'complex' and what is 'merely complicated'. Although in common usage, these terms tend to be interchangeable, the distinction is more than trivial. A different and somewhat unfamiliar toolset is needed to successfully navigate complex problems. This review will explore a methodology for the physician to understand what is complex in oculofacial surgery, the tools needed to optimize performance in a complex healthcare system and successfully manage patients with complex diseases. RECENT FINDINGS A specific understanding of complexity science in oculofacial surgery is only in its nascent beginnings at this point. Nevertheless, recent advances in closely related fields can provide concrete applications. The practice of oculofacial surgery is optimized within a healthcare network of supporting professionals. Moreover, a newer understanding of the 'complex' nature of disease common to oculofacial surgery, such as neoplasia and inflammation, will direct the physician to recognize the most appropriate therapies. SUMMARY Oculofacial surgery, like all of medicine, is a fluid mixture of problems that are complex and those that are merely complicated. As a different toolset is needed to deal with each, physicians need to recognize these differences and acquire those tools.
Collapse
|
76
|
Soheilypour M, Mofrad MRK. Agent-Based Modeling in Molecular Systems Biology. Bioessays 2018; 40:e1800020. [PMID: 29882969 DOI: 10.1002/bies.201800020] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 04/11/2018] [Indexed: 12/13/2022]
Abstract
Molecular systems orchestrating the biology of the cell typically involve a complex web of interactions among various components and span a vast range of spatial and temporal scales. Computational methods have advanced our understanding of the behavior of molecular systems by enabling us to test assumptions and hypotheses, explore the effect of different parameters on the outcome, and eventually guide experiments. While several different mathematical and computational methods are developed to study molecular systems at different spatiotemporal scales, there is still a need for methods that bridge the gap between spatially-detailed and computationally-efficient approaches. In this review, we summarize the capabilities of agent-based modeling (ABM) as an emerging molecular systems biology technique that provides researchers with a new tool in exploring the dynamics of molecular systems/pathways in health and disease.
Collapse
Affiliation(s)
- Mohammad Soheilypour
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
77
|
Zengler K, Zaramela LS. The social network of microorganisms - how auxotrophies shape complex communities. Nat Rev Microbiol 2018; 16:383-390. [PMID: 29599459 PMCID: PMC6059367 DOI: 10.1038/s41579-018-0004-5] [Citation(s) in RCA: 226] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Microorganisms engage in complex interactions with other organisms and their environment. Recent studies have shown that these interactions are not limited to the exchange of electron donors. Most microorganisms are auxotrophs, thus relying on external nutrients for growth, including the exchange of amino acids and vitamins. Currently, we lack a deeper understanding of auxotrophies in microorganisms and how nutrient requirements differ between different strains and different environments. In this Opinion article, we describe how the study of auxotrophies and nutrient requirements among members of complex communities will enable new insights into community composition and assembly. Understanding this complex network over space and time is crucial for developing strategies to interrogate and shape microbial communities.
Collapse
Affiliation(s)
- Karsten Zengler
- Department of Pediatrics, Division of Host-Microbe Systems & Therapeutics, University of California, San Diego, CA, USA.
- Center for Microbiome Innovation, University of California, San Diego, CA, USA.
| | - Livia S Zaramela
- Department of Pediatrics, Division of Host-Microbe Systems & Therapeutics, University of California, San Diego, CA, USA
| |
Collapse
|
78
|
Hantusch A, Rehm M, Brunner T. Counting on Death – Quantitative aspects of Bcl‐2 family regulation. FEBS J 2018; 285:4124-4138. [DOI: 10.1111/febs.14516] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/27/2018] [Accepted: 05/21/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Annika Hantusch
- Department of Biology Chair of Biochemical Pharmacology University of Konstanz Germany
- Konstanz Research School Chemical Biology University of Konstanz Germany
| | - Markus Rehm
- Department of Physiology & Medical Physics Royal College of Surgeons in Ireland Dublin 2 Ireland
- Centre for Systems Medicine Royal College of Surgeons in Ireland Dublin 2 Ireland
- Institute of Cell Biology and Immunology University of Stuttgart Germany
- Stuttgart Research Center Systems Biology University of Stuttgart Germany
| | - Thomas Brunner
- Department of Biology Chair of Biochemical Pharmacology University of Konstanz Germany
- Konstanz Research School Chemical Biology University of Konstanz Germany
| |
Collapse
|
79
|
Seekhao N, Shung C, JaJa J, Mongeau L, Li-Jessen NYK. High-Performance Agent-Based Modeling Applied to Vocal Fold Inflammation and Repair. Front Physiol 2018; 9:304. [PMID: 29706894 PMCID: PMC5906585 DOI: 10.3389/fphys.2018.00304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/13/2018] [Indexed: 01/13/2023] Open
Abstract
Fast and accurate computational biology models offer the prospect of accelerating the development of personalized medicine. A tool capable of estimating treatment success can help prevent unnecessary and costly treatments and potential harmful side effects. A novel high-performance Agent-Based Model (ABM) was adopted to simulate and visualize multi-scale complex biological processes arising in vocal fold inflammation and repair. The computational scheme was designed to organize the 3D ABM sub-tasks to fully utilize the resources available on current heterogeneous platforms consisting of multi-core CPUs and many-core GPUs. Subtasks are further parallelized and convolution-based diffusion is used to enhance the performance of the ABM simulation. The scheme was implemented using a client-server protocol allowing the results of each iteration to be analyzed and visualized on the server (i.e., in-situ) while the simulation is running on the same server. The resulting simulation and visualization software enables users to interact with and steer the course of the simulation in real-time as needed. This high-resolution 3D ABM framework was used for a case study of surgical vocal fold injury and repair. The new framework is capable of completing the simulation, visualization and remote result delivery in under 7 s per iteration, where each iteration of the simulation represents 30 min in the real world. The case study model was simulated at the physiological scale of a human vocal fold. This simulation tracks 17 million biological cells as well as a total of 1.7 billion signaling chemical and structural protein data points. The visualization component processes and renders all simulated biological cells and 154 million signaling chemical data points. The proposed high-performance 3D ABM was verified through comparisons with empirical vocal fold data. Representative trends of biomarker predictions in surgically injured vocal folds were observed.
Collapse
Affiliation(s)
- Nuttiiya Seekhao
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, United States
| | - Caroline Shung
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Joseph JaJa
- Department of Electrical and Computer Engineering, University of Maryland, College Park, MD, United States
| | - Luc Mongeau
- Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Nicole Y K Li-Jessen
- School of Communication Sciences and Disorders, McGill University, Montreal, QC, Canada
| |
Collapse
|
80
|
Soleimani S, Shamsi M, Ghazani MA, Modarres HP, Valente KP, Saghafian M, Ashani MM, Akbari M, Sanati-Nezhad A. Translational models of tumor angiogenesis: A nexus of in silico and in vitro models. Biotechnol Adv 2018; 36:880-893. [PMID: 29378235 DOI: 10.1016/j.biotechadv.2018.01.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/10/2018] [Accepted: 01/20/2018] [Indexed: 12/13/2022]
Abstract
Emerging evidence shows that endothelial cells are not only the building blocks of vascular networks that enable oxygen and nutrient delivery throughout a tissue but also serve as a rich resource of angiocrine factors. Endothelial cells play key roles in determining cancer progression and response to anti-cancer drugs. Furthermore, the endothelium-specific deposition of extracellular matrix is a key modulator of the availability of angiocrine factors to both stromal and cancer cells. Considering tumor vascular network as a decisive factor in cancer pathogenesis and treatment response, these networks need to be an inseparable component of cancer models. Both computational and in vitro experimental models have been extensively developed to model tumor-endothelium interactions. While informative, they have been developed in different communities and do not yet represent a comprehensive platform. In this review, we overview the necessity of incorporating vascular networks for both in vitro and in silico cancer models and discuss recent progresses and challenges of in vitro experimental microfluidic cancer vasculature-on-chip systems and their in silico counterparts. We further highlight how these two approaches can merge together with the aim of presenting a predictive combinatorial platform for studying cancer pathogenesis and testing the efficacy of single or multi-drug therapeutics for cancer treatment.
Collapse
Affiliation(s)
- Shirin Soleimani
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Milad Shamsi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, AB T2N 1N4, Canada; Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 8415683111, Iran
| | - Mehran Akbarpour Ghazani
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 8415683111, Iran
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Karolina Papera Valente
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Mohsen Saghafian
- Department of Mechanical Engineering, Isfahan University of Technology, Isfahan 8415683111, Iran
| | - Mehdi Mohammadi Ashani
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, Calgary, AB T2N 1N4, Canada; Center for BioEngineering Research and Education, University of Calgary, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
81
|
Abstract
New technologies to generate, store and retrieve medical and research data are inducing a rapid change in clinical and translational research and health care. Systems medicine is the interdisciplinary approach wherein physicians and clinical investigators team up with experts from biology, biostatistics, informatics, mathematics and computational modeling to develop methods to use new and stored data to the benefit of the patient. We here provide a critical assessment of the opportunities and challenges arising out of systems approaches in medicine and from this provide a definition of what systems medicine entails. Based on our analysis of current developments in medicine and healthcare and associated research needs, we emphasize the role of systems medicine as a multilevel and multidisciplinary methodological framework for informed data acquisition and interdisciplinary data analysis to extract previously inaccessible knowledge for the benefit of patients.
Collapse
|
82
|
Lux SA. Individual-Based Modeling Approach to Assessment of the Impacts of Landscape Complexity and Climate on Dispersion, Detectability and Fate of Incipient Medfly Populations. Front Physiol 2018; 8:1121. [PMID: 29375396 PMCID: PMC5767299 DOI: 10.3389/fphys.2017.01121] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 12/18/2017] [Indexed: 11/13/2022] Open
Affiliation(s)
- Slawomir A. Lux
- inSilico-IPM, Konstancin-Jeziorna, Poland
- Formely: Department of Applied Entomology, Warsaw University of Life Sciences, Warsaw, Poland
- *Correspondence: Slawomir A. Lux ;
| |
Collapse
|
83
|
Rao RT, Scherholz ML, Hartmanshenn C, Bae SA, Androulakis IP. On the analysis of complex biological supply chains: From Process Systems Engineering to Quantitative Systems Pharmacology. Comput Chem Eng 2017; 107:100-110. [PMID: 29353945 DOI: 10.1016/j.compchemeng.2017.06.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The use of models in biology has become particularly relevant as it enables investigators to develop a mechanistic framework for understanding the operating principles of living systems as well as in quantitatively predicting their response to both pathological perturbations and pharmacological interventions. This application has resulted in a synergistic convergence of systems biology and pharmacokinetic-pharmacodynamic modeling techniques that has led to the emergence of quantitative systems pharmacology (QSP). In this review, we discuss how the foundational principles of chemical process systems engineering inform the progressive development of more physiologically-based systems biology models.
Collapse
Affiliation(s)
- Rohit T Rao
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Megerle L Scherholz
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Clara Hartmanshenn
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Seul-A Bae
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854
| | - Ioannis P Androulakis
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854.,Department of Biomedical Engineering, Rutgers The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854
| |
Collapse
|
84
|
Read MN, Alden K, Rose LM, Timmis J. Automated multi-objective calibration of biological agent-based simulations. J R Soc Interface 2017; 13:rsif.2016.0543. [PMID: 27628175 DOI: 10.1098/rsif.2016.0543] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 08/22/2016] [Indexed: 12/27/2022] Open
Abstract
Computational agent-based simulation (ABS) is increasingly used to complement laboratory techniques in advancing our understanding of biological systems. Calibration, the identification of parameter values that align simulation with biological behaviours, becomes challenging as increasingly complex biological domains are simulated. Complex domains cannot be characterized by single metrics alone, rendering simulation calibration a fundamentally multi-metric optimization problem that typical calibration techniques cannot handle. Yet calibration is an essential activity in simulation-based science; the baseline calibration forms a control for subsequent experimentation and hence is fundamental in the interpretation of results. Here, we develop and showcase a method, built around multi-objective optimization, for calibrating ABSs against complex target behaviours requiring several metrics (termed objectives) to characterize. Multi-objective calibration (MOC) delivers those sets of parameter values representing optimal trade-offs in simulation performance against each metric, in the form of a Pareto front. We use MOC to calibrate a well-understood immunological simulation against both established a priori and previously unestablished target behaviours. Furthermore, we show that simulation-borne conclusions are broadly, but not entirely, robust to adopting baseline parameter values from different extremes of the Pareto front, highlighting the importance of MOC's identification of numerous calibration solutions. We devise a method for detecting overfitting in a multi-objective context, not previously possible, used to save computational effort by terminating MOC when no improved solutions will be found. MOC can significantly impact biological simulation, adding rigour to and speeding up an otherwise time-consuming calibration process and highlighting inappropriate biological capture by simulations that cannot be well calibrated. As such, it produces more accurate simulations that generate more informative biological predictions.
Collapse
Affiliation(s)
- Mark N Read
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, New South Wales, Australia Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
| | - Kieran Alden
- Department of Electronics, University of York, York, UK
| | - Louis M Rose
- Department of Computer Science, University of York, York, UK
| | - Jon Timmis
- Department of Electronics, University of York, York, UK
| |
Collapse
|
85
|
Abstract
Introduction Tuberculosis (TB), one of the most common infectious diseases, requires treatment with multiple antibiotics taken over at least 6 months. This long treatment often results in poor patient-adherence, which can lead to the emergence of multi-drug resistant TB. New antibiotic treatment strategies are sorely needed. New antibiotics are being developed or repurposed to treat TB, but as there are numerous potential antibiotics, dosing sizes and potential schedules, the regimen design space for new treatments is too large to search exhaustively. Here we propose a method that combines an agent-based multi-scale model capturing TB granuloma formation with algorithms for mathematical optimization to identify optimal TB treatment regimens. Methods We define two different single-antibiotic treatments to compare the efficiency and accuracy in predicting optimal treatment regimens of two optimization algorithms: genetic algorithms (GA) and surrogate-assisted optimization through radial basis function (RBF) networks. We also illustrate the use of RBF networks to optimize double-antibiotic treatments. Results We found that while GAs can locate optimal treatment regimens more accurately, RBF networks provide a more practical strategy to TB treatment optimization with fewer simulations, and successfully estimated optimal double-antibiotic treatment regimens. Conclusions Our results indicate surrogate-assisted optimization can locate optimal TB treatment regimens from a larger set of antibiotics, doses and schedules, and could be applied to solve optimization problems in other areas of research using systems biology approaches. Our findings have important implications for the treatment of diseases like TB that have lengthy protocols or for any disease that requires multiple drugs.
Collapse
|
86
|
Das AA, Ajayakumar Darsana T, Jacob E. Agent-based re-engineering of ErbB signaling: a modeling pipeline for integrative systems biology. Bioinformatics 2017; 33:726-732. [PMID: 27998938 DOI: 10.1093/bioinformatics/btw709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/08/2016] [Indexed: 11/14/2022] Open
Abstract
Motivation Experiments in systems biology are generally supported by a computational model which quantitatively estimates the parameters of the system by finding the best fit to the experiment. Mathematical models have proved to be successful in reverse engineering the system. The data generated is interpreted to understand the dynamics of the underlying phenomena. The question we have sought to answer is that - is it possible to use an agent-based approach to re-engineer a biological process, making use of the available knowledge from experimental and modelling efforts? Can the bottom-up approach benefit from the top-down exercise so as to create an integrated modelling formalism for systems biology? We propose a modelling pipeline that learns from the data given by reverse engineering, and uses it for re-engineering the system, to carry out in-silico experiments. Results A mathematical model that quantitatively predicts co-expression of EGFR-HER2 receptors in activation and trafficking has been taken for this study. The pipeline architecture takes cues from the population model that gives the rates of biochemical reactions, to formulate knowledge-based rules for the particle model. Agent-based simulations using these rules, support the existing facts on EGFR-HER2 dynamics. We conclude that, re-engineering models, built using the results of reverse engineering, opens up the possibility of harnessing the power pack of data which now lies scattered in literature. Virtual experiments could then become more realistic when empowered with the findings of empirical cell biology and modelling studies. Availability and Implementation Implemented on the Agent Modelling Framework developed in-house. C ++ code templates available in Supplementary material . Contact liz.csir@gmail.com. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
|
87
|
Simple and complex retinal dystrophies are associated with profoundly different disease networks. Sci Rep 2017; 7:41835. [PMID: 28139756 PMCID: PMC5282568 DOI: 10.1038/srep41835] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 12/28/2016] [Indexed: 12/20/2022] Open
Abstract
Retinopathies are a group of monogenetic or complex retinal diseases associated with high unmet medical need. Monogenic disorders are caused by rare genetic variation and usually arise early in life. Other diseases, such as age-related macular degeneration (AMD), develop late in life and are considered to be of complex origin as they develop from a combination of genetic, ageing, environmental and lifestyle risk factors. Here, we contrast the underlying disease networks and pathological mechanisms of monogenic as opposed to complex retinopathies, using AMD as an example of the latter. We show that, surprisingly, genes associated with the different forms of retinopathies in general do not overlap despite their overlapping retinal phenotypes. Further, AMD risk genes participate in multiple networks with interaction partners that link to different ubiquitous pathways affecting general tissue integrity and homeostasis. Thus AMD most likely represents an endophenotype with differing underlying pathogenesis in different subjects. Localising these pathomechanisms and processes within and across different retinal anatomical compartments provides a novel representation of AMD that may be extended to complex disease in general. This approach may generate improved treatment options that target multiple processes with the aim of restoring tissue homeostasis and maintaining vision.
Collapse
|
88
|
Wang Q, Holmes WR, Sosnik J, Schilling T, Nie Q. Cell Sorting and Noise-Induced Cell Plasticity Coordinate to Sharpen Boundaries between Gene Expression Domains. PLoS Comput Biol 2017; 13:e1005307. [PMID: 28135279 PMCID: PMC5279720 DOI: 10.1371/journal.pcbi.1005307] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 12/09/2016] [Indexed: 12/13/2022] Open
Abstract
A fundamental question in biology is how sharp boundaries of gene expression form precisely in spite of biological variation/noise. Numerous mechanisms position gene expression domains across fields of cells (e.g. morphogens), but how these domains are refined remains unclear. In some cases, domain boundaries sharpen through differential adhesion-mediated cell sorting. However, boundaries can also sharpen through cellular plasticity, with cell fate changes driven by up- or down-regulation of gene expression. In this context, we have argued that noise in gene expression can help cells transition to the correct fate. Here we investigate the efficacy of cell sorting, gene expression plasticity, and their combination in boundary sharpening using multi-scale, stochastic models. We focus on the formation of hindbrain segments (rhombomeres) in the developing zebrafish as an example, but the mechanisms investigated apply broadly to many tissues. Our results indicate that neither sorting nor plasticity is sufficient on its own to sharpen transition regions between different rhombomeres. Rather the two have complementary strengths and weaknesses, which synergize when combined to sharpen gene expression boundaries. In many developing systems, chemical gradients control the formation of segmental domains of gene expression, specifying distinct domains that go on to form different tissues and structures, in a concentration-dependent manner. These gradients are noisy however, raising the question of how sharply delineated boundaries between distinct segments form. It is crucial that developing systems be able to cope with stochasticity and generate well-defined boundaries between different segmented domains. Previous work suggests that cell sorting and cellular plasticity help sharpen boundaries between segments. However, it remains unclear how effective each of these mechanisms is and what their role in sharpening may be. Motivated by recent experimental observations, we construct a hybrid stochastic model to investigate these questions. We find that neither mechanism is sufficient on its own to sharpen boundaries between different segments. Rather, results indicate each has its own strengths and weaknesses, and that they work together synergistically to promote the development of precise, well defined segment boundaries. Formation of segmented rhombomeres in the zebrafish hindbrain, which later form different components of the central nervous system, is a motivating case for this study.
Collapse
Affiliation(s)
- Qixuan Wang
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA, United States of America
- Department of Mathematics, University of California Irvine, Irvine, CA, United States of America
| | - William R. Holmes
- Department of Physics and Astronomy, Vanderbilt University, Nashville, TN, United States of America
| | - Julian Sosnik
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA, United States of America
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, United States of America
| | - Thomas Schilling
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA, United States of America
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, United States of America
| | - Qing Nie
- Center for Complex Biological Systems, University of California Irvine, Irvine, CA, United States of America
- Department of Mathematics, University of California Irvine, Irvine, CA, United States of America
- * E-mail:
| |
Collapse
|
89
|
An Adaptive Agent-Based Model of Homing Pigeons: A Genetic Algorithm Approach. ISPRS INTERNATIONAL JOURNAL OF GEO-INFORMATION 2017. [DOI: 10.3390/ijgi6010027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
90
|
Varga M, Prokop A, Csukas B. Biosystem models, generated from a complex rule/reaction/influence network and from two functionality prototypes. Biosystems 2017; 152:24-43. [PMID: 28062323 DOI: 10.1016/j.biosystems.2016.12.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 12/23/2016] [Indexed: 12/24/2022]
Abstract
In this work we have further developed the Direct Computer Mapping (DCM) based modelling and simulation methodology. A unified, transition-based representation of complex rule, reaction and influence networks has been introduced and two prototypes (one general state- and another general transition-prototype) have been developed for the unified functional modelling of the state and transition nodes. Starting from the network and from the functional prototypes, an automatic generation method of the graphically editable and extensible GraphML description of biosystem models has been elaborated. The new developments have been implemented in the improved kernel of DCM models. The applied knowledge representation makes possible the unified generation and execution of the balance-based quantitative and influence- or rule-based qualitative, as well as optionally time-driven, multiscale biosystem models. Application of the developed methodology has been illustrated by the improved implementation of the formerly studied and upgraded example biosystem model for combining the detailed, quantitative p53/miR34a signalling system with the pathological model through an extended rule-based coupling model.
Collapse
Affiliation(s)
- M Varga
- Research Group on Process Network Engineering, Kaposvar University, 40 Guba S, 7400, Kaposvar, Hungary.
| | - A Prokop
- Department of Chemical & Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - B Csukas
- Research Group on Process Network Engineering, Kaposvar University, 40 Guba S, 7400, Kaposvar, Hungary
| |
Collapse
|
91
|
Abstract
This chapter aims at discussing the content of multi-agent based simulation (MABS) applied to computational biology i.e., to modelling and simulating biological systems by means of computational models, methodologies, and frameworks. In particular, the adoption of agent-based modelling (ABM) in the field of multicellular systems biology is explored, focussing on the challenging scenarios of developmental biology. After motivating why agent-based abstractions are critical in representing multicellular systems behaviour, MABS is discussed as the source of the most natural and appropriate mechanism for analysing the self-organising behaviour of systems of cells. As a case study, an application of MABS to the development of Drosophila Melanogaster is finally presented, which exploits the ALCHEMIST platform for agent-based simulation.
Collapse
|
92
|
Wedgwood KCA, Richardson SJ, Morgan NG, Tsaneva-Atanasova K. Spatiotemporal Dynamics of Insulitis in Human Type 1 Diabetes. Front Physiol 2016; 7:633. [PMID: 28082906 PMCID: PMC5186767 DOI: 10.3389/fphys.2016.00633] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 12/05/2016] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes (T1D) is an auto-immune disease characterized by the selective destruction of the insulin secreting beta cells in the pancreas during an inflammatory phase known as insulitis. Patients with T1D are typically dependent on the administration of externally provided insulin in order to manage blood glucose levels. Whilst technological developments have significantly improved both the life expectancy and quality of life of these patients, an understanding of the mechanisms of the disease remains elusive. Animal models, such as the NOD mouse model, have been widely used to probe the process of insulitis, but there exist very few data from humans studied at disease onset. In this manuscript, we employ data from human pancreases collected close to the onset of T1D and propose a spatio-temporal computational model for the progression of insulitis in human T1D, with particular focus on the mechanisms underlying the development of insulitis in pancreatic islets. This framework allows us to investigate how the time-course of insulitis progression is affected by altering key parameters, such as the number of the CD20+ B cells present in the inflammatory infiltrate, which has recently been proposed to influence the aggressiveness of the disease. Through the analysis of repeated simulations of our stochastic model, which track the number of beta cells within an islet, we find that increased numbers of B cells in the peri-islet space lead to faster destruction of the beta cells. We also find that the balance between the degradation and repair of the basement membrane surrounding the islet is a critical component in governing the overall destruction rate of the beta cells and their remaining number. Our model provides a framework for continued and improved spatio-temporal modeling of human T1D.
Collapse
Affiliation(s)
- Kyle C. A. Wedgwood
- Centre for Biomedical Modelling and Analysis, University of ExeterExeter, UK
| | | | - Noel G. Morgan
- University of Exeter Medical School, University of ExeterExeter, UK
| | - Krasimira Tsaneva-Atanasova
- College for Engineering, Mathematics and Physical Sciences, University of ExeterExeter, UK
- Engineering and Physical Sciences Research Council Centre for Predictive Modelling in Healthcare, University of ExeterExeter, UK
| |
Collapse
|
93
|
Mogilner A, Manhart A. Agent-based modeling: case study in cleavage furrow models. Mol Biol Cell 2016; 27:3379-3384. [PMID: 27811328 PMCID: PMC5221574 DOI: 10.1091/mbc.e16-01-0013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 08/29/2016] [Accepted: 08/31/2016] [Indexed: 12/30/2022] Open
Abstract
The number of studies in cell biology in which quantitative models accompany experiments has been growing steadily. Roughly, mathematical and computational techniques of these models can be classified as "differential equation based" (DE) or "agent based" (AB). Recently AB models have started to outnumber DE models, but understanding of AB philosophy and methodology is much less widespread than familiarity with DE techniques. Here we use the history of modeling a fundamental biological problem-positioning of the cleavage furrow in dividing cells-to explain how and why DE and AB models are used. We discuss differences, advantages, and shortcomings of these two approaches.
Collapse
Affiliation(s)
- Alex Mogilner
- Courant Institute and Department of Biology, New York University, New York, NY 10012
| | - Angelika Manhart
- Courant Institute and Department of Biology, New York University, New York, NY 10012
| |
Collapse
|
94
|
Zhu X, Deng J, Zuo Z, Lam TN. An Agent-Based Approach to Dynamically Represent the Pharmacokinetic Properties of Baicalein. AAPS JOURNAL 2016; 18:1475-1488. [PMID: 27480317 DOI: 10.1208/s12248-016-9955-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 07/06/2016] [Indexed: 11/30/2022]
Abstract
Baicalein, a typical flavonoid presented in Scutellariae radix, exhibits a unique metabolic profile during first-pass metabolism: parallel glucuronidation and sulfation pathways, with possible substrate inhibition in both pathways. In this project, we aimed to construct an agent-based model to dynamically represent baicalein pharmacokinetics and to verify the substrate inhibition hypothesis. The model consisted of three 3D spaces and two membranes: apical space (S1), intracellular space (S2), basolateral space (S3), apical membrane (M1), and basolateral membrane (M2). In silico enzymes (UDP-glucuronosyltransferases (UGTs) and sulfotransferases (SULTs)) and binder components were placed in S2. The model was then executed to simulate one-pass metabolism experiments of baicalein. With the implementation of a two-site enzyme design, the simulated results captured the preset qualitative and quantitative features of the wet-lab observations. The feasible parameter set showed that substrate inhibition happened in both conjugation pathways of baicalein. The simulation results suggested that the sulfation pathway was dominant at low concentrations and that SULT was more inclined to substrate inhibition than UGT. Cross-model validation was satisfactory. Our findings were consistent with a previously reported catenary model. We conclude that the mechanisms represented by our model are plausible. Our novel modeling approach could dynamically represent the metabolic pathways of baicalein in a Caco-2 system.
Collapse
Affiliation(s)
- Xiao Zhu
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Jianyuan Deng
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Zhong Zuo
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong
| | - Tai Ning Lam
- School of Pharmacy, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong.
| |
Collapse
|
95
|
Lux SA, Wnuk A, Vogt H, Belien T, Spornberger A, Studnicki M. Validation of Individual-Based Markov-Like Stochastic Process Model of Insect Behavior and a "Virtual Farm" Concept for Enhancement of Site-Specific IPM. Front Physiol 2016; 7:363. [PMID: 27602000 PMCID: PMC4993809 DOI: 10.3389/fphys.2016.00363] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 08/08/2016] [Indexed: 11/13/2022] Open
Abstract
The paper reports application of a Markov-like stochastic process agent-based model and a "virtual farm" concept for enhancement of site-specific Integrated Pest Management. Conceptually, the model represents a "bottom-up ethological" approach and emulates behavior of the "primary IPM actors"-large cohorts of individual insects-within seasonally changing mosaics of spatiotemporally complex faming landscape, under the challenge of the local IPM actions. Algorithms of the proprietary PESTonFARM model were adjusted to reflect behavior and ecology of R. cerasi. Model parametrization was based on compiled published information about R. cerasi and the results of auxiliary on-farm experiments. The experiments were conducted on sweet cherry farms located in Austria, Germany, and Belgium. For each farm, a customized model-module was prepared, reflecting its spatiotemporal features. Historical data about pest monitoring, IPM treatments and fruit infestation were used to specify the model assumptions and calibrate it further. Finally, for each of the farms, virtual IPM experiments were simulated and the model-generated results were compared with the results of the real experiments conducted on the same farms. Implications of the findings for broader applicability of the model and the "virtual farm" approach-were discussed.
Collapse
Affiliation(s)
- Slawomir A Lux
- inSilico-IPMKonstancin-Jeziorna, Poland; Department of Applied Entomology, Warsaw University of Life SciencesWarsaw, Poland
| | - Andrzej Wnuk
- Department of Applied Entomology, Warsaw University of Life Sciences Warsaw, Poland
| | - Heidrun Vogt
- Julius Kühn-Institut, Federal Research Centre for Cultivated Plants, Institute for Plant Protection in Fruit Crops and Viticulture Dossenheim, Germany
| | - Tim Belien
- Department of Zoology, pcfruit vzw Sint-Truiden, Belgium
| | - Andreas Spornberger
- Division of Viticulture and Pomology, University of Natural Resources and Life Sciences Vienna, Austria
| | - Marcin Studnicki
- Department of Experimental Design and Bioinformatics, Warsaw University of Life Sciences Warsaw, Poland
| |
Collapse
|
96
|
Shi Z, Chapes SK, Ben-Arieh D, Wu CH. An Agent-Based Model of a Hepatic Inflammatory Response to Salmonella: A Computational Study under a Large Set of Experimental Data. PLoS One 2016; 11:e0161131. [PMID: 27556404 PMCID: PMC4996536 DOI: 10.1371/journal.pone.0161131] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/29/2016] [Indexed: 01/04/2023] Open
Abstract
We present an agent-based model (ABM) to simulate a hepatic inflammatory response (HIR) in a mouse infected by Salmonella that sometimes progressed to problematic proportions, known as "sepsis". Based on over 200 published studies, this ABM describes interactions among 21 cells or cytokines and incorporates 226 experimental data sets and/or data estimates from those reports to simulate a mouse HIR in silico. Our simulated results reproduced dynamic patterns of HIR reported in the literature. As shown in vivo, our model also demonstrated that sepsis was highly related to the initial Salmonella dose and the presence of components of the adaptive immune system. We determined that high mobility group box-1, C-reactive protein, and the interleukin-10: tumor necrosis factor-α ratio, and CD4+ T cell: CD8+ T cell ratio, all recognized as biomarkers during HIR, significantly correlated with outcomes of HIR. During therapy-directed silico simulations, our results demonstrated that anti-agent intervention impacted the survival rates of septic individuals in a time-dependent manner. By specifying the infected species, source of infection, and site of infection, this ABM enabled us to reproduce the kinetics of several essential indicators during a HIR, observe distinct dynamic patterns that are manifested during HIR, and allowed us to test proposed therapy-directed treatments. Although limitation still exists, this ABM is a step forward because it links underlying biological processes to computational simulation and was validated through a series of comparisons between the simulated results and experimental studies.
Collapse
Affiliation(s)
- Zhenzhen Shi
- Health Care Operations Resource Center, Department of Industrial and Manufacturing Systems Engineering, Kansas State University, Manhattan, Kansas, United States of America
| | - Stephen K. Chapes
- Division of Biology, Kansas State University, Manhattan, Kansas, United States of America
| | - David Ben-Arieh
- Health Care Operations Resource Center, Department of Industrial and Manufacturing Systems Engineering, Kansas State University, Manhattan, Kansas, United States of America
| | - Chih-Hang Wu
- Health Care Operations Resource Center, Department of Industrial and Manufacturing Systems Engineering, Kansas State University, Manhattan, Kansas, United States of America
| |
Collapse
|
97
|
Yankeelov TE, An G, Saut O, Luebeck EG, Popel AS, Ribba B, Vicini P, Zhou X, Weis JA, Ye K, Genin GM. Multi-scale Modeling in Clinical Oncology: Opportunities and Barriers to Success. Ann Biomed Eng 2016; 44:2626-41. [PMID: 27384942 DOI: 10.1007/s10439-016-1691-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 06/29/2016] [Indexed: 12/11/2022]
Abstract
Hierarchical processes spanning several orders of magnitude of both space and time underlie nearly all cancers. Multi-scale statistical, mathematical, and computational modeling methods are central to designing, implementing and assessing treatment strategies that account for these hierarchies. The basic science underlying these modeling efforts is maturing into a new discipline that is close to influencing and facilitating clinical successes. The purpose of this review is to capture the state-of-the-art as well as the key barriers to success for multi-scale modeling in clinical oncology. We begin with a summary of the long-envisioned promise of multi-scale modeling in clinical oncology, including the synthesis of disparate data types into models that reveal underlying mechanisms and allow for experimental testing of hypotheses. We then evaluate the mathematical techniques employed most widely and present several examples illustrating their application as well as the current gap between pre-clinical and clinical applications. We conclude with a discussion of what we view to be the key challenges and opportunities for multi-scale modeling in clinical oncology.
Collapse
Affiliation(s)
- Thomas E Yankeelov
- Departments of Biomedical Engineering and Internal Medicine, Institute for Computational and Engineering Sciences, Cockrell School of Engineering, The University of Texas at Austin, 107 W. Dean Keeton, BME Building, 1 University Station, C0800, Austin, TX, 78712, USA.
| | - Gary An
- Department of Surgery and Computation Institute, The University of Chicago, Chicago, IL, USA
| | - Oliver Saut
- Institut de Mathématiques de Bordeaux, Université de Bordeaux and INRIA, Bordeaux, France
| | - E Georg Luebeck
- Program in Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Aleksander S Popel
- Departments of Biomedical Engineering and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Benjamin Ribba
- Pharma Research and Early Development, Clinical Pharmacology, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Paolo Vicini
- Clinical Pharmacology and DMPK, MedImmune, Gaithersburg, MD, USA
| | - Xiaobo Zhou
- Center for Bioinformatics and Systems Biology, Radiology, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Jared A Weis
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kaiming Ye
- Department of Biomedical Engineering, Watson School of Engineering and Applied Science, Binghamton University, State University of New York, Binghamton, NY, USA
| | - Guy M Genin
- Departments of Mechanical Engineering and Materials Science, and Neurological Surgery, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
98
|
Warsinske HC, Wheaton AK, Kim KK, Linderman JJ, Moore BB, Kirschner DE. Computational Modeling Predicts Simultaneous Targeting of Fibroblasts and Epithelial Cells Is Necessary for Treatment of Pulmonary Fibrosis. Front Pharmacol 2016; 7:183. [PMID: 27445819 PMCID: PMC4917547 DOI: 10.3389/fphar.2016.00183] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/10/2016] [Indexed: 11/13/2022] Open
Abstract
Pulmonary fibrosis is pathologic remodeling of lung tissue that can result in difficulty breathing, reduced quality of life, and a poor prognosis for patients. Fibrosis occurs as a result of insult to lung tissue, though mechanisms of this response are not well-characterized. The disease is driven in part by dysregulation of fibroblast proliferation and differentiation into myofibroblast cells, as well as pro-fibrotic mediator-driven epithelial cell apoptosis. The most well-characterized pro-fibrotic mediator associated with pulmonary fibrosis is TGF-β1. Excessive synthesis of, and sensitivity to, pro-fibrotic mediators as well as insufficient production of and sensitivity to anti-fibrotic mediators has been credited with enabling fibroblast accumulation. Available treatments neither halt nor reverse lung damage. In this study we have two aims: to identify molecular and cellular scale mechanisms driving fibroblast proliferation and differentiation as well as epithelial cell survival in the context of fibrosis, and to predict therapeutic targets and strategies. We combine in vitro studies with a multi-scale hybrid agent-based computational model that describes fibroblasts and epithelial cells in co-culture. Within this model TGF-β1 represents a pro-fibrotic mediator and we include detailed dynamics of TGF-β1 receptor ligand signaling in fibroblasts. PGE2 represents an anti-fibrotic mediator. Using uncertainty and sensitivity analysis we identify TGF-β1 synthesis, TGF-β1 activation, and PGE2 synthesis among the key mechanisms contributing to fibrotic outcomes. We further demonstrate that intervention strategies combining potential therapeutics targeting both fibroblast regulation and epithelial cell survival can promote healthy tissue repair better than individual strategies. Combinations of existing drugs and compounds may provide significant improvements to the current standard of care for pulmonary fibrosis. Thus, a two-hit therapeutic intervention strategy may prove necessary to halt and reverse disease dynamics.
Collapse
Affiliation(s)
- Hayley C. Warsinske
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Amanda K. Wheaton
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Kevin K. Kim
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | | | - Bethany B. Moore
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
- Department of Internal Medicine, University of Michigan Medical SchoolAnn Arbor, MI, USA
| | - Denise E. Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical SchoolAnn Arbor, MI, USA
| |
Collapse
|
99
|
Multi-scale Modeling of the Cardiovascular System: Disease Development, Progression, and Clinical Intervention. Ann Biomed Eng 2016; 44:2642-60. [PMID: 27138523 DOI: 10.1007/s10439-016-1628-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 04/22/2016] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death in the western world. With the current development of clinical diagnostics to more accurately measure the extent and specifics of CVDs, a laudable goal is a better understanding of the structure-function relation in the cardiovascular system. Much of this fundamental understanding comes from the development and study of models that integrate biology, medicine, imaging, and biomechanics. Information from these models provides guidance for developing diagnostics, and implementation of these diagnostics to the clinical setting, in turn, provides data for refining the models. In this review, we introduce multi-scale and multi-physical models for understanding disease development, progression, and designing clinical interventions. We begin with multi-scale models of cardiac electrophysiology and mechanics for diagnosis, clinical decision support, personalized and precision medicine in cardiology with examples in arrhythmia and heart failure. We then introduce computational models of vasculature mechanics and associated mechanical forces for understanding vascular disease progression, designing clinical interventions, and elucidating mechanisms that underlie diverse vascular conditions. We conclude with a discussion of barriers that must be overcome to provide enhanced insights, predictions, and decisions in pre-clinical and clinical applications.
Collapse
|
100
|
Pienaar E, Matern WM, Linderman JJ, Bader JS, Kirschner DE. Multiscale Model of Mycobacterium tuberculosis Infection Maps Metabolite and Gene Perturbations to Granuloma Sterilization Predictions. Infect Immun 2016; 84:1650-1669. [PMID: 26975995 PMCID: PMC4862722 DOI: 10.1128/iai.01438-15] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/08/2016] [Indexed: 02/06/2023] Open
Abstract
Granulomas are a hallmark of tuberculosis. Inside granulomas, the pathogen Mycobacterium tuberculosis may enter a metabolically inactive state that is less susceptible to antibiotics. Understanding M. tuberculosis metabolism within granulomas could contribute to reducing the lengthy treatment required for tuberculosis and provide additional targets for new drugs. Two key adaptations of M. tuberculosis are a nonreplicating phenotype and accumulation of lipid inclusions in response to hypoxic conditions. To explore how these adaptations influence granuloma-scale outcomes in vivo, we present a multiscale in silico model of granuloma formation in tuberculosis. The model comprises host immunity, M. tuberculosis metabolism, M. tuberculosis growth adaptation to hypoxia, and nutrient diffusion. We calibrated our model to in vivo data from nonhuman primates and rabbits and apply the model to predict M. tuberculosis population dynamics and heterogeneity within granulomas. We found that bacterial populations are highly dynamic throughout infection in response to changing oxygen levels and host immunity pressures. Our results indicate that a nonreplicating phenotype, but not lipid inclusion formation, is important for long-term M. tuberculosis survival in granulomas. We used virtual M. tuberculosis knockouts to predict the impact of both metabolic enzyme inhibitors and metabolic pathways exploited to overcome inhibition. Results indicate that knockouts whose growth rates are below ∼66% of the wild-type growth rate in a culture medium featuring lipid as the only carbon source are unable to sustain infections in granulomas. By mapping metabolite- and gene-scale perturbations to granuloma-scale outcomes and predicting mechanisms of sterilization, our method provides a powerful tool for hypothesis testing and guiding experimental searches for novel antituberculosis interventions.
Collapse
Affiliation(s)
- Elsje Pienaar
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - William M Matern
- Department of Biomedical Engineering and High-Throughput Biology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jennifer J Linderman
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Joel S Bader
- Department of Biomedical Engineering and High-Throughput Biology Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Denise E Kirschner
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|