1
|
Urdy S, Hanke M, Toledo AI, Ratto N, Jacob E, Peyronnet E, Gourlet JB, Chaves SS, Thommes E, Coudeville L, Boissel JP, Courcelles E, Bruezière L. Multi-strain modeling of influenza vaccine effectiveness in older adults and its dependence on antigenic distance. Sci Rep 2024; 14:27190. [PMID: 39516205 PMCID: PMC11549341 DOI: 10.1038/s41598-024-72716-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 09/10/2024] [Indexed: 11/16/2024] Open
Abstract
Influenza vaccine effectiveness (VE) varies seasonally due to host, virus and vaccine characteristics. To investigate how antigenic matching and dosage impact VE, we developed a mechanistic knowledge-based mathematical model. Immunization with a split vaccine is modeled for exposure to A/H1N1 or A/H3N2 virus strains. The model accounts for cross-reactivity of immune cells elicited during previous immunizations with new antigens. We simulated vaccine effectiveness (sVE) of high dose (HD) versus standard dose (SD) vaccines in the older population, from 2011 to 2022. We find that sVE is highly dependent on antigenic matching and that higher dosage improves immunogenicity, activation and memory formation of immune cells. In alignment with clinical observations, the HD vaccine performs better than the SD vaccine in all simulations, supporting the use of the HD vaccine in the older population. This model could be adapted to predict the impact of alternative virus strain selection on clinical outcomes in future influenza seasons.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sandra S Chaves
- Modeling, Edpidemiology and Data Science (MEDS), Sanofi Vaccines, Lyon, France
| | - Edward Thommes
- Modeling, Edpidemiology and Data Science (MEDS), Sanofi Vaccines, Lyon, France
| | - Laurent Coudeville
- Modeling, Edpidemiology and Data Science (MEDS), Sanofi Vaccines, Lyon, France
| | | | | | | |
Collapse
|
2
|
Whipple B, Miura TA, Hernandez-Vargas EA. Modeling the CD8+ T cell immune response to influenza infection in adult and aged mice. J Theor Biol 2024; 593:111898. [PMID: 38996911 PMCID: PMC11348945 DOI: 10.1016/j.jtbi.2024.111898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
The CD8+ T cell response is the main determinant of viral clearance during influenza infection. However, influenza viral dynamics and the respective immune responses are affected by the host's age. To investigate age-related differences in the CD8+ T cell immune response dynamics, we propose 16 ordinary differential equation models of existing experimental data. These data consist of viral titer and CD8+ T cell counts collected periodically over a period of 19 days from adult and aged mice infected with influenza A/Puerto Rico/8/34 (H1N1). We use the corrected Akaike Information Criterion to identify the models which best represent the considered data. Our model selection process indicates differences in mechanisms which reduce the CD8+ T cell response: linear downregulation is favored for adult mice, while baseline exponential decay is favored for aged mice. Parameter fitting of the top ranked models suggests that the aged population has reduced CD8+ T cell proliferation compared to the adult population. More experimental work is needed to determine the specific immunological features through which age might cause these differences. A better understanding of the immunological mechanisms by which aging leads to discrepant CD8+ T cell dynamics may inform future treatment strategies.
Collapse
Affiliation(s)
- Benjamin Whipple
- Department of Mathematics and Statistical Science, University of Idaho, Moscow, ID, 83844, United States; Bioinformatics and Computational Biology Program, University of Idaho, Moscow, ID, 83844, United States
| | - Tanya A Miura
- Bioinformatics and Computational Biology Program, University of Idaho, Moscow, ID, 83844, United States; Department of Biological Sciences, University of Idaho, Moscow, ID, 83844, United States; Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, ID, 83844, United States
| | - Esteban A Hernandez-Vargas
- Department of Mathematics and Statistical Science, University of Idaho, Moscow, ID, 83844, United States; Bioinformatics and Computational Biology Program, University of Idaho, Moscow, ID, 83844, United States; Institute for Modeling Collaboration and Innovation, University of Idaho, Moscow, ID, 83844, United States.
| |
Collapse
|
3
|
Gazeau S, Deng X, Ooi HK, Mostefai F, Hussin J, Heffernan J, Jenner AL, Craig M. The race to understand immunopathology in COVID-19: Perspectives on the impact of quantitative approaches to understand within-host interactions. IMMUNOINFORMATICS (AMSTERDAM, NETHERLANDS) 2023; 9:100021. [PMID: 36643886 PMCID: PMC9826539 DOI: 10.1016/j.immuno.2023.100021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/16/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
The COVID-19 pandemic has revealed the need for the increased integration of modelling and data analysis to public health, experimental, and clinical studies. Throughout the first two years of the pandemic, there has been a concerted effort to improve our understanding of the within-host immune response to the SARS-CoV-2 virus to provide better predictions of COVID-19 severity, treatment and vaccine development questions, and insights into viral evolution and the impacts of variants on immunopathology. Here we provide perspectives on what has been accomplished using quantitative methods, including predictive modelling, population genetics, machine learning, and dimensionality reduction techniques, in the first 26 months of the COVID-19 pandemic approaches, and where we go from here to improve our responses to this and future pandemics.
Collapse
Affiliation(s)
- Sonia Gazeau
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada
- Sainte-Justine University Hospital Research Centre, Montréal, Canada
| | - Xiaoyan Deng
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada
- Sainte-Justine University Hospital Research Centre, Montréal, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, Toronto, Canada
| | - Fatima Mostefai
- Montréal Heart Institute Research Centre, Montréal, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Julie Hussin
- Montréal Heart Institute Research Centre, Montréal, Canada
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Canada
| | - Jane Heffernan
- Modelling Infection and Immunity Lab, Mathematics Statistics, York University, Toronto, Canada
- Centre for Disease Modelling (CDM), Mathematics Statistics, York University, Toronto, Canada
| | - Adrianne L Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane Australia
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal, Montréal, Canada
- Sainte-Justine University Hospital Research Centre, Montréal, Canada
| |
Collapse
|
4
|
Sumi T, Harada K. Immune response to SARS-CoV-2 in severe disease and long COVID-19. iScience 2022; 25:104723. [PMID: 35813874 PMCID: PMC9251893 DOI: 10.1016/j.isci.2022.104723] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 05/23/2022] [Accepted: 06/29/2022] [Indexed: 01/10/2023] Open
Abstract
COVID-19 is mild to moderate in otherwise healthy individuals but may nonetheless cause life-threatening disease and/or a wide range of persistent symptoms. The general determinant of disease severity is age mainly because the immune response declines in aging patients. Here, we developed a mathematical model of the immune response to SARS-CoV-2 and revealed that typical age-related risk factors such as only a several 10% decrease in innate immune cell activity and inhibition of type-I interferon signaling by autoantibodies drastically increased the viral load. It was reported that the numbers of certain dendritic cell subsets remained less than half those in healthy donors even seven months after infection. Hence, the inflammatory response was ongoing. Our model predicted the persistent DC reduction and showed that certain patients with severe and even mild symptoms could not effectively eliminate the virus and could potentially develop long COVID.
Collapse
Affiliation(s)
- Tomonari Sumi
- Research Institute for Interdisciplinary Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
- Department of Chemistry, Faculty of Science, Okayama University, 3-1-1 Tsushima-Naka, Kita-ku, Okayama 700-8530, Japan
| | - Kouji Harada
- Department of Computer Science and Engineering, Toyohashi University of Technology, Tempaku-cho, Toyohashi 441-8580, Japan
- Center for IT-Based Education, Toyohashi University of Technology, Tempaku-cho, Toyohashi, Aichi 441-8580, Japan
| |
Collapse
|
5
|
Chatterjee B, Singh Sandhu H, Dixit NM. Modeling recapitulates the heterogeneous outcomes of SARS-CoV-2 infection and quantifies the differences in the innate immune and CD8 T-cell responses between patients experiencing mild and severe symptoms. PLoS Pathog 2022; 18:e1010630. [PMID: 35759522 PMCID: PMC9269964 DOI: 10.1371/journal.ppat.1010630] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 07/08/2022] [Accepted: 06/01/2022] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 infection results in highly heterogeneous outcomes, from cure without symptoms to acute respiratory distress and death. Empirical evidence points to the prominent roles of innate immune and CD8 T-cell responses in determining the outcomes. However, how these immune arms act in concert to elicit the outcomes remains unclear. Here, we developed a mathematical model of within-host SARS-CoV-2 infection that incorporates the essential features of the innate immune and CD8 T-cell responses. Remarkably, by varying the strengths and timings of the two immune arms, the model recapitulated the entire spectrum of outcomes realized. Furthermore, model predictions offered plausible explanations of several confounding clinical observations, including the occurrence of multiple peaks in viral load, viral recrudescence after symptom loss, and prolonged viral positivity. We applied the model to analyze published datasets of longitudinal viral load measurements from patients exhibiting diverse outcomes. The model provided excellent fits to the data. The best-fit parameter estimates indicated a nearly 80-fold stronger innate immune response and an over 200-fold more sensitive CD8 T-cell response in patients with mild compared to severe infection. These estimates provide quantitative insights into the likely origins of the dramatic inter-patient variability in the outcomes of SARS-CoV-2 infection. The insights have implications for interventions aimed at preventing severe disease and for understanding the differences between viral variants.
Collapse
Affiliation(s)
- Budhaditya Chatterjee
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| | | | - Narendra M. Dixit
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
6
|
Benest J, Rhodes S, Evans TG, White RG. Mathematical Modelling for Optimal Vaccine Dose Finding: Maximising Efficacy and Minimising Toxicity. Vaccines (Basel) 2022; 10:vaccines10050756. [PMID: 35632511 PMCID: PMC9144167 DOI: 10.3390/vaccines10050756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/03/2022] [Accepted: 05/06/2022] [Indexed: 02/06/2023] Open
Abstract
Vaccination is a key tool to reduce global disease burden. Vaccine dose can affect vaccine efficacy and toxicity. Given the expense of developing vaccines, optimising vaccine dose is essential. Mathematical modelling has been suggested as an approach for optimising vaccine dose by quantitatively establishing the relationships between dose and efficacy/toxicity. In this work, we performed simulation studies to assess the performance of modelling approaches in determining optimal dose. We found that the ability of modelling approaches to determine optimal dose improved with trial size, particularly for studies with at least 30 trial participants, and that, generally, using a peaking or a weighted model-averaging-based dose–efficacy relationship was most effective in finding optimal dose. Most methods of trial dose selection were similarly effective for the purpose of determining optimal dose; however, including modelling to adapt doses during a trial may lead to more trial participants receiving a more optimal dose. Clinical trial dosing around the predicted optimal dose, rather than only at the predicted optimal dose, may improve final dose selection. This work suggests modelling can be used effectively for vaccine dose finding, prompting potential practical applications of these methods in accelerating effective vaccine development and saving lives.
Collapse
Affiliation(s)
- John Benest
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; (S.R.); (R.G.W.)
- Correspondence:
| | - Sophie Rhodes
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; (S.R.); (R.G.W.)
| | - Thomas G. Evans
- Vaccitech Ltd., The Schrodinger Building, Heatley Road, The Oxford Science Park, Oxford OX4 4GE, UK;
| | - Richard G. White
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; (S.R.); (R.G.W.)
| |
Collapse
|
7
|
Padmanabhan P, Desikan R, Dixit NM. Modeling how antibody responses may determine the efficacy of COVID-19 vaccines. NATURE COMPUTATIONAL SCIENCE 2022; 2:123-131. [PMID: 38177523 DOI: 10.1038/s43588-022-00198-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 01/20/2022] [Indexed: 01/06/2024]
Abstract
Predicting the efficacy of COVID-19 vaccines would aid vaccine development and usage strategies, which is of importance given their limited supplies. Here we develop a multiscale mathematical model that proposes mechanistic links between COVID-19 vaccine efficacies and the neutralizing antibody (NAb) responses they elicit. We hypothesized that the collection of all NAbs would constitute a shape space and that responses of individuals are random samples from this space. We constructed the shape space by analyzing reported in vitro dose-response curves of ~80 NAbs. Sampling NAb subsets from the space, we recapitulated the responses of convalescent patients. We assumed that vaccination would elicit similar NAb responses. We developed a model of within-host SARS-CoV-2 dynamics, applied it to virtual patient populations and, invoking the NAb responses above, predicted vaccine efficacies. Our predictions quantitatively captured the efficacies from clinical trials. Our study thus suggests plausible mechanistic underpinnings of COVID-19 vaccines and generates testable hypotheses for establishing them.
Collapse
Affiliation(s)
- Pranesh Padmanabhan
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Queensland, Australia.
| | - Rajat Desikan
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
- Certara QSP, Certara UK Limited, Sheffield, UK
| | - Narendra M Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India.
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India.
| |
Collapse
|
8
|
Day JD, Park S, Ranard BL, Singh H, Chow CC, Vodovotz Y. Divergent COVID-19 Disease Trajectories Predicted by a DAMP-Centered Immune Network Model. Front Immunol 2021; 12:754127. [PMID: 34777366 PMCID: PMC8582279 DOI: 10.3389/fimmu.2021.754127] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 10/04/2021] [Indexed: 01/08/2023] Open
Abstract
COVID-19 presentations range from mild to moderate through severe disease but also manifest with persistent illness or viral recrudescence. We hypothesized that the spectrum of COVID-19 disease manifestations was a consequence of SARS-CoV-2-mediated delay in the pathogen-associated molecular pattern (PAMP) response, including dampened type I interferon signaling, thereby shifting the balance of the immune response to be dominated by damage-associated molecular pattern (DAMP) signaling. To test the hypothesis, we constructed a parsimonious mechanistic mathematical model. After calibration of the model for initial viral load and then by varying a few key parameters, we show that the core model generates four distinct viral load, immune response and associated disease trajectories termed “patient archetypes”, whose temporal dynamics are reflected in clinical data from hospitalized COVID-19 patients. The model also accounts for responses to corticosteroid therapy and predicts that vaccine-induced neutralizing antibodies and cellular memory will be protective, including from severe COVID-19 disease. This generalizable modeling framework could be used to analyze protective and pathogenic immune responses to diverse viral infections.
Collapse
Affiliation(s)
- Judy D Day
- Department of Mathematics, University of Tennessee, Knoxville, TN, United States.,Department of Electrical Engineering & Computer Science, University of Tennessee, Knoxville, TN, United States
| | - Soojin Park
- Department of Neurology & Division of Critical Care and Hospital Neurology, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital - Columbia University Irving Medical Center, New York, NY, United States.,Program for Hospital and Intensive Care Informatics, Department of Neurology, Columbia University College of Physicians and Surgeons, New York, NY, United States
| | - Benjamin L Ranard
- Program for Hospital and Intensive Care Informatics, Department of Neurology, Columbia University College of Physicians and Surgeons, New York, NY, United States.,Division of Pulmonary, Allergy & Critical Care Medicine, Department of Medicine, Columbia University College of Physicians and Surgeons, New York Presbyterian Hospital - Columbia University Irving Medical Center, New York, NY, United States
| | - Harinder Singh
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Carson C Chow
- Mathematical Biology Section, Laboratory of Biological Modeling, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, United States
| | - Yoram Vodovotz
- Center for Systems Immunology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Inflammation and Regeneration Modeling, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Pascucci E, Pugliese A. Modelling Immune Memory Development. Bull Math Biol 2021; 83:118. [PMID: 34687362 DOI: 10.1007/s11538-021-00949-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 09/27/2021] [Indexed: 12/01/2022]
Abstract
The cellular adaptive immune response to influenza has been analyzed through several recent mathematical models. In particular, Zarnitsyna et al. (Front Immunol 7:1-9, 2016) show how central memory CD8+ T cells reach a plateau after repeated infections, and analyze their role in the immune response to further challenges. In this paper, we further investigate the theoretical features of that model by extracting from the infection dynamics a discrete map that describes the build-up of memory cells. Furthermore, we show how the model by Zarnitsyna et al. (Front Immunol 7:1-9, 2016) can be viewed as a fast-scale approximation of a model allowing for recruitment of target epithelial cells. Finally, we analyze which components of the model are essential to understand the progressive build-up of immune memory. This is performed through the analysis of simplified versions of the model that include some components only of immune response. The analysis performed may also provide a theoretical framework for understanding the conditions under which two-dose vaccination strategies can be helpful.
Collapse
Affiliation(s)
- Eleonora Pascucci
- Dipartimento di Matematica, Università degli Studi di Trento, Via Sommarive 14, 38123, Povo, TN, Italy
| | - Andrea Pugliese
- Dipartimento di Matematica, Università degli Studi di Trento, Via Sommarive 14, 38123, Povo, TN, Italy.
| |
Collapse
|
10
|
Alamo T, G Reina D, Millán Gata P, Preciado VM, Giordano G. Data-driven methods for present and future pandemics: Monitoring, modelling and managing. ANNUAL REVIEWS IN CONTROL 2021; 52:448-464. [PMID: 34220287 PMCID: PMC8238691 DOI: 10.1016/j.arcontrol.2021.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/24/2021] [Accepted: 05/27/2021] [Indexed: 05/29/2023]
Abstract
This survey analyses the role of data-driven methodologies for pandemic modelling and control. We provide a roadmap from the access to epidemiological data sources to the control of epidemic phenomena. We review the available methodologies and discuss the challenges in the development of data-driven strategies to combat the spreading of infectious diseases. Our aim is to bring together several different disciplines required to provide a holistic approach to epidemic analysis, such as data science, epidemiology, and systems-and-control theory. A 3M-analysis is presented, whose three pillars are: Monitoring, Modelling and Managing. The focus is on the potential of data-driven schemes to address three different challenges raised by a pandemic: (i) monitoring the epidemic evolution and assessing the effectiveness of the adopted countermeasures; (ii) modelling and forecasting the spread of the epidemic; (iii) making timely decisions to manage, mitigate and suppress the contagion. For each step of this roadmap, we review consolidated theoretical approaches (including data-driven methodologies that have been shown to be successful in other contexts) and discuss their application to past or present epidemics, such as Covid-19, as well as their potential application to future epidemics.
Collapse
Affiliation(s)
- Teodoro Alamo
- Departamento de Ingeniería de Sistemas y Automática, Universidad de Sevilla, Escuela Superior de Ingenieros, Sevilla, Spain
| | - Daniel G Reina
- Departamento de Ingeniería Electrónica, Universidad de Sevilla, Escuela Superior de Ingenieros, Sevilla, Spain
| | - Pablo Millán Gata
- Departamento de Ingeniería, Universidad Loyola Andalucía, Seville, Spain
| | - Victor M Preciado
- Department of Electrical and Systems Engineering, University of Pennsylvania, Philadelphia, USA
| | - Giulia Giordano
- Department of Industrial Engineering, University of Trento, Trento, Italy
| |
Collapse
|
11
|
Barros LRC, Paixão EA, Valli AMP, Naozuka GT, Fassoni AC, Almeida RC. CART math-A Mathematical Model of CAR-T Immunotherapy in Preclinical Studies of Hematological Cancers. Cancers (Basel) 2021; 13:2941. [PMID: 34208323 PMCID: PMC8231202 DOI: 10.3390/cancers13122941] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/05/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy has gained great momentum with chimeric antigen receptor T cell (CAR-T) therapy, in which patient's T lymphocytes are genetically manipulated to recognize tumor-specific antigens, increasing tumor elimination efficiency. In recent years, CAR-T cell immunotherapy for hematological malignancies achieved a great response rate in patients and is a very promising therapy for several other malignancies. Each new CAR design requires a preclinical proof-of-concept experiment using immunodeficient mouse models. The absence of a functional immune system in these mice makes them simple and suitable for use as mathematical models. In this work, we develop a three-population mathematical model to describe tumor response to CAR-T cell immunotherapy in immunodeficient mouse models, encompassing interactions between a non-solid tumor and CAR-T cells (effector and long-term memory). We account for several phenomena, such as tumor-induced immunosuppression, memory pool formation, and conversion of memory into effector CAR-T cells in the presence of new tumor cells. Individual donor and tumor specificities are considered uncertainties in the model parameters. Our model is able to reproduce several CAR-T cell immunotherapy scenarios, with different CAR receptors and tumor targets reported in the literature. We found that therapy effectiveness mostly depends on specific parameters such as the differentiation of effector to memory CAR-T cells, CAR-T cytotoxic capacity, tumor growth rate, and tumor-induced immunosuppression. In summary, our model can contribute to reducing and optimizing the number of in vivo experiments with in silico tests to select specific scenarios that could be tested in experimental research. Such an in silico laboratory is an easy-to-run open-source simulator, built on a Shiny R-based platform called CARTmath. It contains the results of this manuscript as examples and documentation. The developed model together with the CARTmath platform have potential use in assessing different CAR-T cell immunotherapy protocols and its associated efficacy, becoming an accessory for in silico trials.
Collapse
Affiliation(s)
- Luciana R. C. Barros
- Center for Translational Research in Oncology, Instituto do Câncer do Estado de São Paulo, Hospital das Clínicas da Faculdade de Medicina ds Universidade de São Paulo, São Paulo 01246-000, Brazil
| | - Emanuelle A. Paixão
- Graduate Program, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil; (E.A.P.); (G.T.N.)
| | - Andrea M. P. Valli
- Computer Science Department, Universidade Federal do Espírito Santo, Vitória 29075-910, Brazil;
| | - Gustavo T. Naozuka
- Graduate Program, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil; (E.A.P.); (G.T.N.)
| | - Artur C. Fassoni
- Institute for Mathematics and Computer Science, Universidade Federal de Itajubá, Itajubá 37500-903, Brazil;
| | - Regina C. Almeida
- Computational Modeling Department, Laboratório Nacional de Computação Científica, Petrópolis 25651-075, Brazil;
| |
Collapse
|
12
|
Wyatt A, Levy D. Modeling the Effect of Memory in the Adaptive Immune Response. Bull Math Biol 2020; 82:124. [PMID: 32926238 DOI: 10.1007/s11538-020-00798-9] [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: 01/08/2020] [Accepted: 08/25/2020] [Indexed: 11/30/2022]
Abstract
It is well understood that there are key differences between a primary immune response and subsequent responses. Specifically, memory T cells that remain after a primary response drive the clearance of antigen in later encounters. While the existence of memory T cells is widely accepted, the specific mechanisms that govern their function are generally debated. In this paper, we develop a mathematical model of the immune response. This model follows the creation, activation, and regulation of memory T cells, which allows us to explore the differences between the primary and secondary immune responses. Through the incorporation of memory T cells, we demonstrate how the immune system can mount a faster and more effective secondary response. This mathematical model provides a quantitative framework for studying chronic infections and auto-immune diseases.
Collapse
Affiliation(s)
- Asia Wyatt
- Department of Mathematics, University of Maryland, College Park, MD, 20742, USA
| | - Doron Levy
- Department of Mathematics and Center for Scientific Computation and Mathematical Modeling (CSCAMM), University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
13
|
Morris SE, Farber DL, Yates AJ. Tissue-Resident Memory T Cells in Mice and Humans: Towards a Quantitative Ecology. THE JOURNAL OF IMMUNOLOGY 2020; 203:2561-2569. [PMID: 31685700 DOI: 10.4049/jimmunol.1900767] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022]
Abstract
In recent years, tissue-resident memory T cells (TRM) have emerged as essential components of immunological memory. Following antigenic challenge, TRM remain in nonlymphoid tissues and defend against re-exposure. Although accumulating evidence suggests important roles for TRM in mediating protective immunity, fundamental aspects of the population biology of TRM remain poorly understood. In this article, we discuss how results from different systems shed light on the ecological dynamics of TRM in mice and humans. We highlight the importance of dissecting processes contributing to TRM maintenance, and how these might vary across phenotypically and spatially heterogeneous subsets. We also discuss how the diversity of TRM communities within specific tissues may evolve under competition and in response to antigenic perturbation. Throughout, we illustrate how mathematical models can clarify inferences obtained from experimental data and help elucidate the homeostatic mechanisms underpinning the ecology of TRM populations.
Collapse
Affiliation(s)
- Sinead E Morris
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032
| | - Donna L Farber
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032.,Department of Microbiology and Immunology, Columbia University Medical Center, New York, NY 10032; and.,Department of Surgery, Columbia University Medical Center, New York, NY 10032
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032;
| |
Collapse
|
14
|
Personal response to immune checkpoint inhibitors of patients with advanced melanoma explained by a computational model of cellular immunity, tumor growth, and drug. PLoS One 2019; 14:e0226869. [PMID: 31877168 PMCID: PMC6932803 DOI: 10.1371/journal.pone.0226869] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 12/08/2019] [Indexed: 01/22/2023] Open
Abstract
Immune checkpoint inhibitors, such as pembrolizumab, are transforming clinical oncology. Yet, insufficient overall response rate, and accelerated tumor growth rate in some patients, highlight the need for identifying potential responders. To construct a computational model, identifying response predictors, and enabling immunotherapy personalization. The combined dynamics of cellular immunity, pembrolizumab, and the melanoma cancer were modeled by a set of ordinary differential equations. The model relies on a scheme of T memory stem cells, progressively differentiating into effector CD8+ T cells, and additionally includes T cell exhaustion, reinvigoration and senescence. Clinical data of a pembrolizumab-treated patient with advanced melanoma (Patient O’) were used for model calibration and simulations. Virtual patient populations, varying in one parameter or more, were generated for retrieving clinical studies. Simulations captured the major features of Patient O’s disease, displaying a good fit to her clinical data. A temporary increase in tumor burden, as implied by the clinical data, was obtained only when assuming aberrant self-renewal rates. Variation in effector T cell cytotoxicity was sufficient for simulating dynamics that vary from rapid progression to complete cure, while variation in tumor immunogenicity has a delayed and limited effect on response. Simulations of a-specific clinical trial were in good agreement with the clinical results, demonstrating positive correlations between response to pembrolizumab and the ratio of reinvigoration to baseline tumor load. These results were obtained by assuming inter-patient variation in the toxicity of effector CD8+ T cells, and in their intrinsic division rate, as well as by assuming that the intrinsic division rate of cancer cells is correlated with the baseline tumor burden. In conclusion, hyperprogression can result from lower patient-specific effector cytotoxicity, a temporary increase in tumor load is unlikely to result from real tumor growth, and the ratio of reinvigoration to tumor load can predict personal response to pembrolizumab. Upon further validation, the model can serve for immunotherapy personalization.
Collapse
|
15
|
Moore JR, Ahmed H, McGuire D, Akondy R, Ahmed R, Antia R. Dependence of CD8 T Cell Response upon Antigen Load During Primary Infection : Analysis of Data from Yellow Fever Vaccination. Bull Math Biol 2019; 81:2553-2568. [PMID: 31165405 PMCID: PMC6657775 DOI: 10.1007/s11538-019-00618-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Abstract
A major question in immunology is what role antigen load plays in determining the size of the CD8 immune response. Is the amount of antigen important during recruitment, proliferation, and/or memory formation? Animal studies have shown that antigen is only strictly required early during activation of T cells, but the importance of antigen at later timepoints is unclear. Using data from 24 volunteers infected with the yellow fever vaccine virus (YFV), we analyzed the dependence of T cell proliferation upon viral load. We found that volunteers with high viral load initially have greater T cell responses, but by 28 days post-vaccination those with lower viral load are able to 'catch-up.' Using differential equation modeling we show that this pattern is consistent with viral load only affecting recruitment (i.e., programmed proliferation) as opposed to affecting recruitment and proliferation (i.e., antigen-dependent proliferation). A quantitative understanding of the dependence of T cell dynamics on antigen load will be of use to modelers studying not only vaccination, but also cancer immunology and autoimmune disorders.
Collapse
Affiliation(s)
- James R Moore
- Division of Vaccines and Infectious Diseases, Fred Hutchinson Cancer Research Center, Seattle, USA.
| | - Hasan Ahmed
- Department of Biology, Emory University, Atlanta, USA
| | - Don McGuire
- Emory Vaccine Center, Emory University, Atlanta, USA
| | - Rama Akondy
- Department of Microbiology and Immunobiology, Emory University, Atlanta, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University, Atlanta, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, USA
| |
Collapse
|
16
|
Why Are CD8 T Cell Epitopes of Human Influenza A Virus Conserved? J Virol 2019; 93:JVI.01534-18. [PMID: 30626684 PMCID: PMC6401462 DOI: 10.1128/jvi.01534-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/26/2018] [Indexed: 01/10/2023] Open
Abstract
Universal influenza vaccines against the conserved epitopes of influenza A virus have been proposed to minimize the burden of seasonal outbreaks and prepare for the pandemics. However, it is not clear how rapidly T cell-inducing vaccines will select for viruses that escape these T cell responses. Our mathematical models explore the factors that contribute to the conservation of CD8 T cell epitopes and how rapidly the virus will evolve in response to T cell-inducing vaccines. We identify the key biological parameters to be measured and questions that need to be addressed in future studies. The high degree of conservation of CD8 T cell epitopes of influenza A virus (IAV) may allow for the development of T cell-inducing vaccines that provide protection across different strains and subtypes. This conservation is not fully explained by functional constraint, since an additional mutation(s) can compensate for the replicative fitness loss of IAV escape variants. Here, we propose three additional mechanisms that contribute to the conservation of CD8 T cell epitopes of IAV. First, influenza-specific CD8 T cells may protect predominantly against severe pathology rather than infection and may have only a modest effect on transmission. Second, polymorphism of the human major histocompatibility complex class I (MHC-I) gene restricts the advantage of an escape variant to only a small fraction of the human population who carry the relevant MHC-I alleles. Finally, infection with CD8 T cell escape variants may result in a compensatory increase in the responses to other epitopes of IAV. We use a combination of population genetics and epidemiological models to examine how the interplay between these mechanisms affects the rate of invasion of IAV escape variants. We conclude that for a wide range of biologically reasonable parameters, the invasion of an escape variant virus will be slow, with a timescale of a decade or more. The results suggest T cell-inducing vaccines do not engender the rapid evolution of IAV. Finally, we identify key parameters whose measurement will allow for more accurate quantification of the long-term effectiveness and impact of universal T cell-inducing influenza vaccines. IMPORTANCE Universal influenza vaccines against the conserved epitopes of influenza A virus have been proposed to minimize the burden of seasonal outbreaks and prepare for the pandemics. However, it is not clear how rapidly T cell-inducing vaccines will select for viruses that escape these T cell responses. Our mathematical models explore the factors that contribute to the conservation of CD8 T cell epitopes and how rapidly the virus will evolve in response to T cell-inducing vaccines. We identify the key biological parameters to be measured and questions that need to be addressed in future studies.
Collapse
|
17
|
Baral S, Raja R, Sen P, Dixit NM. Towards multiscale modeling of the CD8 + T cell response to viral infections. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2019; 11:e1446. [PMID: 30811096 PMCID: PMC6614031 DOI: 10.1002/wsbm.1446] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/22/2022]
Abstract
The CD8+ T cell response is critical to the control of viral infections. Yet, defining the CD8+ T cell response to viral infections quantitatively has been a challenge. Following antigen recognition, which triggers an intracellular signaling cascade, CD8+ T cells can differentiate into effector cells, which proliferate rapidly and destroy infected cells. When the infection is cleared, they leave behind memory cells for quick recall following a second challenge. If the infection persists, the cells may become exhausted, retaining minimal control of the infection while preventing severe immunopathology. These activation, proliferation and differentiation processes as well as the mounting of the effector response are intrinsically multiscale and collective phenomena. Remarkable experimental advances in the recent years, especially at the single cell level, have enabled a quantitative characterization of several underlying processes. Simultaneously, sophisticated mathematical models have begun to be constructed that describe these multiscale phenomena, bringing us closer to a comprehensive description of the CD8+ T cell response to viral infections. Here, we review the advances made and summarize the challenges and opportunities ahead. This article is categorized under: Analytical and Computational Methods > Computational Methods Biological Mechanisms > Cell Fates Biological Mechanisms > Cell Signaling Models of Systems Properties and Processes > Mechanistic Models.
Collapse
Affiliation(s)
- Subhasish Baral
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
| | - Rubesh Raja
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
| | - Pramita Sen
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India
| | - Narendra M Dixit
- Department of Chemical Engineering, Indian Institute of Science, Bangalore, India.,Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
18
|
Yan AWC, Zaloumis SG, Simpson JA, McCaw JM. Sequential infection experiments for quantifying innate and adaptive immunity during influenza infection. PLoS Comput Biol 2019; 15:e1006568. [PMID: 30653522 PMCID: PMC6353225 DOI: 10.1371/journal.pcbi.1006568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/30/2019] [Accepted: 10/16/2018] [Indexed: 12/20/2022] Open
Abstract
Laboratory models are often used to understand the interaction of related pathogens via host immunity. For example, recent experiments where ferrets were exposed to two influenza strains within a short period of time have shown how the effects of cross-immunity vary with the time between exposures and the specific strains used. On the other hand, studies of the workings of different arms of the immune response, and their relative importance, typically use experiments involving a single infection. However, inferring the relative importance of different immune components from this type of data is challenging. Using simulations and mathematical modelling, here we investigate whether the sequential infection experiment design can be used not only to determine immune components contributing to cross-protection, but also to gain insight into the immune response during a single infection. We show that virological data from sequential infection experiments can be used to accurately extract the timing and extent of cross-protection. Moreover, the broad immune components responsible for such cross-protection can be determined. Such data can also be used to infer the timing and strength of some immune components in controlling a primary infection, even in the absence of serological data. By contrast, single infection data cannot be used to reliably recover this information. Hence, sequential infection data enhances our understanding of the mechanisms underlying the control and resolution of infection, and generates new insight into how previous exposure influences the time course of a subsequent infection.
Collapse
Affiliation(s)
- Ada W. C. Yan
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, United Kingdom
| | - Sophie G. Zaloumis
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Julie A. Simpson
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
| | - James M. McCaw
- School of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria, Australia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia
- Modelling and Simulation, Infection and Immunity Theme, Murdoch Childrens Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
| |
Collapse
|
19
|
Melville K, Rodriguez T, Dobrovolny HM. Investigating Different Mechanisms of Action in Combination Therapy for Influenza. Front Pharmacol 2018; 9:1207. [PMID: 30405419 PMCID: PMC6206389 DOI: 10.3389/fphar.2018.01207] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/03/2018] [Indexed: 01/15/2023] Open
Abstract
Combination therapy for influenza can have several benefits, from reducing the emergence of drug resistant virus strains to decreasing the cost of antivirals. However, there are currently only two classes of antivirals approved for use against influenza, limiting the possible combinations that can be considered for treatment. However, new antivirals are being developed that target different parts of the viral replication cycle, and their potential for use in combination therapy should be considered. The role of antiviral mechanism of action in the effectiveness of combination therapy has not yet been systematically investigated to determine whether certain antiviral mechanisms of action pair well in combination. Here, we use a mathematical model of influenza to model combination treatment with antivirals having different mechanisms of action to measure peak viral load, infection duration, and synergy of different drug combinations. We find that antivirals that lower the infection rate and antivirals that increase the duration of the eclipse phase perform poorly in combination with other antivirals.
Collapse
Affiliation(s)
- Kelli Melville
- Physics Department, East Carolina University, Greenville, NC, United States
| | - Thalia Rodriguez
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| | - Hana M. Dobrovolny
- Department of Physics and Astronomy, Texas Christian University, Fort Worth, TX, United States
| |
Collapse
|
20
|
Wu X, Wu P, Shen Y, Jiang X, Xu F. CD8 + Resident Memory T Cells and Viral Infection. Front Immunol 2018; 9:2093. [PMID: 30283442 PMCID: PMC6156262 DOI: 10.3389/fimmu.2018.02093] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 08/24/2018] [Indexed: 12/24/2022] Open
Abstract
Tissue-resident memory T (Trm) cells are a subset of recently identified memory T cells that mainly reside and serve as sentinels in non-lymphoid peripheral tissues. Unlike the well-characterized circulating central memory T (Tcm) cells and effector memory T (Tem) cells, Trm cells persist in the tissues, do not recirculate into blood, and offer immediate protection against pathogens upon reinfection. In this review, we focus on CD8+ Trm cells and briefly introduce their characteristics, development, maintenance, and function during viral infection. We also discuss some unresolved problems, such as how CD8+ Trm cells adapt to the local tissue microenvironment, how Trm cells interact with other immune cells during their development and maintenance, and the mechanisms by which CD8+ Trm cells confer immune protection. We believe that a better understanding of these problems is of great clinical and therapeutic value and may contribute to more effective vaccination and treatments against viral infection.
Collapse
Affiliation(s)
- Xuejie Wu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pin Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifei Shen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Xiaodong Jiang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, United States
| | - Feng Xu
- Department of Infectious Diseases, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
21
|
Abstract
Influenza virus infections are a leading cause of morbidity and mortality worldwide. This is due in part to the continual emergence of new viral variants and to synergistic interactions with other viruses and bacteria. There is a lack of understanding about how host responses work to control the infection and how other pathogens capitalize on the altered immune state. The complexity of multi-pathogen infections makes dissecting contributing mechanisms, which may be non-linear and occur on different time scales, challenging. Fortunately, mathematical models have been able to uncover infection control mechanisms, establish regulatory feedbacks, connect mechanisms across time scales, and determine the processes that dictate different disease outcomes. These models have tested existing hypotheses and generated new hypotheses, some of which have been subsequently tested and validated in the laboratory. They have been particularly a key in studying influenza-bacteria coinfections and will be undoubtedly be useful in examining the interplay between influenza virus and other viruses. Here, I review recent advances in modeling influenza-related infections, the novel biological insight that has been gained through modeling, the importance of model-driven experimental design, and future directions of the field.
Collapse
Affiliation(s)
- Amber M Smith
- University of Tennessee Health Science CenterMemphisTNUSA
| |
Collapse
|
22
|
Zitzmann C, Kaderali L. Mathematical Analysis of Viral Replication Dynamics and Antiviral Treatment Strategies: From Basic Models to Age-Based Multi-Scale Modeling. Front Microbiol 2018; 9:1546. [PMID: 30050523 PMCID: PMC6050366 DOI: 10.3389/fmicb.2018.01546] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 06/21/2018] [Indexed: 12/14/2022] Open
Abstract
Viral infectious diseases are a global health concern, as is evident by recent outbreaks of the middle east respiratory syndrome, Ebola virus disease, and re-emerging zika, dengue, and chikungunya fevers. Viral epidemics are a socio-economic burden that causes short- and long-term costs for disease diagnosis and treatment as well as a loss in productivity by absenteeism. These outbreaks and their socio-economic costs underline the necessity for a precise analysis of virus-host interactions, which would help to understand disease mechanisms and to develop therapeutic interventions. The combination of quantitative measurements and dynamic mathematical modeling has increased our understanding of the within-host infection dynamics and has led to important insights into viral pathogenesis, transmission, and disease progression. Furthermore, virus-host models helped to identify drug targets, to predict the treatment duration to achieve cure, and to reduce treatment costs. In this article, we review important achievements made by mathematical modeling of viral kinetics on the extracellular, intracellular, and multi-scale level for Human Immunodeficiency Virus, Hepatitis C Virus, Influenza A Virus, Ebola Virus, Dengue Virus, and Zika Virus. Herein, we focus on basic mathematical models on the population scale (so-called target cell-limited models), detailed models regarding the most important steps in the viral life cycle, and the combination of both. For this purpose, we review how mathematical modeling of viral dynamics helped to understand the virus-host interactions and disease progression or clearance. Additionally, we review different types and effects of therapeutic strategies and how mathematical modeling has been used to predict new treatment regimens.
Collapse
Affiliation(s)
- Carolin Zitzmann
- Institute of Bioinformatics and Center for Functional Genomics of Microbes, University Medicine Greifswald, Greifswald, Germany
| | - Lars Kaderali
- Institute of Bioinformatics and Center for Functional Genomics of Microbes, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
23
|
Muruganandah V, Sathkumara HD, Navarro S, Kupz A. A Systematic Review: The Role of Resident Memory T Cells in Infectious Diseases and Their Relevance for Vaccine Development. Front Immunol 2018; 9:1574. [PMID: 30038624 PMCID: PMC6046459 DOI: 10.3389/fimmu.2018.01574] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022] Open
Abstract
Background Resident memory T cells have emerged as key players in the immune response generated against a number of pathogens. Their ability to take residence in non-lymphoid peripheral tissues allows for the rapid deployment of secondary effector responses at the site of pathogen entry. This ability to provide enhanced regional immunity has gathered much attention, with the generation of resident memory T cells being the goal of many novel vaccines. Objectives This review aimed to systematically analyze published literature investigating the role of resident memory T cells in human infectious diseases. Known effector responses mounted by these cells are summarized and key strategies that are potentially influential in the rational design of resident memory T cell inducing vaccines have also been highlighted. Methods A Boolean search was applied to Medline, SCOPUS, and Web of Science. Studies that investigated the effector response generated by resident memory T cells and/or evaluated strategies for inducing these cells were included irrespective of published date. Studies must have utilized an established technique for identifying resident memory T cells such as T cell phenotyping. Results While over 600 publications were revealed by the search, 147 articles were eligible for inclusion. The reference lists of included articles were also screened for other eligible publications. This resulted in the inclusion of publications that studied resident memory T cells in the context of over 25 human pathogens. The vast majority of studies were conducted in mouse models and demonstrated that resident memory T cells mount protective immune responses. Conclusion Although the role resident memory T cells play in providing immunity varies depending on the pathogen and anatomical location they resided in, the evidence overall suggests that these cells are vital for the timely and optimal protection against a number of infectious diseases. The induction of resident memory T cells should be further investigated and seriously considered when designing new vaccines.
Collapse
Affiliation(s)
- Visai Muruganandah
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Harindra D Sathkumara
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| | - Severine Navarro
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia.,QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Andreas Kupz
- Centre for Biosecurity and Tropical Infectious Diseases, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
24
|
Zarnitsyna VI, Bulusheva I, Handel A, Longini IM, Halloran ME, Antia R. Intermediate levels of vaccination coverage may minimize seasonal influenza outbreaks. PLoS One 2018; 13:e0199674. [PMID: 29944709 PMCID: PMC6019388 DOI: 10.1371/journal.pone.0199674] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/12/2018] [Indexed: 11/30/2022] Open
Abstract
For most pathogens, vaccination reduces the spread of the infection and total number of cases; thus, public policy usually advocates maximizing vaccination coverage. We use simple mathematical models to explore how this may be different for pathogens, such as influenza, which exhibit strain variation. Our models predict that the total number of seasonal influenza infections is minimized at an intermediate (rather than maximal) level of vaccination, and, somewhat counter-intuitively, further increasing the level of the vaccination coverage may lead to higher number of influenza infections and be detrimental to the public interest. This arises due to the combined effects of: competition between multiple co-circulating strains; limited breadth of protection afforded by the vaccine; and short-term strain-transcending immunity following natural infection. The study highlights the need for better quantification of the components of vaccine efficacy and longevity of strain-transcending cross-immunity in order to generate nuanced recommendations for influenza vaccine coverage levels.
Collapse
Affiliation(s)
- Veronika I. Zarnitsyna
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30322, United States of America
- * E-mail: (VZ); (RA)
| | - Irina Bulusheva
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, 141701, Russia
| | - Andreas Handel
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, GA, 30602, United States of America
| | - Ira M. Longini
- Department of Biostatistics, University of Florida, Gainesville, FL, 32611, United States of America
| | - M. Elizabeth Halloran
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, 98109, United States of America
- Department of Biostatistics, University of Washington, Seattle, WA, United States of America
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA, 30322, United States of America
- * E-mail: (VZ); (RA)
| |
Collapse
|
25
|
Goronzy JJ, Weyand CM. Successful and Maladaptive T Cell Aging. Immunity 2017; 46:364-378. [PMID: 28329703 DOI: 10.1016/j.immuni.2017.03.010] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 02/27/2017] [Accepted: 03/07/2017] [Indexed: 12/21/2022]
Abstract
Throughout life, the T cell system adapts to shifting resources and demands, resulting in a fundamentally restructured immune system in older individuals. Here we review the cellular and molecular features of an aged immune system and discuss the trade-offs inherent to these adaptive mechanisms. Processes include homeostatic proliferation that maintains compartment size at the expense of partial loss in stemness and incomplete differentiation and the activation of negative regulatory programs, which constrain effector T cell expansion and prevent increasing oligoclonality but also interfere with memory cell generation. We propose that immune failure occurs when adaptive strategies developed by the aging T cell system fail and also discuss how, in some settings, the programs associated with T cell aging culminates in a maladaptive response that directly contributes to chronic inflammatory disease.
Collapse
Affiliation(s)
- Jörg J Goronzy
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304, USA.
| | - Cornelia M Weyand
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Palo Alto Veterans Administration Health Care System, Palo Alto, CA 94304, USA.
| |
Collapse
|
26
|
The Mechanisms for Within-Host Influenza Virus Control Affect Model-Based Assessment and Prediction of Antiviral Treatment. Viruses 2017; 9:v9080197. [PMID: 28933757 PMCID: PMC5580454 DOI: 10.3390/v9080197] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 12/28/2022] Open
Abstract
Models of within-host influenza viral dynamics have contributed to an improved understanding of viral dynamics and antiviral effects over the past decade. Existing models can be classified into two broad types based on the mechanism of viral control: models utilising target cell depletion to limit the progress of infection and models which rely on timely activation of innate and adaptive immune responses to control the infection. In this paper, we compare how two exemplar models based on these different mechanisms behave and investigate how the mechanistic difference affects the assessment and prediction of antiviral treatment. We find that the assumed mechanism for viral control strongly influences the predicted outcomes of treatment. Furthermore, we observe that for the target cell-limited model the assumed drug efficacy strongly influences the predicted treatment outcomes. The area under the viral load curve is identified as the most reliable predictor of drug efficacy, and is robust to model selection. Moreover, with support from previous clinical studies, we suggest that the target cell-limited model is more suitable for modelling in vitro assays or infection in some immunocompromised/immunosuppressed patients while the immune response model is preferred for predicting the infection/antiviral effect in immunocompetent animals/patients.
Collapse
|
27
|
Cao P, Wang Z, Yan AWC, McVernon J, Xu J, Heffernan JM, Kedzierska K, McCaw JM. On the Role of CD8 + T Cells in Determining Recovery Time from Influenza Virus Infection. Front Immunol 2016; 7:611. [PMID: 28066421 PMCID: PMC5167728 DOI: 10.3389/fimmu.2016.00611] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 12/02/2016] [Indexed: 01/02/2023] Open
Abstract
Myriad experiments have identified an important role for CD8+ T cell response mechanisms in determining recovery from influenza A virus infection. Animal models of influenza infection further implicate multiple elements of the immune response in defining the dynamical characteristics of viral infection. To date, influenza virus models, while capturing particular aspects of the natural infection history, have been unable to reproduce the full gamut of observed viral kinetic behavior in a single coherent framework. Here, we introduce a mathematical model of influenza viral dynamics incorporating innate, humoral, and cellular immune components and explore its properties with a particular emphasis on the role of cellular immunity. Calibrated against a range of murine data, our model is capable of recapitulating observed viral kinetics from a multitude of experiments. Importantly, the model predicts a robust exponential relationship between the level of effector CD8+ T cells and recovery time, whereby recovery time rapidly decreases to a fixed minimum recovery time with an increasing level of effector CD8+ T cells. We find support for this relationship in recent clinical data from influenza A (H7N9) hospitalized patients. The exponential relationship implies that people with a lower level of naive CD8+ T cells may receive significantly more benefit from induction of additional effector CD8+ T cells arising from immunological memory, itself established through either previous viral infection or T cell-based vaccines.
Collapse
Affiliation(s)
- Pengxing Cao
- School of Mathematics and Statistics, The University of Melbourne , Melbourne, VIC , Australia
| | - Zhongfang Wang
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, VIC, Australia; Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ada W C Yan
- School of Mathematics and Statistics, The University of Melbourne , Melbourne, VIC , Australia
| | - Jodie McVernon
- Doherty Epidemiology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital, Melbourne, VIC, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia; Modelling and Simulation, Infection and Immunity Theme, Murdoch Childrens Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Key Laboratory of Medical Molecular Virology of Ministry of Education/Health, Shanghai Medical College, Institutes of Biomedical Sciences, Fudan University , Shanghai , China
| | - Jane M Heffernan
- Modelling Infection and Immunity Lab, Centre for Disease Modelling, York Institute for Health Research, Mathematics and Statistics, York University , Toronto, ON , Canada
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of Melbourne and Royal Melbourne Hospital , Melbourne, VIC , Australia
| | - James M McCaw
- School of Mathematics and Statistics, The University of Melbourne, Melbourne, VIC, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia; Modelling and Simulation, Infection and Immunity Theme, Murdoch Childrens Research Institute, The Royal Children's Hospital, Melbourne, VIC, Australia
| |
Collapse
|
28
|
Yan AWC, Cao P, Heffernan JM, McVernon J, Quinn KM, La Gruta NL, Laurie KL, McCaw JM. Modelling cross-reactivity and memory in the cellular adaptive immune response to influenza infection in the host. J Theor Biol 2016; 413:34-49. [PMID: 27856216 DOI: 10.1016/j.jtbi.2016.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 11/02/2016] [Accepted: 11/05/2016] [Indexed: 01/05/2023]
Abstract
The cellular adaptive immune response plays a key role in resolving influenza infection. Experiments where individuals are successively infected with different strains within a short timeframe provide insight into the underlying viral dynamics and the role of a cross-reactive immune response in resolving an acute infection. We construct a mathematical model of within-host influenza viral dynamics including three possible factors which determine the strength of the cross-reactive cellular adaptive immune response: the initial naive T cell number, the avidity of the interaction between T cells and the epitopes presented by infected cells, and the epitope abundance per infected cell. Our model explains the experimentally observed shortening of a second infection when cross-reactivity is present, and shows that memory in the cellular adaptive immune response is necessary to protect against a second infection.
Collapse
Affiliation(s)
- Ada W C Yan
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Pengxing Cao
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jane M Heffernan
- Department of Mathematics and Statistics, York University, Toronto, Ontario, Canada M3J 1P3; Modelling Infection and Immunity Lab, Centre for Disease Modelling, York Institute for Health Research, York University, Toronto, Ontario, Canada M3J 1P3
| | - Jodie McVernon
- Doherty Epidemiology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC 3010, Australia; Modelling and Simulation, Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia
| | - Kylie M Quinn
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Nicole L La Gruta
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia; Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Karen L Laurie
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; School of Applied and Biomedical Sciences, Federation University, Churchill, VIC 3842, Australia; Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3010, Australia
| | - James M McCaw
- School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, VIC 3010, Australia; Modelling and Simulation, Infection and Immunity Theme, Murdoch Children's Research Institute, Parkville, VIC 3052, Australia.
| |
Collapse
|