Quantifying innate and adaptive immunity during influenza infection using sequential infection experiments and mathematical models

Recent experiments where ferrets were exposed to two influenza strains within a short time have provided insight into the timing and contribution of each immune component to cross-protection. While mathematical models which capture the novel observations have been developed and analysed, they have many parameters and it is unclear whether the contributions of each immune component to cross-protection can be recovered from the data. We show using Markov chain Monte Carlo methods that within a simulation estimation framework, a model fitted to sequential infection data accurately captures the timing and extent of cross-protection, and attributes such cross-protection to the correct broad immune components. In addition, sequential infection data enables recovery of the timing and role of each immune component in controlling a primary infection. A major limitation of previous studies using single infection data is that they have produced discrepant estimates of these quantities. This evaluation demonstrates that sequential infection experiments provide richer information for quantifying the contribution of each immune component to cross-protection. This information 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.

[1]  Pengxing Cao,et al.  On the extinction probability in models of within-host infection: the role of latency and immunity , 2016, Journal of mathematical biology.

[2]  Udo Reichl,et al.  Mathematical model of influenza A virus production in large-scale microcarrier culture. , 2005, Biotechnology and bioengineering.

[3]  Guy Boivin,et al.  The H275Y Neuraminidase Mutation of the Pandemic A/H1N1 Influenza Virus Lengthens the Eclipse Phase and Reduces Viral Output of Infected Cells, Potentially Compromising Fitness in Ferrets , 2012, Journal of Virology.

[4]  A. Handel,et al.  Mathematical analysis of a mouse experiment suggests little role for resource depletion in controlling influenza infection within host , 2017, 1705.02565.

[5]  Donald Geman,et al.  Stochastic relaxation, Gibbs distributions, and the Bayesian restoration of images , 1984 .

[6]  Andreas Handel,et al.  Towards a quantitative understanding of the within-host dynamics of influenza A infections , 2010, Journal of The Royal Society Interface.

[7]  D. Wodarz,et al.  Does programmed CTL proliferation optimize virus control? , 2005, Trends in immunology.

[8]  Mats O. Karlsson,et al.  Likelihood based approaches to handling data below the quantification limit using NONMEM VI , 2008, Journal of Pharmacokinetics and Pharmacodynamics.

[9]  M. Plummer,et al.  CODA: convergence diagnosis and output analysis for MCMC , 2006 .

[10]  David B Chang,et al.  Simple scaling laws for influenza A rise time, duration, and severity. , 2007, Journal of theoretical biology.

[11]  D. Nayak,et al.  Defective-interfering (DI) RNAs of influenza viruses: origin, structure, expression, and interference. , 1985, Current topics in microbiology and immunology.

[12]  H. A. M. Wols Plasma Cells , 2005 .

[13]  Susan M. Kaech,et al.  Molecular and Functional Profiling of Memory CD8 T Cell Differentiation , 2002, Cell.

[14]  J. A. Misplon,et al.  Heterosubtypic immunity to influenza A virus in mice lacking IgA, all Ig, NKT cells, or gamma delta T cells. , 2001, Journal of immunology.

[15]  A. Perelson,et al.  Kinetics of Influenza A Virus Infection in Humans , 2006, Journal of Virology.

[16]  N. Dimmock,et al.  Defective interfering viruses and their potential as antiviral agents , 2010, Reviews in medical virology.

[17]  James M. McCaw,et al.  Interval Between Infections and Viral Hierarchy Are Determinants of Viral Interference Following Influenza Virus Infection in a Ferret Model , 2015, The Journal of infectious diseases.

[18]  J. Bloom,et al.  Estimating the Fitness Advantage Conferred by Permissive Neuraminidase Mutations in Recent Oseltamivir-Resistant A(H1N1)pdm09 Influenza Viruses , 2014, PLoS pathogens.

[19]  D. Rubin,et al.  Inference from Iterative Simulation Using Multiple Sequences , 1992 .

[20]  C. Beauchemin,et al.  Impact of the H275Y and I223V Mutations in the Neuraminidase of the 2009 Pandemic Influenza Virus In Vitro and Evaluating Experimental Reproducibility , 2015, PloS one.

[21]  L. Wahl,et al.  Perspectives on the basic reproductive ratio , 2005, Journal of The Royal Society Interface.

[22]  K. Toellner,et al.  Intrinsic Constraint on Plasmablast Growth and Extrinsic Limits of Plasma Cell Survival , 2000, The Journal of experimental medicine.

[23]  Andreas Handel,et al.  Neuraminidase Inhibitor Resistance in Influenza: Assessing the Danger of Its Generation and Spread , 2007, PLoS Comput. Biol..

[24]  Peter A. J. Hilbers,et al.  A Bayesian approach to targeted experiment design , 2012, Bioinform..

[25]  Dominik Wodarz,et al.  Direct quantitation of rapid elimination of viral antigen‐positive lymphocytes by antiviral CD8+ T cells in vivo , 2000, European journal of immunology.

[26]  J. McVernon,et al.  On the Role of CD8+ T Cells in Determining Recovery Time from Influenza Virus Infection , 2016, Front. Immunol..

[27]  J. D. de Pablo,et al.  Optimal allocation of replicas in parallel tempering simulations. , 2005, The Journal of chemical physics.

[28]  Stephen P. Schoenberger,et al.  Naïve CTLs require a single brief period of antigenic stimulation for clonal expansion and differentiation , 2001, Nature Immunology.

[29]  Alan S. Perelson,et al.  Modeling Within-Host Dynamics of Influenza Virus Infection Including Immune Responses , 2012, PLoS Comput. Biol..

[30]  Alan S. Perelson,et al.  Quantifying the Early Immune Response and Adaptive Immune Response Kinetics in Mice Infected with Influenza A Virus , 2010, Journal of Virology.

[31]  Ann Cullinane,et al.  Dynamics of Influenza Virus Infection and Pathology , 2010, Journal of Virology.

[32]  J. McVernon,et al.  Quantifying relative within-host replication fitness in influenza virus competition experiments. , 2015, Journal of theoretical biology.

[33]  Evan C. Haskell,et al.  The logistic growth model as an approximating model for viral load measurements of influenza A virus , 2017, Math. Comput. Simul..

[34]  R. C. Layton,et al.  Higher Level of Replication Efficiency of 2009 (H1N1) Pandemic Influenza Virus than Those of Seasonal and Avian Strains: Kinetics from Epithelial Cell Culture and Computational Modeling , 2010, Journal of Virology.

[35]  K. Yap,et al.  Cytotoxic T Cells in the Lungs of Mice Infected with an Influenza A Virus , 1978, Scandinavian journal of immunology.

[36]  G. Bocharov,et al.  Mathematical model of antiviral immune response. III. Influenza A virus infection. , 1994, Journal of theoretical biology.

[37]  Andrew J. McMichael,et al.  Clonal Selection, Clonal Senescence, and Clonal Succession: The Evolution of the T Cell Response to Infection with a Persistent Virus1 , 2002, The Journal of Immunology.

[38]  A. Hindmarsh,et al.  CVODE, a stiff/nonstiff ODE solver in C , 1996 .

[39]  G. Marchuk,et al.  Mathematical model of antiviral immune response. I. Data analysis, generalized picture construction and parameters evaluation for hepatitis B. , 1991, Journal of theoretical biology.

[40]  Kylie M. Quinn,et al.  Modelling cross-reactivity and memory in the cellular adaptive immune response to influenza infection in the host. , 2016, Journal of theoretical biology.

[41]  D. Allman,et al.  What Is and What Should Always Have Been: Long-Lived Plasma Cells Induced by T Cell–Independent Antigens , 2013, The Journal of Immunology.

[42]  Andreas Handel,et al.  Modeling inoculum dose dependent patterns of acute virus infections. , 2014, Journal of theoretical biology.

[43]  D. Kofke,et al.  Selection of temperature intervals for parallel-tempering simulations. , 2005, The Journal of chemical physics.

[44]  T. Iwasaki,et al.  Defense mechanisms against primary influenza virus infection in mice. I. The roles of interferon and neutralizing antibodies and thymus dependence of interferon and antibody production. , 1977, Journal of immunology.

[45]  Cross-reactive human B cell and T cell epitopes between influenza A and B viruses , 2013, Virology Journal.

[46]  David B. Dunson,et al.  Bayesian Data Analysis , 2010 .

[47]  J. Altman,et al.  Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. , 1998, Immunity.

[48]  Stephanie Forrest,et al.  A stochastic model of cytotoxic T cell responses. , 2004, Journal of theoretical biology.

[49]  Alan S. Perelson,et al.  Recruitment Times, Proliferation, and Apoptosis Rates during the CD8+ T-Cell Response to Lymphocytic Choriomeningitis Virus , 2001, Journal of Virology.

[50]  M. Keeling,et al.  An in-host model of acute infection: measles as a case study. , 2008, Theoretical population biology.

[51]  James M. McCaw,et al.  Reducing Uncertainty in Within-Host Parameter Estimates of Influenza Infection by Measuring Both Infectious and Total Viral Load , 2013, PloS one.

[52]  Henrique Veiga-Fernandes,et al.  Response of naïve and memory CD8+ T cells to antigen stimulation in vivo , 2000, Nature Immunology.

[53]  Ursula Klingmüller,et al.  Structural and practical identifiability analysis of partially observed dynamical models by exploiting the profile likelihood , 2009, Bioinform..

[54]  Michael W Deem,et al.  Parallel tempering: theory, applications, and new perspectives. , 2005, Physical chemistry chemical physics : PCCP.

[55]  J. Yewdell,et al.  Heterosubtypic immunity to influenza A virus: where do we stand? , 2008, Microbes and infection.

[56]  A. Perelson,et al.  Influenza A virus infection kinetics: quantitative data and models , 2011, Wiley interdisciplinary reviews. Systems biology and medicine.

[57]  M. Simon,et al.  Mechanism of recovery from acute virus infection , 1985, Medical Microbiology and Immunology.

[58]  R. Ramphal,et al.  Passive serum antibody causes temporary recovery from influenza virus infection of the nose, trachea and lung of nude mice. , 1988, Immunology.

[59]  Susan M. Kaech,et al.  Memory CD8+ T cell differentiation: initial antigen encounter triggers a developmental program in naïve cells , 2001, Nature Immunology.

[60]  L M Wahl,et al.  Viral dynamics of primary viremia and antiretroviral therapy in simian immunodeficiency virus infection , 1997, Journal of virology.

[61]  N. Metropolis,et al.  Equation of State Calculations by Fast Computing Machines , 1953, Resonance.

[62]  Joshua M. Epstein,et al.  Controlling Pandemic Flu: The Value of International Air Travel Restrictions , 2007, PloS one.

[63]  Ruy M Ribeiro,et al.  Modeling amantadine treatment of influenza A virus in vitro. , 2008, Journal of theoretical biology.

[64]  C. Geyer Markov Chain Monte Carlo Maximum Likelihood , 1991 .

[65]  James M. McCaw,et al.  Innate Immunity and the Inter-exposure Interval Determine the Dynamics of Secondary Influenza Virus Infection and Explain Observed Viral Hierarchies , 2015, PLoS Comput. Biol..

[66]  A. Perelson,et al.  Simulation and Prediction of the Adaptive Immune Response to Influenza A Virus Infection , 2009, Journal of Virology.

[67]  R. Webster,et al.  Lethal H5N1 influenza viruses escape host anti-viral cytokine responses , 2002, Nature Medicine.

[68]  Hana M. Dobrovolny,et al.  Assessing Mathematical Models of Influenza Infections Using Features of the Immune Response , 2013, PloS one.

[69]  Gilles Clermont,et al.  A Dynamical Model of Human Immune Response to Influenza a Virus Infection , 2006 .