Parameter Identification for a Model of Neonatal Fc Receptor-Mediated Recycling of Endogenous Immunoglobulin G in Humans

Salvage of endogenous immunoglobulin G (IgG) by the neonatal Fc receptor (FcRn) is implicated in many clinical areas, including therapeutic monoclonal antibody kinetics, patient monitoring in IgG multiple myeloma, and antibody-mediated transplant rejection. There is a clear clinical need for a fully parameterized model of FcRn-mediated recycling of endogenous IgG to allow for predictive modeling, with the potential for optimizing therapeutic regimens for better patient outcomes. In this paper we study a mechanism-based model incorporating nonlinear FcRn-IgG binding kinetics. The aim of this study is to determine whether parameter values can be estimated using the limited in vivo human data, available in the literature, from studies of the kinetics of radiolabeled IgG in humans. We derive mathematical descriptions of the experimental observations—timecourse data and fractional catabolic rate (FCR) data—based on the underlying physiological model. Structural identifiability analyses are performed to determine which, if any, of the parameters are unique with respect to the observations. Structurally identifiable parameters are then estimated from the data. It is found that parameter values estimated from timecourse data are not robust, suggesting that the model complexity is not supported by the available data. Based upon the structural identifiability analyses, a new expression for the FCR is derived. This expression is fitted to the FCR data to estimate unknown parameter values. Using these parameter estimates, the plasma IgG response is simulated under clinical conditions. Finally a suggestion is made for a reduced-order model based upon the newly derived expression for the FCR. The reduced-order model is used to predict the plasma IgG response, which is compared with the original four-compartment model, showing good agreement. This paper shows how techniques for compartmental model analysis—structural identifiability analysis, linearization, and reparameterization—can be used to ensure robust parameter identification.

[1]  T. Waldmann,et al.  Clinical and experimental metabolism of normal 6.6s gamma-globulin in normal subjects and in patients with macroglobulinemia and multiple myeloma. , 1963, The Journal of laboratory and clinical medicine.

[2]  W Strober,et al.  Metabolism of immunoglobulins. , 1969, Progress in allergy.

[3]  R. Bellman,et al.  On structural identifiability , 1970 .

[4]  S. Shaughnessy,et al.  Do No Harm: Health Systems’ Duty to Promote Clinician Well-Being , 2022, American Journal of Hospital Medicine.

[5]  Monoclonal anti-cea antibody , 1982 .

[6]  David H. Anderson Compartmental Modeling and Tracer Kinetics , 1983 .

[7]  T. Waldmann,et al.  Familial hypercatabolic hypoproteinemia. A disorder of endogenous catabolism of albumin and immunoglobulin. , 1990, The Journal of clinical investigation.

[8]  S. Huang,et al.  The effects of measurement errors in the plasma radioactivity curve on parameter estimation in positron emission tomography. , 1991, Physics in medicine and biology.

[9]  C. Anderson,et al.  The protection receptor for IgG catabolism is the beta2-microglobulin-containing neonatal intestinal transport receptor. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[10]  Rainer Storn,et al.  Differential Evolution – A Simple and Efficient Heuristic for global Optimization over Continuous Spaces , 1997, J. Glob. Optim..

[11]  Claudio Cobelli,et al.  Tracer Kinetics In Biomedical Research , 2000 .

[12]  R. Hansen,et al.  Pharmacokinetic/pharmacodynamic modeling of the effects of intravenous immunoglobulin on the disposition of antiplatelet antibodies in a rat model of immune thrombocytopenia. , 2003, Journal of pharmaceutical sciences.

[13]  Claudio Cobelli,et al.  Generalized Sensitivity Functions in Physiological System Identification , 1999, Annals of Biomedical Engineering.

[14]  Anna M. Wu,et al.  A Predictive Model of Therapeutic Monoclonal Antibody Dynamics and Regulation by the Neonatal Fc Receptor (FcRn) , 2005, Annals of Biomedical Engineering.

[15]  Jonghan Kim,et al.  Kinetics of FcRn-mediated recycling of IgG and albumin in human: pathophysiology and therapeutic implications using a simplified mechanism-based model. , 2007, Clinical immunology.

[16]  Joseph P. Balthasar,et al.  Physiologically-based pharmacokinetic (PBPK) model to predict IgG tissue kinetics in wild-type and FcRn-knockout mice , 2007, Journal of Pharmacokinetics and Pharmacodynamics.

[17]  W Wang,et al.  Monoclonal Antibody Pharmacokinetics and Pharmacodynamics , 2008, Clinical pharmacology and therapeutics.

[18]  Lanyan Fang,et al.  Predictive Physiologically Based Pharmacokinetic Model for Antibody-Directed Enzyme Prodrug Therapy , 2008, Drug Metabolism and Disposition.

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

[20]  John Hattersley,et al.  Mathematical modelling of immune condition dynamics : a clinical perspective , 2009 .

[21]  Joseph P Balthasar,et al.  Physiologically based pharmacokinetic model for T84.66: a monoclonal anti-CEA antibody. , 2010, Journal of pharmaceutical sciences.

[22]  E. J. Routh A Treatise on the Stability of a Given State of Motion: Particularly Steady Motion , 2010 .

[23]  Dhaval K. Shah,et al.  Towards a platform PBPK model to characterize the plasma and tissue disposition of monoclonal antibodies in preclinical species and human , 2011, Journal of Pharmacokinetics and Pharmacodynamics.

[24]  W. Krzyzanski,et al.  Methods of solving rapid binding target-mediated drug disposition model for two drugs competing for the same receptor , 2012, Journal of Pharmacokinetics and Pharmacodynamics.

[25]  F. Theil,et al.  Subcutaneous bioavailability of therapeutic antibodies as a function 
of FcRn binding affinity in mice , 2012, mAbs.

[26]  Jim J. Xiao Pharmacokinetic Models for FcRn-Mediated IgG Disposition , 2012, Journal of biomedicine & biotechnology.

[27]  Evaluation of a Catenary PBPK Model for Predicting the In Vivo Disposition of mAbs Engineered for High-Affinity Binding to FcRn , 2012, The AAPS Journal.

[28]  Neil D. Evans,et al.  Describing the effectiveness of immunosuppression drugs and apheresis in the treatment of transplant patients , 2013, Comput. Methods Programs Biomed..

[29]  S. Iyer,et al.  Modeling approach to investigate the effect of neonatal Fc receptor binding affinity and anti-therapeutic antibody on the pharmacokinetic of humanized monoclonal anti-tumor necrosis factor-α IgG antibody in cynomolgus monkey. , 2014, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

[30]  W. Huisinga,et al.  Monoclonal antibody disposition: a simplified PBPK model and its implications for the derivation and interpretation of classical compartment models , 2014, Journal of Pharmacokinetics and Pharmacodynamics.

[31]  Masoud Jamei,et al.  Simulation of Monoclonal Antibody Pharmacokinetics in HumansUsing a Minimal Physiologically Based Model , 2014, The AAPS Journal.

[32]  Subhojit Ghosh A differential evolution based approach for estimating minimal model parameters from IVGTT data , 2014, Comput. Biol. Medicine.

[33]  H. Goldschmidt,et al.  International Myeloma Working Group consensus criteria for response and minimal residual disease assessment in multiple myeloma. , 2016, The Lancet. Oncology.

[34]  F. Kappel,et al.  Generalized sensitivity functions for multiple output systems , 2016 .

[35]  A. Dispenzieri,et al.  High sensitivity blood-based M-protein detection in sCR patients with multiple myeloma , 2017, Blood Cancer Journal.

[36]  M. Chappell,et al.  Analysis of a Compartmental Model of Endogenous Immunoglobulin G Metabolism with Application to Multiple Myeloma , 2017, Front. Physiol..

[37]  S. Lonial,et al.  Influence of Disease and Patient Characteristics on Daratumumab Exposure and Clinical Outcomes in Relapsed or Refractory Multiple Myeloma , 2017, Clinical Pharmacokinetics.

[38]  J. Lancet,et al.  ASXL1 frameshift mutations drive inferior outcomes in CMML without negative impact in MDS , 2016, Blood Cancer Journal.