Elsevier

Journal of Theoretical Biology

Volume 458, 7 December 2018, Pages 31-46
Journal of Theoretical Biology

Methods for determining key components in a mathematical model for tumor–immune dynamics in multiple myeloma

https://doi.org/10.1016/j.jtbi.2018.08.037Get rights and content

Highlights

  • We explore a mathematical model of multiple myeloma that was presented previously.

  • We justify model parameter ranges and values using published data and calculations.

  • We use global methods to determine parameters the model is most sensitive to.

  • We determine which of those parameters could be estimated from data.

  • We numerically explore the behavior of the model over time.

Abstract

In this work, we analyze a mathematical model we introduced previously for the dynamics of multiple myeloma and the immune system. We focus on four main aspects: (1) obtaining and justifying ranges and values for all parameters in the model; (2) determining a subset of parameters to which the model is most sensitive; (3) determining which parameters in this subset can be uniquely estimated given certain types of data; and (4) exploring the model numerically. Using global sensitivity analysis techniques, we found that the model is most sensitive to certain growth, loss, and efficacy parameters. This analysis provides the foundation for a future application of the model: prediction of optimal combination regimens in patients with multiple myeloma.

Introduction

Multiple myeloma (MM) is a cancer of plasma B cells. Although there are almost two dozen treatments approved in the US and others currently in clinical trials, many patients do not survive more than ten years (Kazandjian and Landgren, 2016). Regimens usually combine multiple therapies (two, three, or more drugs) simultaneously. Patients receive additional combination therapies if they do not respond to the initial combination, or when they relapse. Without direct comparison studies, it is difficult to know the best combinations and doses for treatment, and questions remain regarding both treatment choice and timing (Kazandjian and Landgren, 2016). A mathematical model that accurately captures important features of the disease and therapy dynamics can be used to test regimens in silico. It can also be used to calculate a regimen that is predicted to perform optimally.

A number of mathematical models for the progression of MM and its response to treatments have been developed previously. Most of these models have made use of the correlation between MM tumor burden and a protein that is shed by MM cells, M protein, that can be measured in the peripheral blood (Durie, Salmon, 1975, Salmon, Smith, 1970). Sullivan and Salmon (1972) published a simple mathematical model to characterize chemotherapy-induced tumor regression in patients with MM. Swan and Vincent applied optimal control in Swan and Vincent (1977) to predict an optimal chemotherapy dosing strategy for patients with MM. Hokanson et al. (1977) fit individual M-protein data with mathematical models of myeloma cell populations that were sensitive or resistant to chemotherapy. More recently, several groups have examined the disease progression in the presence of various treatments. Jonsson et al. (2015) published a model of patient M-protein levels in response to treatment with carfilzomib. Tang et al. (2016) published a model for patient M-protein levels in clinical trials of bortezomib-based chemotherapy. They predicted that rational combination treatments with decreased selection pressure on myeloma cells could lead to a longer remission period. Nanavati et al. (2017) published a semi-mechanistic protein production and signaling model with seven main compartments. The model was fit to aggregate in vivo xenograft data (digitized from the literature) for mice treated with vorinostat.

These prior studies have not directly examined the role of the immune system in MM disease dynamics, though there is a long history of tumor–immune dynamics models (cf. d’Onofrio, 2005, Kirschner, Panetta, 1998, Kuznetsov, Makalkin, Taylor, Perelson, 1994, Moore, Li, 2004, de Pillis, Radunskaya, Wiseman, 2005, Stepanova, 1980, de Vladar, González, 2004). One contribution of our work is the incorporation and careful parameterization of immune-disease interactions in a mathematical model for MM. This is important as several immunomodulatory drugs have been approved for use in patients with MM, and more are currently in clinical trials (Kazandjian and Landgren, 2016). A well-justified model of tumor–immune dynamics would support in silico exploration of regimens that include immunomodulatory therapies.

In Gallaher et al. (2018), we introduced a semi-mechanistic mathematical model of MM (tumor) and immune system dynamics in a hypothetical patient. We focused on the structure of the model, and how best to incorporate tumor–immune dynamics. The mathematical model consists of a dynamical system that tracks tumor and immune components in the peripheral blood of patients with MM. Although MM is a disease based in the bone marrow, myeloma cells overproduce a myeloma protein or M protein, which can circulate outside the bone marrow. Our model uses the level of M protein in the peripheral blood as a surrogate of tumor burden in patients with MM (Durie, Salmon, 1975, Salmon, Smith, 1970). The immune cells included in our model play important roles in disease control or progression, and can also be measured in the peripheral blood. In addition to justifying the structure of the full model, this work also considered a reduced version of the model that still captures key long-term dynamics, and analyzed equilibria and stability of this reduced model.

In this paper, we present methods to determine important components or interactions in the model. This information can be used to help decide which therapies to consider in combination regimens. We began with extensive literature searches to determine and justify parameter values and ranges to use in the model. For the ranges considered feasible for the parameters, we used sampling to perform global sensitivity analyses. We found eight parameters that have the largest effect on long-term levels of M protein. We refer to this subset as “sensitive parameters”. We numerically explored how small changes in the sensitive parameters can lead to qualitatively different types of model behavior. In particular, changes in values of the sensitive parameters can result in a switch between high and low tumor burden states.

With all except the sensitive parameters fixed, we used identifiability analysis to determine that the model is globally structurally identifiable. In other words, the eight sensitive parameters could be uniquely estimated if we had continuous data from all four populations in our model. Although we did not find time-series data in the literature, we did find steady-state patient values for the four tumor and immune populations included in our model (Greipp, Miguel, Durie, Crowley, Barlogie, Bladé, Boccadoro, Child, Avet-Loiseau, Kyle, Lahuerta, Ludwig, Morgan, Powles, Shimizu, Shustik, Sonneveld, Tosi, Turesson, Westin, 2005, Pessoa de Magalhães, Vidriales, Paiva, Fernandez-Gimenez, García-Sanz, Mateos, Gutierrez, Lecrevisse, Blanco, Hernández, de las Heras, Martinez-Lopez, Roig, Costa, Ocio, Perez-Andres, Maiolino, Nucci, Rubia, Lahuerta, San-Miguel, Orfao, 2013, Tang, Zhao, van de Velde, Tross, Mitsiades, Viselli, Neuwirth, Esseltine, Anderson, Ghobrial, Miguel, Richardson, Tomasson, Michor, 2016). Fitting to this small amount of data, we were able to explore relationships between the sensitive parameters.

We propose a model for tumor–immune dynamics of multiple myeloma that can be used in future work. The model we propose was developed in Gallaher et al. (2018), has parameter values/ranges we based on literature searches, and has fixed values for all but the eight most sensitive parameters found in this current work. The analysis and findings in this and previous work (Gallaher et al., 2018) add to our overall confidence in the model. If individual-level clinical data become available for each of the four populations in the model, the eight sensitive parameters could be estimated. In silico exploration and optimization could then identify regimens predicted to have best outcomes. Such regimens could be tested either preclinically or clinically and compared to others to evaluate outcomes and calibrate the results.

Section snippets

The mathematical model

Our mathematical model, originally presented in Gallaher et al. (2018), consists of a system of ordinary differential equations (ODEs) which represent interactions between myeloma cells and the immune system. We include the following four populations in the peripheral blood, considered as functions of time, t: M protein produced by MM cells, M(t); natural killer (NK) cells, N(t); cytotoxic T lymphocytes (CTLs), TC(t); and regulatory T cells (Tregs), TR(t). A detailed description and

Numerical simulation

We used numerical simulation to explore the spectrum of disease dynamics given by the range of parameter values in our model system.

Sensitivity analysis

Due to the high levels of uncertainty and variability in the model parameter values, we performed sensitivity analysis to see if the values of some of the parameters are more important than others in determining the outcome of the system. We used the long-term M-protein level M as the outcome of interest, as M protein continues to be an important measure of tumor burden in patients with MM (Dimopoulos, Kyle, Fermand, Rajkumar, Miguel, Chanan-Khan, Ludwig, Joshua, Mehta, Gertz, Avet-Loiseau,

Identifiability

Model identifiability is an important step that considers the possibility of obtaining unique parameter values from data. Structural identifiability considers the problem of fitting parameters to perfect data, continuous in time and space, and is related to model structure and independent of the parameter values. Practical identifiability considers the problem for more realistic data availability and error within. We consider both types in this section.

There are three possible outcomes for

Conclusion

In this work, we explored a mathematical model of tumor–immune dynamics for MM that we originally presented in Gallaher et al. (2018). Our model uses M-protein and immune cell populations in the peripheral blood to represent the dynamics of disease burden and immune response in a patient with MM. The value of a model depends not only on the model structure, but also on the parameter values used with the model. In Gallaher et al. (2018), our focus was on the model structure; in this new work,

Disclosure statement

Zhu, Passey, Robbins, Bezman, Shelat, and Moore were employed by Bristol-Myers Squibb at the time this work was performed.

Acknowledgment

This work was initiated during the Association for Women in Mathematics collaborative workshop Women Advancing Mathematical Biology hosted by the Mathematical Biosciences Institute (MBI) at Ohio State University in April 2017. Funding for the workshop was provided by MBI, NSF ADVANCE “Career Advancement for Women Through Research-Focused Networks” (NSF-HRD 1500481), the Society for Mathematical Biology, and Microsoft Research. Editorial assistance was provided by Caudex, funded by Bristol-Myers

References (87)

  • A. Shanker et al.

    Cooperative action of CD8 T lymphocytes and natural killer cells controls tumour growth under conditions of restricted T-cell receptor diversity

    Immunology

    (2010)
  • D.Q. Tran

    TGF-β: the sword, the wand, and the shield of FOXP3+ regulatory T cells

    J Mol Cell Biol

    (2012)
  • Y.-J. Wen et al.

    Tumor lysate-specific cytotoxic t lymphocytes in multiple myeloma: promising effector cells for immunotherapy

    Blood

    (2002)
  • A.K. Abbas et al.

    Cellular and Molecular Immunology

    (2015)
  • J. Arciero et al.

    A mathematical model of tumor-immune evasion and siRNA treatment

    Discrete Continuous Dyn. Syst. Ser. B

    (2004)
  • E. Balsa-Canto et al.

    An iterative identification procedure for dynamic modeling of biochemical networks

    BMC Syst. Biol.

    (2010)
  • R.J. de Boer et al.

    Macrophage T lymphocyte interactions in the anti-tumor immune response: a mathematical model

    J. Immunol.

    (1985)
  • R.J. de Boer et al.

    Different dynamics of CD4+ and CD8+ T cell responses during and after acute lymphocytic choriomeningitis virus infection

    J. Immunol.

    (2003)
  • O. Boyman et al.

    The role of interleukin-2 during homeostasis and activation of the immune system

    Nat. Rev. Immunol.

    (2012)
  • S.P. Brooks

    Markov chain Monte Carlo method and its application

    Statistician

    (1998)
  • R. Brown et al.

    The expression of T cell related costimulatory molecules in multiple myeloma

    Leukemia Lymphoma

    (1998)
  • A. Cerwenka et al.

    Ectopic expression of retinoic acid early inducible-1 gene (RAE-1) permits natural killer cell-mediated rejection of a MHC class I-bearing tumor in vivo

    Proc. Natl. Acad. Sci.

    (2001)
  • M.-L. Chen et al.

    Regulatory T cells suppress tumor-specific CD8 T cell cytotoxicity through TGF-β signals in vivo

    Proc. Natl. Acad. Sci.

    (2005)
  • O. Chiş et al.

    GenSSI: a software toolbox for structural identifiability analysis of biological models

    Bioinformatics

    (2011)
  • G. D’Arena et al.

    Circulating regulatory T-cells in monoclonal gammopathies of uncertain significance and multiple myeloma: In search of a role

    J. Immunol. Res.

    (2016)
  • M.V. Dhodapkar et al.

    A reversible defect in natural killer T cell function characterizes the progression of premalignant to malignant multiple myeloma

    J. Exp. Med.

    (2003)
  • A. Diefenbach et al.

    Rae1 and H60 ligands of the NKG2D receptor stimulate tumor immunity

    Nature

    (2001)
  • M. Dimopoulos et al.

    Consensus recommendations for standard investigative workup: report of the International Myeloma Workshop Consensus Panel 3

    Blood

    (2011)
  • A. d’Onofrio

    A general framework for modelling tumor-immune system competition and immunotherapy: mathematical analysis and biomedical inferences

    Physica D

    (2005)
  • B.G. Durie et al.

    A clinical staging system for multiple myeloma: correlation of measured myeloma cell mass with presenting clinical features, response to treatment, and survival

    Cancer

    (1975)
  • J. Favaloro et al.

    Myeloma skews regulatory T and pro-inflammatory T helper 17 cell balance in favor of a suppressive state

    Leukemia Lymphoma

    (2014)
  • S. Feyler et al.

    CD4+CD25+FoxP3+ regulatory T cells are increased whilst CD3+CDCD8abTCR+ double negative T cells are decreased in the peripheral blood of patients with multiple myeloma which correlates with disease burden

    Br. J. Haematol.

    (2009)
  • S. Feyler et al.

    Tumour cell generation of inducible regulatory T-cells in multiple myeloma is contact-dependent and antigen-presenting cell-independent

    PLoS ONE

    (2012)
  • C. Frohn et al.

    Anti-myeloma activity of natural killer lymphocytes

    Br. J. Haematol.

    (2002)
  • J. Gallaher et al.

    A mathematical model for tumor-immune dynamics in multiple myeloma

    Understanding Complex Biol. Syst. Math

    (2018)
  • M. Gao et al.

    Myeloma cells resistance to NK cell lysis mainly involves an HLA class I-dependent mechanism

    Acta Biochimica et Biophysica Sinica (Shanghai)

    (2014)
  • F. Ghiringhelli et al.

    The role of regulatory T cells in the control of natural killer cells: relevance during tumor progression

    Immunol. Rev.

    (2006)
  • F. Ghiringhelli et al.

    CD4+ CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-β-dependent manner

    J. Exp. Med.

    (2005)
  • M. van der Giessen et al.

    Quantification of IgG subclasses in sera of normal adults and healthy children between 4 and 12 years of age

    Clin. Exp. Immunol.

    (1975)
  • G.H. Golub et al.

    Matrix Computations

    (2012)
  • A. Gonzalez-Qunitela et al.

    Serum levels of immunoglobulins (igg, iga, igm) in a general adult population and their relationship with alcohol consumption, smoking and common metabolic abnormalities

    Clin. Exp. Immunol.

    (2008)
  • P.R. Greipp et al.

    International staging system for multiple myeloma

    J. Clin. Oncol.

    (2005)
  • C.T. Hansen et al.

    Evaluation of the serum free light chain (sFLC) analysis in prediction of response in symptomatic multiple myeloma patients: Rapid profound reduction in involved FLC predicts achievement of VGPR

    Eur. J. Haematol.

    (2014)
  • Cited by (16)

    • On modeling the synergy of cancer immunotherapy with radiotherapy

      2023, Communications in Nonlinear Science and Numerical Simulation
      Citation Excerpt :

      This interplay has led to a revision of the ideas of mere cancer immunosurveillance (i.e., the immune system is able to control the cancerous growth) into what now more commonly is called cancer immunoediting [3]. Mathematical models that describe tumor-immune system interactions are plentiful ranging from low-dimensional minimally parameterized models [4–7] to large-scale agent-based, PDE and hybrid models [8–11]. While the latter enable sophisticated simulation studies, the former are amenable to a theoretical analysis [12].

    • Multi-method global sensitivity analysis of mathematical models

      2022, Journal of Theoretical Biology
      Citation Excerpt :

      Global sensitivity techniques are increasingly preferred because of the deeper insight gained when exploring the response of model output by varying all the parameters at the same time over a range of uncertainty (finite or sometimes infinite). Examples of global methods include DGSM (Derivative-based Global Sensitivity Measures) (Kucherenko et al., 2009; Kucherenko et al., 2012), the Morris screening method (Shin et al., 2013; Tian, 2013), variance-based methods such as Sobol’s method (Sobol, 1993) and the extended Fourier amplitude sensitivity test (eFAST) (Saltelli et al., 1999; Saltelli et al., 2000), analysis via singular value decomposition followed by QR factorization (Olufsen and Ottesen, 2013; Gallaher et al., 2018), and correlation-based methods such as the Partial Rank Correlation Coefficient method (PRCC) (Pearson et al., 1895; Stigler, 1989), just to name a few. Unlike local methods, global techniques are robust and can be employed in a wide variety of applications, but they are not yet as commonly used as local methods.

    • Global sensitivity analysis of biological multiscale models

      2019, Current Opinion in Biomedical Engineering
      Citation Excerpt :

      Sensitivity analysis is an area of study that involves the development of tools that assess sources of uncertainty in computational models of biological systems and that help elucidate the relationships between parameters/mechanisms and model outcomes. This type of analysis has been used in many different systems biology settings, such as models for cancer [11,18,27,6], immune response [1,8,40], ecology [4,21], drug resistance and antibiotic treatment [26], and vaccine efficacy [30,14]. There are also many other uses for sensitivity analysis, such as assisting in model fitting/calibration, evaluating differences between different modeling approaches, and determining where models can be simplified to reduce computational cost or to increase understanding of, and confidence in, simulated results.

    View all citing articles on Scopus
    1

    Contributed equally to the specification of the model and the conceptual framework.

    View full text