Population modeling of tumor growth curves and the reduced Gompertz model improve prediction of the age of experimental tumors

Tumor growth curves are classically modeled by means of ordinary differential equations. In analyzing the Gompertz model several studies have reported a striking correlation between the two parameters of the model, which could be used to reduce the dimensionality and improve predictive power. We analyzed tumor growth kinetics within the statistical framework of nonlinear mixed-effects (population approach). This allowed the simultaneous modeling of tumor dynamics and inter-animal variability. Experimental data comprised three animal models of breast and lung cancers, with 833 measurements in 94 animals. Candidate models of tumor growth included the exponential, logistic and Gompertz models. The exponential and—more notably—logistic models failed to describe the experimental data whereas the Gompertz model generated very good fits. The previously reported population-level correlation between the Gompertz parameters was further confirmed in our analysis (R2 > 0.92 in all groups). Combining this structural correlation with rigorous population parameter estimation, we propose a reduced Gompertz function consisting of a single individual parameter (and one population parameter). Leveraging the population approach using Bayesian inference, we estimated times of tumor initiation using three late measurement timepoints. The reduced Gompertz model was found to exhibit the best results, with drastic improvements when using Bayesian inference as compared to likelihood maximization alone, for both accuracy and precision. Specifically, mean accuracy (prediction error) was 12.2% versus 78% and mean precision (width of the 95% prediction interval) was 15.6 days versus 210 days, for the breast cancer cell line. These results demonstrate the superior predictive power of the reduced Gompertz model, especially when combined with Bayesian estimation. They offer possible clinical perspectives for personalized prediction of the age of a tumor from limited data at diagnosis. The code and data used in our analysis are publicly available at https://github.com/cristinavaghi/plumky.

[1]  Athanassios Iliadis,et al.  Mathematical modeling of tumor growth and metastatic spreading: validation in tumor-bearing mice. , 2014, Cancer research.

[2]  S. Benzekry Machine learning versus mechanistic modeling for prediction of metastatic relapse in breast cancer , 2019 .

[3]  Angelo Iollo,et al.  Prediction of the Evolution of Thyroidal Lung Nodules Using a Mathematical Model , 2010, ERCIM News.

[4]  G. Steel,et al.  Growth kinetics of tumours : cell population kinetics in relation to the growth and treatment of cancer , 1977 .

[5]  Jan Fagerberg,et al.  Model-based prediction of phase III overall survival in colorectal cancer on the basis of phase II tumor dynamics. , 2009, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[6]  L. G. Pillis,et al.  A Comparison and Catalog of Intrinsic Tumor Growth Models , 2013, Bulletin of mathematical biology.

[7]  Ken Ito,et al.  An Integrated Computational/Experimental Model of Lymphoma Growth , 2013, PLoS Comput. Biol..

[8]  Elena Bogdanovic,et al.  Vascular endothelial growth factor-mediated decrease in plasma soluble vascular endothelial growth factor receptor-2 levels as a surrogate biomarker for tumor growth. , 2008, Cancer research.

[9]  E. Rofstad,et al.  Growth characteristics of human melanoma xenografts , 1982, Cell and tissue kinetics.

[10]  Alberto Gandolfi,et al.  A family of models of angiogenesis and anti-angiogenesis anti-cancer therapy. , 2008, Mathematical medicine and biology : a journal of the IMA.

[11]  M. Carré,et al.  From 3D spheroids to tumor bearing mice: efficacy and distribution studies of trastuzumab-docetaxel immunoliposome in breast cancer , 2018, International journal of nanomedicine.

[12]  F. Barlesi,et al.  Quantitative mathematical modeling of clinical brain metastasis dynamics in non-small cell lung cancer , 2019, Scientific Reports.

[13]  Andrej Pázman,et al.  Nonlinear Regression , 2019, Handbook of Regression Analysis With Applications in R.

[14]  James P. Freyer,et al.  Tumor growthin vivo and as multicellular spheroids compared by mathematical models , 1994, Bulletin of mathematical biology.

[15]  M. Firat,et al.  Nonlinear mixed effects modeling of growth in Japanese quail. , 2013, Poultry science.

[16]  C. Nicolò,et al.  Machine learning and mechanistic modeling for prediction of metastatic relapse in early-stage breast cancer , 2019, bioRxiv.

[17]  L. Norton A Gompertzian model of human breast cancer growth. , 1988, Cancer research.

[18]  Z. Agur,et al.  The growth law of primary breast cancer as inferred from mammography screening trials data. , 1998, British Journal of Cancer.

[19]  Athanassios Iliadis,et al.  Revisiting Dosing Regimen Using Pharmacokinetic/Pharmacodynamic Mathematical Modeling: Densification and Intensification of Combination Cancer Therapy , 2016, Clinical Pharmacokinetics.

[20]  John K Kruschke,et al.  Bayesian data analysis. , 2010, Wiley interdisciplinary reviews. Cognitive science.

[21]  M. Zuidhof,et al.  Estimation of growth parameters using a nonlinear mixed Gompertz model. , 2004, Poultry science.

[22]  V. P. Collins,et al.  Observations on growth rates of human tumors. , 1956, The American journal of roentgenology, radium therapy, and nuclear medicine.

[23]  C. Winsor,et al.  The Gompertz Curve as a Growth Curve. , 1932, Proceedings of the National Academy of Sciences of the United States of America.

[24]  J S Spratt,et al.  Decelerating growth and human breast cancer , 1993, Cancer.

[25]  T E Wheldon,et al.  THE GOMPERTZ EQUATION AND THE CONSTRUCTION OF TUMOUR GROWTH CURVES , 1980, Cell and tissue kinetics.

[26]  Hervé Delingette,et al.  Image Guided Personalization of Reaction-Diffusion Type Tumor Growth Models Using Modified Anisotropic Eikonal Equations , 2010, IEEE Transactions on Medical Imaging.

[27]  Zinnia P. Parra-Guillen,et al.  Systematic Modeling and Design Evaluation of Unperturbed Tumor Dynamics in Xenografts , 2018, The Journal of Pharmacology and Experimental Therapeutics.

[28]  Yuri Kogan,et al.  Predicting Outcomes of Prostate Cancer Immunotherapy by Personalized Mathematical Models , 2010, PloS one.

[29]  Carl M. O’Brien,et al.  Mixed Effects Models for the Population Approach: Models, Tasks, Methods and Tools , 2015 .

[30]  L. Aarons,et al.  Mixed Effects Models for the Population Approach: Models, Tasks, Methods, and Tools , 2015, CPT: Pharmacometrics & Systems Pharmacology.

[31]  A. d’Onofrio Fractal growth of tumors and other cellular populations : linking the mechanistic to the phenomenological modeling and vice versa , 2014 .

[32]  Yuri Kogan,et al.  Reconsidering the paradigm of cancer immunotherapy by computationally aided real-time personalization. , 2012, Cancer research.

[33]  Matija Snuderl,et al.  Coevolution of solid stress and interstitial fluid pressure in tumors during progression: implications for vascular collapse. , 2013, Cancer research.

[34]  Philipp M. Altrock,et al.  The mathematics of cancer: integrating quantitative models , 2015, Nature Reviews Cancer.

[35]  Nicole Radde,et al.  Hamiltonian Monte Carlo methods for efficient parameter estimation in steady state dynamical systems , 2014, BMC Bioinformatics.

[36]  T. Wheldon,et al.  Characteristic Species Dependent Growth Patterns of Mammalian Neoplasms , 1978, Cell and tissue kinetics.

[37]  Laird Ak DYNAMICS OF TUMOR GROWTH. , 1964 .

[38]  Vinay G. Vaidya,et al.  Evaluation of some mathematical models for tumor growth. , 1982, International journal of bio-medical computing.

[39]  S. Salmon,et al.  Kinetics of tumor growth and regression in IgG multiple myeloma. , 1972, The Journal of clinical investigation.

[40]  David Basanta,et al.  Invasion and proliferation kinetics in enhancing gliomas predict IDH1 mutation status. , 2014, Neuro-oncology.

[41]  Albert E. Casey,et al.  The Experimental Alteration of Malignancy with an Homologous Mammalian Tumor Material: I. Results with Intratesticular Inoculation , 1934 .

[42]  Paolo Magni,et al.  Predictive Pharmacokinetic-Pharmacodynamic Modeling of Tumor Growth Kinetics in Xenograft Models after Administration of Anticancer Agents , 2004, Cancer Research.

[43]  Nicolas André,et al.  Computational oncology — mathematical modelling of drug regimens for precision medicine , 2016, Nature Reviews Clinical Oncology.

[44]  L. Norton,et al.  Predicting the course of Gompertzian growth , 1976, Nature.

[45]  Johan Pallud,et al.  A Tumor Growth Inhibition Model for Low-Grade Glioma Treated with Chemotherapy or Radiotherapy , 2012, Clinical Cancer Research.

[46]  Michalis Mastri,et al.  Modeling Spontaneous Metastasis following Surgery: An In Vivo-In Silico Approach. , 2016, Cancer research.

[47]  É. Moulines,et al.  Convergence of a stochastic approximation version of the EM algorithm , 1999 .

[48]  R Demicheli Growth of testicular neoplasm lung metastases: tumor-specific relation between two Gompertzian parameters. , 1980, European journal of cancer.

[49]  G G Steel SPECIES‐DEPENDENT GROWTH PATTERNS FOR MAMMALIAN NEOPLASMS , 1980, Cell and tissue kinetics.

[50]  L E Friberg,et al.  A Review of Mixed-Effects Models of Tumor Growth and Effects of Anticancer Drug Treatment Used in Population Analysis , 2014, CPT: pharmacometrics & systems pharmacology.

[51]  T E Wheldon,et al.  PREDICTION OF THE COMPLETE GROWTH PATTERN OF HUMAN MULTIPLE MYELOMA FROM RESTRICTED INITIAL MEASUREMENTS , 1977, Cell and tissue kinetics.

[52]  Stacey D. Finley,et al.  Effect of tumor microenvironment on tumor VEGF during anti-VEGF treatment: systems biology predictions. , 2013, Journal of the National Cancer Institute.

[53]  A S Glicksman,et al.  Growth in solid heterogeneous human colon adenocarcinomas: comparison of simple logistical models , 1987, Cell and tissue kinetics.

[54]  Jiqiang Guo,et al.  Stan: A Probabilistic Programming Language. , 2017, Journal of statistical software.

[55]  T. Wheldon Mathematical models in cancer research , 1988 .

[56]  J. Bertram,et al.  Establishment of a cloned line of Lewis Lung Carcinoma cells adapted to cell culture. , 1980, Cancer letters.

[57]  D P Fyhrie,et al.  Gompertzian growth curves in parathyroid tumours: further evidence for the set‐point hypothesis , 1997, Cell proliferation.

[58]  C. Frenzen,et al.  A cellk kinetics justification for Gompertz' equation , 1986 .

[59]  L. V. van't Veer,et al.  70-Gene Signature as an Aid to Treatment Decisions in Early-Stage Breast Cancer. , 2016, The New England journal of medicine.

[60]  John M. L. Ebos,et al.  Classical Mathematical Models for Description and Prediction of Experimental Tumor Growth , 2014, PLoS Comput. Biol..

[61]  Z Bajzer,et al.  Analysis of growth of multicellular tumour spheroids by mathematical models , 1994, Cell proliferation.