Integrating multimodal data sets into a mathematical framework to describe and predict therapeutic resistance in cancer

A significant challenge in the field of biomedicine is the development of methods to integrate the multitude of dispersed data sets into comprehensive frameworks to be used to generate optimal clinical decisions. Recent technological advances in single cell analysis allow for high-dimensional molecular characterization of cells and populations, but to date, few mathematical models have attempted to integrate measurements from the single cell scale with other data types. Here, we present a framework that actionizes static outputs from a machine learning model and leverages these as measurements of state variables in a dynamic mechanistic model of treatment response. We apply this framework to breast cancer cells to integrate single cell transcriptomic data with longitudinal population-size data. We demonstrate that the explicit inclusion of the transcriptomic information in the parameter estimation is critical for identification of the model parameters and enables accurate prediction of new treatment regimens. Inclusion of the transcriptomic data improves predictive accuracy in new treatment response dynamics with a concordance correlation coefficient (CCC) of 0.89 compared to a prediction accuracy of CCC = 0.79 without integration of the single cell RNA sequencing (scRNA-seq) data directly into the model calibration. To the best our knowledge, this is the first work that explicitly integrates single cell clonally-resolved transcriptome datasets with longitudinal treatment response data into a mechanistic mathematical model of drug resistance dynamics. We anticipate this approach to be a first step that demonstrates the feasibility of incorporating multimodal data sets into identifiable mathematical models to develop optimized treatment regimens from data.

[1]  Geoffrey J Maher,et al.  Chromatin and Single-Cell RNA-Seq Profiling Reveal Dynamic Signaling and Metabolic Transitions during Human Spermatogonial Stem Cell Development , 2017, Cell stem cell.

[2]  Virginia L. Stonick,et al.  On a natural homotopy between linear and nonlinear single-layer networks , 1996, IEEE Trans. Neural Networks.

[3]  Thomas E Yankeelov,et al.  Clinically Relevant Modeling of Tumor Growth and Treatment Response , 2013, Science Translational Medicine.

[4]  Boris Jerchow,et al.  Molecular evolution of a novel hyperactive Sleeping Beauty transposase enables robust stable gene transfer in vertebrates , 2009, Nature Genetics.

[5]  Douglas A. Lauffenburger,et al.  Analysis of Single-Cell RNA-Seq Identifies Cell-Cell Communication Associated with Tumor Characteristics , 2018, Cell reports.

[6]  Doron Levy,et al.  Modeling intrinsic heterogeneity and growth of cancer cells. , 2015, Journal of theoretical biology.

[7]  Beate Vieth,et al.  A systematic evaluation of single cell RNA-seq analysis pipelines , 2019, Nature Communications.

[8]  H. Cho,et al.  Modelling acute myeloid leukaemia in a continuum of differentiation states , 2018, Letters in biomathematics.

[9]  Angela M. Jarrett,et al.  Calibrating a Predictive Model of Tumor Growth and Angiogenesis with Quantitative MRI , 2019, Annals of Biomedical Engineering.

[10]  Michael Hinczewski,et al.  The 2019 mathematical oncology roadmap , 2019, Physical biology.

[11]  Eduardo D. Sontag,et al.  Validation of a Mathematical Model of Cancer Incorporating Spontaneous and Induced Evolution to Drug Resistance , 2019, bioRxiv.

[12]  Erik Sundström,et al.  RNA velocity of single cells , 2018, Nature.

[13]  Thomas E. Yankeelov,et al.  Variable Cell Line Pharmacokinetics Contribute to Non-Linear Treatment Response in Heterogeneous Cell Populations , 2018, Annals of Biomedical Engineering.

[14]  J. Marioni,et al.  Pooling across cells to normalize single-cell RNA sequencing data with many zero counts , 2016, Genome Biology.

[15]  Eduardo D. Sontag,et al.  Mathematical Details on a Cancer Resistance Model , 2018, bioRxiv.

[16]  Vincent A. Traag,et al.  From Louvain to Leiden: guaranteeing well-connected communities , 2018, Scientific Reports.

[17]  Thomas E. Yankeelov,et al.  Characterizing Trastuzumab-Induced Alterations in Intratumoral Heterogeneity with Quantitative Imaging and Immunohistochemistry in HER2+ Breast Cancer , 2018, Neoplasia.

[18]  Robert A. Gatenby,et al.  Leveraging transcriptional dynamics to improve BRAF inhibitor responses in melanoma , 2019, EBioMedicine.

[19]  Kaitlyn E. Johnson,et al.  A multi-state model of chemoresistance to characterize phenotypic dynamics in breast cancer , 2017, Scientific Reports.

[20]  Ryan R. Wick,et al.  Unicycler: Resolving bacterial genome assemblies from short and long sequencing reads , 2016, bioRxiv.

[21]  Fabian J Theis,et al.  SCANPY: large-scale single-cell gene expression data analysis , 2018, Genome Biology.

[22]  Amy Brock,et al.  Control of Lineage-Specific Gene Expression by Functionalized gRNA Barcodes. , 2018, ACS synthetic biology.

[23]  Tian Zhang,et al.  Prostate-specific antigen dynamics predict individual responses to intermittent androgen deprivation , 2019, Nature Communications.

[24]  Itay Tirosh,et al.  Single-Cell RNA Sequencing in Cancer: Lessons Learned and Emerging Challenges. , 2019, Molecular cell.

[25]  Jinzhou Yuan,et al.  Single-Cell Transcriptomic Analysis of Tumor Heterogeneity. , 2018, Trends in cancer.

[26]  Y. Censor Pareto optimality in multiobjective problems , 1977 .

[27]  Thomas E Yankeelov,et al.  Toward a science of tumor forecasting for clinical oncology. , 2015, Cancer research.

[28]  Kaitlyn E. Johnson,et al.  Directional inconsistency between Response Evaluation Criteria in Solid Tumors (RECIST) time to progression and response speed and depth. , 2019, European journal of cancer.

[29]  B. Frieden,et al.  Adaptive therapy. , 2009, Cancer research.

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

[31]  Richard Ballweg,et al.  Designing combination therapies with modeling chaperoned machine learning , 2019, PLoS Comput. Biol..

[32]  Thomas E Yankeelov,et al.  A Predictive Mathematical Modeling Approach for the Study of Doxorubicin Treatment in Triple Negative Breast Cancer , 2017, Scientific Reports.

[33]  Gioele La Manno,et al.  Quantitative single-cell RNA-seq with unique molecular identifiers , 2013, Nature Methods.

[34]  Philipp M. Altrock,et al.  Leveraging Single-Cell RNA Sequencing Experiments to Model Intratumor Heterogeneity , 2018, JCO clinical cancer informatics.

[35]  Jan Poleszczuk,et al.  Cancer Stem Cell Plasticity as Tumor Growth Promoter and Catalyst of Population Collapse , 2015, bioRxiv.

[36]  Marisa C. Eisenberg,et al.  Input-output equivalence and identifiability: some simple generalizations of the differential algebra approach , 2013 .

[37]  Patrick S. Stumpf,et al.  Stem Cell Differentiation as a Non-Markov Stochastic Process , 2017, Cell systems.

[38]  Angela M. Jarrett,et al.  Mathematical modelling of trastuzumab-induced immune response in an in vivo murine model of HER2+ breast cancer. , 2018, Mathematical medicine and biology : a journal of the IMA.

[39]  Sarah A Teichmann,et al.  A test metric for assessing single-cell RNA-seq batch correction , 2018, Nature Methods.

[40]  Shuang Wu,et al.  Evaluation of single-cell classifiers for single-cell RNA sequencing data sets , 2019, Briefings Bioinform..

[41]  Kujtim Latifi,et al.  A proliferation saturation index to predict radiation response and personalize radiotherapy fractionation , 2015, Radiation oncology.

[42]  Alissa M. Weaver,et al.  Tumor Morphology and Phenotypic Evolution Driven by Selective Pressure from the Microenvironment , 2006, Cell.

[43]  Ava Kwong,et al.  Long non-coding RNA NEAT1 confers oncogenic role in triple-negative breast cancer through modulating chemoresistance and cancer stemness , 2019, Cell Death & Disease.

[44]  Amy Brock,et al.  Single-cell RNA sequencing of lung adenocarcinoma reveals heterogeneity of immune response-related genes. , 2018, JCI insight.

[45]  Jan Poleszczuk,et al.  The Optimal Radiation Dose to Induce Robust Systemic Anti-Tumor Immunity , 2018, International journal of molecular sciences.

[46]  R. Marschalek,et al.  Optimized Sleeping Beauty transposons rapidly generate stable transgenic cell lines. , 2015, Biotechnology journal.

[47]  Angela M. Jarrett,et al.  Mathematical models of tumor cell proliferation: A review of the literature , 2018, Expert review of anticancer therapy.

[48]  Eduardo Sontag,et al.  On two definitions of observation spaces , 1989 .

[49]  Eduardo D. Sontag Dynamic compensation, parameter identifiability, and equivariances , 2017, PLoS Comput. Biol..

[50]  N G Cogan,et al.  Global sensitivity analysis used to interpret biological experimental results , 2015, Journal of mathematical biology.

[51]  Fabian J Theis,et al.  Current best practices in single‐cell RNA‐seq analysis: a tutorial , 2019, Molecular systems biology.

[52]  J. Mesirov,et al.  Automated high-dimensional flow cytometric data analysis , 2009, Proceedings of the National Academy of Sciences.

[53]  Charles H. Yoon,et al.  Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq , 2016, Science.

[54]  J. Ajani,et al.  iTALK: an R Package to Characterize and Illustrate Intercellular Communication , 2019, bioRxiv.

[55]  Jin He,et al.  Personalized Approaches to Gastrointestinal Cancers: Importance of Integrating Genomic Information to Guide Therapy. , 2015, The Surgical clinics of North America.

[56]  Rafael Meza,et al.  A Systematic Approach to Determining the Identifiability of Multistage Carcinogenesis Models , 2017, Risk analysis : an official publication of the Society for Risk Analysis.

[57]  E. Holland,et al.  Optimization of radiation dosing schedules for proneural glioblastoma , 2016, Journal of mathematical biology.

[58]  Eduardo D Sontag,et al.  Mathematical Approach to Differentiate Spontaneous and Induced Evolution to Drug Resistance During Cancer Treatment , 2019, JCO clinical cancer informatics.

[59]  Suzanne L Robertson,et al.  Identifiability and estimation of multiple transmission pathways in cholera and waterborne disease. , 2013, Journal of theoretical biology.

[60]  Kaitlyn E. Johnson,et al.  Expressed barcodes enable clonal characterization of chemotherapeutic responses in chronic lymphocytic leukemia , 2019, bioRxiv.

[61]  Thomas E. Yankeelov,et al.  Precision Medicine with Imprecise Therapy: Computational Modeling for Chemotherapy in Breast Cancer1 , 2018, Translational oncology.