A systems biology approach to discovering pathway signaling dysregulation in metastasis

Total metastatic burden is the primary cause of death for many cancer patients. While the process of metastasis has been studied widely, much remains to be understood. Moreover, few agents have been developed that specifically target the major steps of the metastatic cascade. Many individual genes and pathways have been implicated in metastasis but a holistic view of how these interact and cooperate to regulate and execute the process remains somewhat rudimentary. It is unclear whether all of the signaling features that regulate and execute metastasis are yet fully understood. Novel features of a complex system such as metastasis can often be discovered by taking a systems-based approach. We introduce the concepts of systems modeling and define some of the central challenges facing the application of a multidisciplinary systems-based approach to understanding metastasis and finding actionable targets therein. These challenges include appreciating the unique properties of the high-dimensional omics data often used for modeling, limitations in knowledge of the system (metastasis), tumor heterogeneity and sampling bias, and some of the issues key to understanding critical features of molecular signaling in the context of metastasis. We also provide a brief introduction to integrative modeling that focuses on both the nodes and edges of molecular signaling networks. Finally, we offer some observations on future directions as they relate to developing a systems-based model of the metastatic cascade.

[1]  Robert Clarke,et al.  UNDO: a Bioconductor R package for unsupervised deconvolution of mixed gene expressions in tumor samples , 2015, Bioinform..

[2]  Ali Najafi,et al.  Cancer systems biology and modeling: microscopic scale and multiscale approaches. , 2015, Seminars in cancer biology.

[3]  Jason W. Locasale,et al.  Transcriptional diversity and bioenergetic shift in human breast cancer metastasis revealed by single-cell RNA sequencing , 2020, Nature Cell Biology.

[4]  Jonathan R. Karr,et al.  A Whole-Cell Computational Model Predicts Phenotype from Genotype , 2012, Cell.

[5]  S. Elmore Apoptosis: A Review of Programmed Cell Death , 2007, Toxicologic pathology.

[6]  Eduard Batlle,et al.  Transforming Growth Factor-β Signaling in Immunity and Cancer. , 2019, Immunity.

[7]  E. Gehan,et al.  The properties of high-dimensional data spaces: implications for exploring gene and protein expression data , 2008, Nature Reviews Cancer.

[8]  K. Lamperska,et al.  2D and 3D cell cultures – a comparison of different types of cancer cell cultures , 2016, Archives of medical science : AMS.

[9]  Peter C. Searson,et al.  In Vitro Tumor Models: Advantages, Disadvantages, Variables, and Selecting the Right Platform , 2016, Front. Bioeng. Biotechnol..

[10]  Ryung S. Kim,et al.  Dormancy Signatures and Metastasis in Estrogen Receptor Positive and Negative Breast Cancer , 2012, PloS one.

[11]  D. Lauffenburger,et al.  Modeling a Snap-Action, Variable-Delay Switch Controlling Extrinsic Cell Death , 2008, PLoS biology.

[12]  Antonio Reverter,et al.  A Differential Wiring Analysis of Expression Data Correctly Identifies the Gene Containing the Causal Mutation , 2009, PLoS Comput. Biol..

[13]  Katherine C. Chen,et al.  Integrative analysis of cell cycle control in budding yeast. , 2004, Molecular biology of the cell.

[14]  Katherine C. Chen,et al.  Sniffers, buzzers, toggles and blinkers: dynamics of regulatory and signaling pathways in the cell. , 2003, Current opinion in cell biology.

[15]  F. J. Geske,et al.  Early stages of p53-induced apoptosis are reversible , 2001, Cell Death and Differentiation.

[16]  John J Tyson,et al.  A model for restriction point control of the mammalian cell cycle. , 2004, Journal of theoretical biology.

[17]  Pavel Kraikivski,et al.  Computational Model of G2-M DNA Damage Checkpoint Regulation in Normal and p53-null Cancer Cells , 2020, bioRxiv.

[18]  Ronald J. Moore,et al.  Integrated Proteogenomic Characterization of Human High-Grade Serous Ovarian Cancer , 2016, Cell.

[19]  Yibin Kang,et al.  Unravelling the complexity of metastasis — molecular understanding and targeted therapies , 2011, Nature Reviews Cancer.

[20]  T. M. Murali,et al.  Modeling and analysis of the macronutrient signaling network in budding yeast , 2020, bioRxiv.

[21]  Quy H. Nguyen,et al.  Experimental Considerations for Single-Cell RNA Sequencing Approaches , 2018, Front. Cell Dev. Biol..

[22]  S. Lipton,et al.  Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018 , 2018, Cell Death & Differentiation.

[23]  P. Friedl,et al.  Classifying collective cancer cell invasion , 2012, Nature Cell Biology.

[24]  S. Narod,et al.  Triple-Negative Breast Cancer: Clinical Features and Patterns of Recurrence , 2007, Clinical Cancer Research.

[25]  Robert Clarke,et al.  Systems biology: perspectives on multiscale modeling in research on endocrine-related cancers. , 2019, Endocrine-related cancer.

[26]  W. Baumann,et al.  Dynamic Modeling of the Interaction Between Autophagy and Apoptosis in Mammalian Cells , 2013, CPT: pharmacometrics & systems pharmacology.

[27]  J. Tyson,et al.  A simple model of circadian rhythms based on dimerization and proteolysis of PER and TIM. , 1999, Biophysical journal.

[28]  N. Dimitrova,et al.  InFlo: a novel systems biology framework identifies cAMP-CREB1 axis as a key modulator of platinum resistance in ovarian cancer , 2016, Oncogene.

[29]  Sarah J. Parker,et al.  Proteomic Architecture of Human Coronary and Aortic Atherosclerosis , 2018, Circulation.

[30]  S. Sleijfer,et al.  The genomic landscape of metastatic breast cancer highlights changes in mutation and signature frequencies , 2019, Nature Genetics.

[31]  E. Hoffman,et al.  Mathematical modelling of transcriptional heterogeneity identifies novel markers and subpopulations in complex tissues , 2016, Scientific Reports.

[32]  Zhongming Zhao,et al.  Transcriptome- and proteome-oriented identification of dysregulated eIF4G, STAT3, and Hippo pathways altered by PIK3CAH1047R in HER2/ER-positive breast cancer , 2016, Breast Cancer Research and Treatment.

[33]  Francisco Avila Cobos,et al.  Computational deconvolution of transcriptomics data from mixed cell populations , 2018, Bioinform..

[34]  E. Andres Houseman,et al.  Reference-free deconvolution of DNA methylation data and mediation by cell composition effects , 2016, BMC Bioinformatics.

[35]  James C. Whisstock,et al.  Perforin and granzymes: function, dysfunction and human pathology , 2015, Nature Reviews Immunology.

[36]  K. Hunter,et al.  Genetic insights into the morass of metastatic heterogeneity , 2018, Nature Reviews Cancer.

[37]  Benjamin J. Raphael,et al.  Pan-Cancer Network Analysis Identifies Combinations of Rare Somatic Mutations across Pathways and Protein Complexes , 2014, Nature Genetics.

[38]  Weiqing Wang,et al.  Perturbation Biology: Inferring Signaling Networks in Cellular Systems , 2013, PLoS Comput. Biol..

[39]  J. Ferrell,et al.  Modeling the Cell Cycle: Why Do Certain Circuits Oscillate? , 2011, Cell.

[40]  Jen-Huang Huang,et al.  Plug-and-Play In Vitro Metastasis System toward Recapitulating the Metastatic Cascade , 2019, Scientific Reports.

[41]  Sridhar Ramaswamy,et al.  Circulating Tumor Cell Clusters Are Oligoclonal Precursors of Breast Cancer Metastasis , 2014, Cell.

[42]  E. Ruppin,et al.  Computational reconstruction of tissue-specific metabolic models: application to human liver metabolism , 2010, Molecular systems biology.

[43]  E. Marcotte,et al.  Insights into the regulation of protein abundance from proteomic and transcriptomic analyses , 2012, Nature Reviews Genetics.

[44]  John J Tyson,et al.  A Dynamical Paradigm for Molecular Cell Biology. , 2020, Trends in cell biology.

[45]  Kohei Miyazono,et al.  TGFβ signalling: a complex web in cancer progression , 2010, Nature Reviews Cancer.

[46]  Maw-Shang Chang,et al.  Dynamic sensitivity analysis of biological systems , 2008, BMC Bioinformatics.

[47]  Robert Clarke,et al.  Differential dependency network analysis to identify condition-specific topological changes in biological networks , 2009, Bioinform..

[48]  Christopher J Tape,et al.  Systems Biology Analysis of Heterocellular Signaling. , 2016, Trends in biotechnology.

[49]  Paul Brazhnik,et al.  Computational analysis of dynamical responses to the intrinsic pathway of programmed cell death. , 2009, Biophysical journal.

[50]  Christina H. Stuelten,et al.  Cell motility in cancer invasion and metastasis: insights from simple model organisms , 2018, Nature Reviews Cancer.

[51]  Daniel B. Forger,et al.  A mechanism for robust circadian timekeeping via stoichiometric balance , 2012, Molecular systems biology.

[52]  R. Clarke,et al.  Approaches to working in high-dimensional data spaces: gene expression microarrays , 2008, British Journal of Cancer.

[53]  Johannes G. Reiter,et al.  An analysis of genetic heterogeneity in untreated cancers , 2019, Nature Reviews Cancer.

[54]  Jonathan W. Uhr,et al.  Controversies in clinical cancer dormancy , 2011, Proceedings of the National Academy of Sciences.

[55]  T M Murali,et al.  From START to FINISH: computational analysis of cell cycle control in budding yeast , 2015, npj Systems Biology and Applications.

[56]  Katherine C. Chen,et al.  Kinetic analysis of a molecular model of the budding yeast cell cycle. , 2000, Molecular biology of the cell.

[57]  Robert A Gatenby,et al.  Application of Evolutionary Principles to Cancer Therapy. , 2015, Cancer research.

[58]  Brendan F. Kohrn,et al.  Extensive subclonal mutational diversity in human colorectal cancer and its significance , 2019, Proceedings of the National Academy of Sciences.

[59]  Andrea Califano,et al.  ARACNe-AP: gene network reverse engineering through adaptive partitioning inference of mutual information , 2016, Bioinform..

[60]  Ioannis Xenarios,et al.  A method for the generation of standardized qualitative dynamical systems of regulatory networks , 2005, Theoretical Biology and Medical Modelling.

[61]  Robert Clarke,et al.  Dynamic modelling of oestrogen signalling and cell fate in breast cancer cells , 2011, Nature Reviews Cancer.

[62]  Jonathan R. Karr,et al.  Accelerated discovery via a whole-cell model , 2013, Nature Methods.

[63]  Mohit Kumar Jolly,et al.  Epithelial/mesenchymal plasticity: how have quantitative mathematical models helped improve our understanding? , 2017, Molecular oncology.

[64]  Randy Heiland,et al.  A Review of Cell-Based Computational Modeling in Cancer Biology , 2019, JCO clinical cancer informatics.

[65]  Yunyun Zhou,et al.  An epigenetically distinct breast cancer cell subpopulation promotes collective invasion. , 2015, The Journal of clinical investigation.

[66]  S. Adams,et al.  Variation in the Incidence and Magnitude of Tumor-Infiltrating Lymphocytes in Breast Cancer Subtypes: A Systematic Review. , 2016, JAMA oncology.

[67]  Albert Goldbeter,et al.  Temporal self-organization of the cyclin/Cdk network driving the mammalian cell cycle , 2009, Proceedings of the National Academy of Sciences.

[68]  Thomas R. Cox,et al.  Pre-metastatic niches: organ-specific homes for metastases , 2017, Nature Reviews Cancer.

[69]  D. Hanahan,et al.  The Hallmarks of Cancer , 2000, Cell.

[70]  Jingyu Zhang,et al.  TGF-β–induced epithelial-to-mesenchymal transition proceeds through stepwise activation of multiple feedback loops , 2014, Science Signaling.

[71]  Matteo Barberis,et al.  Quantitative Systems Biology to decipher design principles of a dynamic cell cycle network: the “Maximum Allowable mammalian Trade–Off–Weight” (MAmTOW) , 2017, npj Systems Biology and Applications.

[72]  C. Sander,et al.  Models from experiments: combinatorial drug perturbations of cancer cells , 2008, Molecular systems biology.

[73]  Andrea Califano,et al.  A Systems Approach to Drug Discovery in Alzheimer’s Disease , 2014, Neurotherapeutics.

[74]  Sookkyung Lim,et al.  Mathematical modeling and validation of glucose compensation of the neurospora circadian clock. , 2015, Biophysical journal.

[75]  Yahia Adnani,et al.  Genomic characterization of metastatic breast cancers , 2019, Nature.

[76]  John J Tyson,et al.  Modeling the dynamic behavior of biochemical regulatory networks. , 2019, Journal of theoretical biology.

[77]  B. Kholodenko,et al.  Control analysis of glycolytic oscillations. , 1996, Biophysical chemistry.

[78]  Xiao-ming Meng,et al.  TGF-β: the master regulator of fibrosis , 2016, Nature Reviews Nephrology.

[79]  G. Pearson,et al.  Control of Invasion by Epithelial-to-Mesenchymal Transition Programs during Metastasis , 2019, Journal of clinical medicine.

[80]  Christian A. Rees,et al.  Molecular portraits of human breast tumours , 2000, Nature.

[81]  Benjamin J. Raphael,et al.  Multiplatform Analysis of 12 Cancer Types Reveals Molecular Classification within and across Tissues of Origin , 2014, Cell.

[82]  D. Hanahan,et al.  Hallmarks of Cancer: The Next Generation , 2011, Cell.