DRUG-NEM: Optimizing drug combinations using single-cell perturbation response to account for intratumoral heterogeneity

Significance Single-cell high-throughput technologies enable the ability to identify combination cancer therapies that account for intratumoral heterogeneity, a phenomenon that has been shown to influence the effectiveness of cancer treatment. We developed and applied an approach that identifies top-ranking drug combinations based on the single-cell perturbation response when an individual tumor sample is screened against a panel of single drugs. This approach optimizes drug combinations by choosing the minimum number of drugs that produce the maximal intracellular desired effects for an individual sample. An individual malignant tumor is composed of a heterogeneous collection of single cells with distinct molecular and phenotypic features, a phenomenon termed intratumoral heterogeneity. Intratumoral heterogeneity poses challenges for cancer treatment, motivating the need for combination therapies. Single-cell technologies are now available to guide effective drug combinations by accounting for intratumoral heterogeneity through the analysis of the signaling perturbations of an individual tumor sample screened by a drug panel. In particular, Mass Cytometry Time-of-Flight (CyTOF) is a high-throughput single-cell technology that enables the simultaneous measurements of multiple (>40) intracellular and surface markers at the level of single cells for hundreds of thousands of cells in a sample. We developed a computational framework, entitled Drug Nested Effects Models (DRUG-NEM), to analyze CyTOF single-drug perturbation data for the purpose of individualizing drug combinations. DRUG-NEM optimizes drug combinations by choosing the minimum number of drugs that produce the maximal desired intracellular effects based on nested effects modeling. We demonstrate the performance of DRUG-NEM using single-cell drug perturbation data from tumor cell lines and primary leukemia samples.

[1]  Ursula Klingmüller,et al.  Short-term information processing, long-term responses: Insights by mathematical modeling of signal transduction , 2012, BioEssays : news and reviews in molecular, cellular and developmental biology.

[2]  Gordon K Smyth,et al.  Linear Models and Empirical Bayes Methods for Assessing Differential Expression in Microarray Experiments , 2004, Statistical applications in genetics and molecular biology.

[3]  C. I. Bliss THE TOXICITY OF POISONS APPLIED JOINTLY1 , 1939 .

[4]  Kevin B. Kim,et al.  Trametinib (GSK1120212) in the treatment of melanoma , 2013, Expert opinion on pharmacotherapy.

[5]  Rainer Spang,et al.  Analyzing synergistic and non-synergistic interactions in signalling pathways using Boolean Nested Effect Models , 2015, Bioinform..

[6]  Holger Fröhlich,et al.  Analyzing gene perturbation screens with nested effects models in R and bioconductor , 2008, Bioinform..

[7]  Karen A. Ryall,et al.  Systems biology approaches for advancing the discovery of effective drug combinations , 2015, Journal of Cheminformatics.

[8]  Joshua M. Stuart,et al.  A Factor Graph Nested Effects Model To Identify Networks from Genetic Perturbations , 2009, PLoS Comput. Biol..

[9]  Kiyoshi Miyagawa,et al.  The human pre-B cell line Nalm-6 is highly proficient in gene targeting by homologous recombination. , 2006, DNA and cell biology.

[10]  Steven M Kornblau,et al.  Treatment of Philadelphia chromosome-positive acute lymphocytic leukemia with hyper-CVAD and imatinib mesylate. , 2004, Blood.

[11]  Soo-Jeong Lim,et al.  p38MAPK inhibitor SB203580 sensitizes human SNU-C4 colon cancer cells to exisulind-induced apoptosis. , 2006, Oncology reports.

[12]  Achim Tresch,et al.  Modeling the temporal interplay of molecular signaling and gene expression by using dynamic nested effects models , 2009, Proceedings of the National Academy of Sciences.

[13]  M F Greaves,et al.  A non-T, non-B human leukemia cell line (NALM-1): establishment of the cell line and presence of leukemia-associated antigens. , 1977, Journal of the National Cancer Institute.

[14]  Bo Zhang,et al.  Network target for screening synergistic drug combinations with application to traditional Chinese medicine , 2011, BMC Systems Biology.

[15]  Philip J Brown,et al.  Empirical Bayes Logistic Regression , 2008, Statistical applications in genetics and molecular biology.

[16]  P. Sorger,et al.  Sequential Application of Anticancer Drugs Enhances Cell Death by Rewiring Apoptotic Signaling Networks , 2012, Cell.

[17]  Ulrich Göbel,et al.  Long-term outcome in children with relapsed ALL by risk-stratified salvage therapy: results of trial acute lymphoblastic leukemia-relapse study of the Berlin-Frankfurt-Münster Group 87. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[18]  Ronald Koschny,et al.  The promise of TRAIL—potential and risks of a novel anticancer therapy , 2007, Journal of Molecular Medicine.

[19]  Jing Chen,et al.  Effective and selective targeting of leukemia cells using a TORC1/2 kinase inhibitor , 2010, Nature Medicine.

[20]  Douglas A. Lauffenburger,et al.  A Dynamical Systems Model for Combinatorial Cancer Therapy Enhances Oncolytic Adenovirus Efficacy by MEK-Inhibition , 2011, PLoS Comput. Biol..

[21]  Sean C. Bendall,et al.  Extracting a Cellular Hierarchy from High-dimensional Cytometry Data with SPADE , 2011, Nature Biotechnology.

[22]  Holger Fröhlich,et al.  MC EMiNEM Maps the Interaction Landscape of the Mediator , 2012, PLoS Comput. Biol..

[23]  B. Fang,et al.  Mechanisms of resistance to TRAIL-induced apoptosis in cancer , 2005, Cancer Gene Therapy.

[24]  V. Dixit,et al.  Death receptors: signaling and modulation. , 1998, Science.

[25]  Karen Sachs,et al.  Multiplexed mass cytometry profiling of cellular states perturbed by small-molecule regulators , 2012, Nature Biotechnology.

[26]  Marc Hafner,et al.  Profiles of Basal and Stimulated Receptor Signaling Networks Predict Drug Response in Breast Cancer Lines , 2013, Science Signaling.

[27]  Olga G. Troyanskaya,et al.  Nested effects models for high-dimensional phenotyping screens , 2007, ISMB/ECCB.

[28]  Mark M. Davis,et al.  Automatic Classification of Cellular Expression by Nonlinear Stochastic Embedding (ACCENSE) , 2013, Proceedings of the National Academy of Sciences.

[29]  P. Gaynon,et al.  Relapsed acute lymphoblastic leukemia: Current status and future opportunities , 2008, Current oncology reports.

[30]  Douglas A Lauffenburger,et al.  Addressing genetic tumor heterogeneity through computationally predictive combination therapy. , 2013, Cancer discovery.

[31]  Rainer Spang,et al.  Detecting hierarchical structure in molecular characteristics of disease using transitive approximations of directed graphs , 2008, Bioinform..

[32]  Lit-Hsin Loo,et al.  Early spatiotemporal-specific changes in intermediate signals are predictive of cytotoxic sensitivity to TNFα and co-treatments , 2017, Scientific Reports.

[33]  Rainer Spang,et al.  Non-transcriptional pathway features reconstructed from secondary effects of RNA interference , 2005, Bioinform..

[34]  Holger Fröhlich,et al.  Estimating large-scale signaling networks through nested effect models with intervention effects from microarray data , 2008, Bioinform..

[35]  F Hecht,et al.  Philadelphia chromosome-positive acute lymphoblastic leukemia cell lines without classical breakpoint cluster region rearrangement. , 1988, Cancer research.

[36]  Sean C. Bendall,et al.  Single-Cell Mass Cytometry of Differential Immune and Drug Responses Across a Human Hematopoietic Continuum , 2011, Science.

[37]  Sean C. Bendall,et al.  Conditional density-based analysis of T cell signaling in single-cell data , 2014, Science.

[38]  Achim Tresch,et al.  Structure Learning in Nested Effects Models , 2007, Statistical applications in genetics and molecular biology.

[39]  Garry P Nolan,et al.  Visualization and cellular hierarchy inference of single-cell data using SPADE , 2016, Nature Protocols.

[40]  Xi Zhao,et al.  CCAST: A Model-Based Gating Strategy to Isolate Homogeneous Subpopulations in a Heterogeneous Population of Single Cells , 2014, PLoS Comput. Biol..