Identifying Drug Effects via Pathway Alterations using an Integer Linear Programming Optimization Formulation on Phosphoproteomic Data

Understanding the mechanisms of cell function and drug action is a major endeavor in the pharmaceutical industry. Drug effects are governed by the intrinsic properties of the drug (i.e., selectivity and potency) and the specific signaling transduction network of the host (i.e., normal vs. diseased cells). Here, we describe an unbiased, phosphoproteomic-based approach to identify drug effects by monitoring drug-induced topology alterations. With our proposed method, drug effects are investigated under diverse stimulations of the signaling network. Starting with a generic pathway made of logical gates, we build a cell-type specific map by constraining it to fit 13 key phopshoprotein signals under 55 experimental conditions. Fitting is performed via an Integer Linear Program (ILP) formulation and solution by standard ILP solvers; a procedure that drastically outperforms previous fitting schemes. Then, knowing the cell's topology, we monitor the same key phosphoprotein signals under the presence of drug and we re-optimize the specific map to reveal drug-induced topology alterations. To prove our case, we make a topology for the hepatocytic cell-line HepG2 and we evaluate the effects of 4 drugs: 3 selective inhibitors for the Epidermal Growth Factor Receptor (EGFR) and a non-selective drug. We confirm effects easily predictable from the drugs' main target (i.e., EGFR inhibitors blocks the EGFR pathway) but we also uncover unanticipated effects due to either drug promiscuity or the cell's specific topology. An interesting finding is that the selective EGFR inhibitor Gefitinib inhibits signaling downstream the Interleukin-1alpha (IL1α) pathway; an effect that cannot be extracted from binding affinity-based approaches. Our method represents an unbiased approach to identify drug effects on small to medium size pathways which is scalable to larger topologies with any type of signaling interventions (small molecules, RNAi, etc). The method can reveal drug effects on pathways, the cornerstone for identifying mechanisms of drug's efficacy.

[1]  M. Gerstein,et al.  A Bayesian Networks Approach for Predicting Protein-Protein Interactions from Genomic Data , 2003, Science.

[2]  S. Schreiber,et al.  Printing proteins as microarrays for high-throughput function determination. , 2000, Science.

[3]  P. Shannon,et al.  Cytoscape: a software environment for integrated models of biomolecular interaction networks. , 2003, Genome research.

[4]  E. Butcher,et al.  Can cell systems biology rescue drug discovery? , 2007, Ernst Schering Research Foundation workshop.

[5]  David Kendrick,et al.  GAMS, a user's guide , 1988, SGNM.

[6]  V. Grantcharova,et al.  Therapeutically Targeting ErbB3: A Key Node in Ligand-Induced Activation of the ErbB Receptor–PI3K Axis , 2009, Science Signaling.

[7]  Mindy I. Davis,et al.  A quantitative analysis of kinase inhibitor selectivity , 2008, Nature Biotechnology.

[8]  John G. Albeck,et al.  Cue-Signal-Response Analysis of TNF-Induced Apoptosis by Partial Least Squares Regression of Dynamic Multivariate Data , 2004, J. Comput. Biol..

[9]  S. L. Wong,et al.  Towards a proteome-scale map of the human protein–protein interaction network , 2005, Nature.

[10]  Lili X. Peng,et al.  A High-throughput Quantitative Multiplex Kinase Assay for Monitoring Information Flow in Signaling Networks , 2003, Molecular & Cellular Proteomics.

[11]  P. Bork,et al.  Literature mining for the biologist: from information retrieval to biological discovery , 2006, Nature Reviews Genetics.

[12]  S. Hall,et al.  Chemoproteomics-driven drug discovery: addressing high attrition rates. , 2006, Drug discovery today.

[13]  M. Chaparro,et al.  Review article: pharmacological therapy for hepatocellular carcinoma with sorafenib and other oral agents , 2008, Alimentary pharmacology & therapeutics.

[14]  Paul I. Barton,et al.  Process-wide integration of solvent mixtures , 1999 .

[15]  Diego di Bernardo,et al.  Identifying Network of Drug Mode of Action by Gene Expression Profiling , 2009, J. Comput. Biol..

[16]  J. McCubrey,et al.  BAY-43-9006 Bayer/Onyx. , 2003, Current opinion in investigational drugs.

[17]  G. Siemeister,et al.  Off‐Target Decoding of a Multitarget Kinase Inhibitor by Chemical Proteomics , 2009, Chembiochem : a European journal of chemical biology.

[18]  Min Wu,et al.  Fishing for targets: novel approaches using small molecule baits. , 2004, Drug discovery today. Technologies.

[19]  Steven A Carr,et al.  Identifying the proteins to which small-molecule probes and drugs bind in cells , 2009, Proceedings of the National Academy of Sciences.

[20]  Hiroaki Kitano,et al.  Foundations of systems biology , 2001 .

[21]  Bernhard Kuster,et al.  Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors , 2007, Nature Biotechnology.

[22]  Steffen Klamt,et al.  The Logic of EGFR/ErbB Signaling: Theoretical Properties and Analysis of High-Throughput Data , 2009, PLoS Comput. Biol..

[23]  Julio Saez-Rodriguez,et al.  Fuzzy Logic Analysis of Kinase Pathway Crosstalk in TNF/EGF/Insulin-Induced Signaling , 2007, PLoS Comput. Biol..

[24]  Masao Nagasaki,et al.  Foundations of Systems Biology - Using Cell Illustrator® and Pathway Databases , 2009, Computational Biology.

[25]  L. Wodicka,et al.  A small molecule–kinase interaction map for clinical kinase inhibitors , 2005, Nature Biotechnology.

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

[27]  K. Sachs,et al.  Causal Protein-Signaling Networks Derived from Multiparameter Single-Cell Data , 2005, Science.

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

[29]  D. Auclair,et al.  BAY 43-9006 Exhibits Broad Spectrum Oral Antitumor Activity and Targets the RAF/MEK/ERK Pathway and Receptor Tyrosine Kinases Involved in Tumor Progression and Angiogenesis , 2004, Cancer Research.

[30]  Paul A Clemons,et al.  The Connectivity Map: Using Gene-Expression Signatures to Connect Small Molecules, Genes, and Disease , 2006, Science.

[31]  Hong Ma,et al.  Anti-tumor activity of GW572016: a dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways , 2002, Oncogene.

[32]  S. Brahmachari,et al.  Boolean network analysis of a neurotransmitter signaling pathway. , 2007, Journal of theoretical biology.

[33]  Julio Saez-Rodriguez,et al.  Flexible informatics for linking experimental data to mathematical models via DataRail , 2008, Bioinform..

[34]  Klaus Truemper,et al.  Logic Integer Programming Models for Signaling Networks , 2008, J. Comput. Biol..

[35]  Steffen Klamt,et al.  Structural and functional analysis of cellular networks with CellNetAnalyzer , 2007, BMC Systems Biology.

[36]  Alexander Mitsos,et al.  Optimal automatic reaction and species elimination in kinetic mechanisms , 2008 .

[37]  Arthur W. Westerberg,et al.  Optimization for design problems having more than one objective , 1983 .

[38]  M. Mann,et al.  Kinase-selective enrichment enables quantitative phosphoproteomics of the kinome across the cell cycle. , 2008, Molecular cell.

[39]  D. Lauffenburger,et al.  A Compendium of Signals and Responses Triggered by Prodeath and Prosurvival Cytokines*S , 2005, Molecular & Cellular Proteomics.

[40]  D. Lauffenburger,et al.  Discrete logic modelling as a means to link protein signalling networks with functional analysis of mammalian signal transduction , 2009, Molecular systems biology.

[41]  S. Kauffman Metabolic stability and epigenesis in randomly constructed genetic nets. , 1969, Journal of theoretical biology.

[42]  Krystal J Alligood,et al.  A Unique Structure for Epidermal Growth Factor Receptor Bound to GW572016 (Lapatinib) , 2004, Cancer Research.

[43]  J. Collins,et al.  Chemogenomic profiling on a genome-wide scale using reverse-engineered gene networks , 2005, Nature Biotechnology.

[44]  Mark Robinson,et al.  A short-term operational planning model for natural gas production systems† , 2008 .

[45]  Julio Saez-Rodriguez,et al.  High‐Throughput Protein‐Based Technologies and Computational Models for Drug Development, Efficacy, and Toxicity , 2008 .

[46]  T. Gardner,et al.  The mode-of-action by network identification (MNI) algorithm: a network biology approach for molecular target identification , 2006, Nature Protocols.

[47]  Steffen Klamt,et al.  Hypergraphs and Cellular Networks , 2009, PLoS Comput. Biol..

[48]  P Norman,et al.  OSI-774 OSI Pharmaceuticals. , 2001, Current opinion in investigational drugs.

[49]  Kevan M Shokat,et al.  Features of selective kinase inhibitors. , 2005, Chemistry & biology.

[50]  P. Zarrinkar,et al.  High-throughput kinase profiling as a platform for drug discovery , 2008, Nature Reviews Drug Discovery.