Quantitative and Systems Pharmacology in the Post-genomic Era : New Approaches to Discovering Drugs and Understanding Therapeutic

In a situation where the overall behavior of the system depends on many different components, horizontal integration synthesizes a composite model from several components having the same spatial and/or temporal granularity. In multi-cellular organisms, many cell and tissue behaviors are the consequences of interactions among local components. Integrated system models are expected to be able to uncover (i) precisely how components (at multiple levels) interact, (ii) the magnitudes of emergent properties (multiscale phenomena not explained by the sum total of local mechanisms), and how this emergent behavior can be modeled, (iii) the extent to which different biological circuits conform to common principles of “design”, and (iv) optimal ways to alter cellular and organ phenotypes using pharmacological agents. Discussion Systems integration is the act of assembling a composite system—computer models in QSP—from previously autonomous components. Horizontal integration synthesizes a composite from components having the same spatial and/or temporal granularity. An example might be a cell pathway interaction model composed of various networks within and across different cells in various cell systems, but not including tissue or molecular dynamics. A clear statement of current and future uses to which an integrated system will be put is a precondition of systems integration for scientific research (this represents the requirement of fit-to-purpose for models). A use statement typically begins with the current capabilities of the individual components followed by listing the expected capabilities of the integrated system. It seems unlikely that biological component interactions will be exclusively either horizontal or vertical. We must therefore anticipate that issues of horizontal and vertical component integration will overlap and even merge into a single problem of multi-scale integration. Impact What are the uses of horizontally integrated system models? Such systems will be used as stand-alone software components to study specific networks, and they will also be used as components in larger horizontally and vertically integrated systems. Such uses depend on the driving biological problems. If the integrated model is intended to represent an organism's pharmacological response, for example, then the duration of the response cycle, the number of cycles considered, and required response granularity each become determining aspects. With that in mind, model and component reuse, flexibility, and adaptability, become important and that feeds back into model and component design. Therefore, it should be relatively easy to reconfigure components to represent different mechanistic hypotheses or different aspects of a key attribute under different experimental conditions. It should also be relatively simple to accommodate additional aspects at the current level of granularity or alter usage and assumptions, without requiring significant component or system reengineering. Components should be constructed so that they can be adapted easily to function as components in different, integrated models. By way of example, the ability to target signaling and transcriptional pathways that drive diseases such as cancer will be enhanced by a more global understanding of how these pathways interconnect to create, through feedback and cross-talk mechanisms, the full signaling network that integrates all signals into a net outcome or phenotype (e.g. oncogenic transformations). The broader biomedical research community is searching for the underlying rules that govern signaling, while cancer researchers are simultaneously addressing through technology development the need to measure variations between tumor types, between tumors from different patients, and even within tumors through single cell measurements (e.g. integrated microfluidic-based assays). Using cancer models such as Bcr-Abl driven leukemic transformation, we are already collecting high dimensionality phosphoprofiling signaling data focused specifically on subnetworks that involve the cross-talk between a small numbers of signaling modules. Simplification of the problem through subnetwork analysis, allows us to first focus on a more tractable scale, while retaining clinical relevance, with the hope that

[1]  J. A. Hendricks,et al.  In vivo PET imaging of histone deacetylases by 18F-suberoylanilide hydroxamic acid (18F-SAHA). , 2011, Journal of medicinal chemistry.

[2]  D. Swinney,et al.  How were new medicines discovered? , 2011, Nature Reviews Drug Discovery.

[3]  Neil Benson,et al.  Systems Pharmacology: Bridging Systems Biology and Pharmacokinetics-Pharmacodynamics (PKPD) in Drug Discovery and Development , 2011, Pharmaceutical Research.

[4]  Andrew Johnston,et al.  Genome-Wide Expression Profiling of Five Mouse Models Identifies Similarities and Differences with Human Psoriasis , 2011, PloS one.

[5]  M. Duffy,et al.  Use of molecular markers for predicting therapy response in cancer patients. , 2011, Cancer treatment reviews.

[6]  Peter K. Sorger,et al.  Measuring and Modeling Apoptosis in Single Cells , 2011, Cell.

[7]  M. Joyner,et al.  Physiology: alone at the bottom, alone at the top , 2011, The Journal of physiology.

[8]  J. DiMasi,et al.  Pharmaceutical Innovation in the 21st Century: New Drug Approvals in the First Decade, 2000–2009 , 2011, Clinical pharmacology and therapeutics.

[9]  Bin Hu,et al.  Hierarchical graphs for rule-based modeling of biochemical systems , 2011, BMC Bioinformatics.

[10]  M. Elowitz,et al.  Functional roles for noise in genetic circuits , 2010, Nature.

[11]  Marc Pfister,et al.  Fostering Culture and Optimizing Organizational Structure for Implementing Model‐Based Drug Development , 2010, Journal of clinical pharmacology.

[12]  William W. Chen,et al.  Classic and contemporary approaches to modeling biochemical reactions. , 2010, Genes & development.

[13]  G. Verheyen,et al.  Screening for phospholipidosis induced by central nervous drugs: comparing the predictivity of an in vitro assay to high throughput in silico assays. , 2010, Toxicology in vitro : an international journal published in association with BIBRA.

[14]  P Vicini,et al.  Multiscale Modeling in Drug Discovery and Development: Future Opportunities and Present Challenges , 2010, Clinical pharmacology and therapeutics.

[15]  R. Iyengar,et al.  Training in Systems Pharmacology: Predoctoral Program in Pharmacology and Systems Biology at Mount Sinai School of Medicine , 2010, Clinical pharmacology and therapeutics.

[16]  K. Burrage,et al.  The Systems Biology Approach to Drug Development: Application to Toxicity Assessment of Cardiac Drugs , 2010, Clinical pharmacology and therapeutics.

[17]  P. Sorger,et al.  Dissecting Variability in Responses to Cancer Chemotherapy Through Systems Pharmacology , 2010, Clinical pharmacology and therapeutics.

[18]  Mariano J. Alvarez,et al.  A human B-cell interactome identifies MYB and FOXM1 as master regulators of proliferation in germinal centers , 2010, Molecular systems biology.

[19]  G. Ramani,et al.  Update on the clinical utility of sildenafil in the treatment of pulmonary arterial hypertension , 2010, Drug design, development and therapy.

[20]  R. Iyengar,et al.  Systems approaches to polypharmacology and drug discovery. , 2010, Current opinion in drug discovery & development.

[21]  D. Lauffenburger,et al.  Networks Inferred from Biochemical Data Reveal Profound Differences in Toll-like Receptor and Inflammatory Signaling between Normal and Transformed Hepatocytes* , 2010, Molecular & Cellular Proteomics.

[22]  R. Weissleder,et al.  Hybrid PET-optical imaging using targeted probes , 2010, Proceedings of the National Academy of Sciences.

[23]  Charles C. Persinger,et al.  How to improve R&D productivity: the pharmaceutical industry's grand challenge , 2010, Nature Reviews Drug Discovery.

[24]  J. Uhm,et al.  The transcriptional network for mesenchymal transformation of brain tumours , 2010 .

[25]  Sabrina L Spencer,et al.  Non-genetic Cell-to-cell Variability and the Consequences for Pharmacology This Review Comes from a Themed Issue on Omics Edited the Distribution of Protein Abundance and Resulting Variability in Phenotype Measuring Cell-to-cell Variation , 2022 .

[26]  Johan Gabrielsson,et al.  Pharmacokinetic-pharmacodynamic reasoning in drug discovery and early development. , 2009, Future medicinal chemistry.

[27]  Dorothy D. Sears,et al.  Mechanisms of human insulin resistance and thiazolidinedione-mediated insulin sensitization , 2009, Proceedings of the National Academy of Sciences.

[28]  Michael J. Keiser,et al.  Predicting new molecular targets for known drugs , 2009, Nature.

[29]  Sarala M. Wimalaratne,et al.  The Systems Biology Graphical Notation , 2009, Nature Biotechnology.

[30]  Aviv Regev,et al.  Transcriptional Regulatory Circuits: Predicting Numbers from Alphabets , 2009, Science.

[31]  M. Boguski,et al.  Repurposing with a Difference , 2009, Science.

[32]  P. Sorger,et al.  Non-genetic origins of cell-to-cell variability in TRAIL-induced apoptosis , 2009, Nature.

[33]  Janice M Reichert,et al.  Development trends for therapeutic antibody fragments , 2009, Nature Biotechnology.

[34]  Vincent Danos,et al.  Internal coarse-graining of molecular systems , 2009, Proceedings of the National Academy of Sciences.

[35]  B. Agoram,et al.  Use of pharmacokinetic/ pharmacodynamic modelling for starting dose selection in first-in-human trials of high-risk biologics. , 2009, British journal of clinical pharmacology.

[36]  Tanya Coleman,et al.  In Silico and in Vitro Modeling of Hepatocyte Drug Transport Processes: Importance of ABCC2 Expression Levels in the Disposition of Carboxydichlorofluroscein , 2009, Drug Metabolism and Disposition.

[37]  Ravi Iyengar,et al.  Systems pharmacology and genome medicine: a future perspective , 2009, Genome Medicine.

[38]  James R Faeder,et al.  Rule-based modeling of biochemical systems with BioNetGen. , 2009, Methods in molecular biology.

[39]  A. Ferrando,et al.  Gamma-secretase inhibitors reverse glucocorticoid resistance in T-ALL , 2008, Nature Medicine.

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

[41]  G. Augustine,et al.  A Positive Feedback Signal Transduction Loop Determines Timing of Cerebellar Long-Term Depression , 2008, Neuron.

[42]  Stephen S. Taylor,et al.  Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs. , 2008, Cancer cell.

[43]  DE Mager,et al.  Development of Translational Pharmacokinetic–Pharmacodynamic Models , 2008, Clinical pharmacology and therapeutics.

[44]  Marc R Gastonguay,et al.  Pharmacometrics: A Multidisciplinary Field to Facilitate Critical Thinking in Drug Development and Translational Research Settings , 2008, Journal of clinical pharmacology.

[45]  K. Basso,et al.  A systems biology approach to prediction of oncogenes and molecular perturbation targets in B-cell lymphomas , 2008, Molecular systems biology.

[46]  A. Barabasi,et al.  Drug—target network , 2007, Nature Biotechnology.

[47]  P. Donnelly,et al.  New models of collaboration in genome-wide association studies: the Genetic Association Information Network , 2007, Nature Genetics.

[48]  R. Lalonde,et al.  Introduction of Quantitative Methods in Pharmacology and Clinical Pharmacology: A Historical Overview , 2007, Clinical pharmacology and therapeutics.

[49]  W Ewy,et al.  Model‐based Drug Development , 2007, Clinical pharmacology and therapeutics.

[50]  Rajesh Krishna,et al.  Effective integration of systems biology, biomarkers, biosimulation and modelling in streamlining drug development. , 2007, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

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

[52]  D. Kell Systems biology, metabolic modelling and metabolomics in drug discovery and development. , 2006, Drug discovery today.

[53]  D. Lauffenburger,et al.  Physicochemical modelling of cell signalling pathways , 2006, Nature Cell Biology.

[54]  M. Meyerson,et al.  Genomic Approaches to Lung Cancer , 2006, Clinical Cancer Research.

[55]  Patrice Degoulet,et al.  Building an ontology of adverse drug reactions for automated signal generation in pharmacovigilance , 2006, Comput. Biol. Medicine.

[56]  G. V. Paolini,et al.  Global mapping of pharmacological space , 2006, Nature Biotechnology.

[57]  Timothy J Mitchison,et al.  Small molecule screening by imaging. , 2006, Current opinion in chemical biology.

[58]  Eric J Kunkel,et al.  Systems biology in drug discovery. , 2006, Conference proceedings : ... Annual International Conference of the IEEE Engineering in Medicine and Biology Society. IEEE Engineering in Medicine and Biology Society. Annual Conference.

[59]  B. Corrigan,et al.  How Modeling and Simulation Have Enhanced Decision Making in New Drug Development , 2005, Journal of Pharmacokinetics and Pharmacodynamics.

[60]  Garry Jennings,et al.  Finding improved medicines: the role of academic–industrial collaboration , 2005, Nature Reviews Drug Discovery.

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

[62]  Y. Lazebnik Can a biologist fix a radio? — or, what I learned while studying apoptosis , 2004, Biochemistry (Moscow).

[63]  W. Bialek,et al.  Physical limits to biochemical signaling. , 2003, Proceedings of the National Academy of Sciences of the United States of America.

[64]  David E Hill,et al.  Academia-industry collaboration: an integral element for building "omic" resources. , 2004, Genome research.

[65]  I. Kola,et al.  Can the pharmaceutical industry reduce attrition rates? , 2004, Nature Reviews Drug Discovery.

[66]  J. Paulsson Summing up the noise in gene networks , 2004, Nature.

[67]  Hartmut Derendorf,et al.  Modeling of Pharmacokinetic/Pharmacodynamic (PK/PD) Relationships: Concepts and Perspectives , 1999, Pharmaceutical Research.

[68]  D. L. Taylor,et al.  Advances in high content screening for drug discovery. , 2003, Assay and drug development technologies.

[69]  D. Noble Modeling the Heart--from Genes to Cells to the Whole Organ , 2002, Science.

[70]  E. Oren,et al.  Potential role of pharmacogenomics in reducing adverse drug reactions: a systematic review. , 2001, JAMA.

[71]  U. Bhalla,et al.  Emergent properties of networks of biological signaling pathways. , 1999, Science.

[72]  S. Shenolikar,et al.  Gating of CaMKII by cAMP-regulated protein phosphatase activity during LTP. , 1998, Science.

[73]  L B Sheiner,et al.  Learning versus confirming in clinical drug development , 1997, Clinical pharmacology and therapeutics.

[74]  C. Grosvenor,et al.  Role of dopamine in anterior pituitary tubulin pools in the lactating rat. , 1990, Neuroreport.

[75]  H. Berg,et al.  Physics of chemoreception. , 1977, Biophysical journal.

[76]  J. Black,et al.  Definition and Antagonism of Histamine H2-receptors , 1972, Nature.

[77]  T. G. Coleman,et al.  Circulation: overall regulation. , 1972, Annual review of physiology.

[78]  A. M. Lands,et al.  Differentiation of Receptor Systems activated by Sympathomimetic Amines , 1967, Nature.

[79]  A. Hodgkin,et al.  A quantitative description of membrane current and its application to conduction and excitation in nerve , 1952, The Journal of physiology.

[80]  R. Ahlquist,et al.  A study of the adrenotropic receptors. , 1948, The American journal of physiology.

[81]  G. Beadle,et al.  Genetic Control of Biochemical Reactions in Neurospora , 1941 .