1
|
Abrams RE, Pierre K, El-Murr N, Seung E, Wu L, Luna E, Mehta R, Li J, Larabi K, Ahmed M, Pelekanou V, Yang ZY, van de Velde H, Stamatelos SK. Quantitative systems pharmacology modeling sheds light into the dose response relationship of a trispecific T cell engager in multiple myeloma. Sci Rep 2022; 12:10976. [PMID: 35768621 PMCID: PMC9243109 DOI: 10.1038/s41598-022-14726-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 06/10/2022] [Indexed: 02/08/2023] Open
Abstract
In relapsed and refractory multiple myeloma (RRMM), there are few treatment options once patients progress from the established standard of care. Several bispecific T-cell engagers (TCE) are in clinical development for multiple myeloma (MM), designed to promote T-cell activation and tumor killing by binding a T-cell receptor and a myeloma target. In this study we employ both computational and experimental tools to investigate how a novel trispecific TCE improves activation, proliferation, and cytolytic activity of T-cells against MM cells. In addition to binding CD3 on T-cells and CD38 on tumor cells, the trispecific binds CD28, which serves as both co-stimulation for T-cell activation and an additional tumor target. We have established a robust rule-based quantitative systems pharmacology (QSP) model trained against T-cell activation, cytotoxicity, and cytokine data, and used it to gain insight into the complex dose response of this drug. We predict that CD3-CD28-CD38 killing capacity increases rapidly in low dose levels, and with higher doses, killing plateaus rather than following the bell-shaped curve typical of bispecific TCEs. We further predict that dose–response curves are driven by the ability of tumor cells to form synapses with activated T-cells. When competition between cells limits tumor engagement with active T-cells, response to therapy may be diminished. We finally suggest a metric related to drug efficacy in our analysis—“effective” receptor occupancy, or the proportion of receptors engaged in synapses. Overall, this study predicts that the CD28 arm on the trispecific antibody improves efficacy, and identifies metrics to inform potency of novel TCEs.
Collapse
Affiliation(s)
- R E Abrams
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA.,Daichi Sankyo, 211 Mt. Airy Rd., Basking Ridge, NJ, 07920, USA
| | - K Pierre
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA.
| | - N El-Murr
- Sanofi, 13 quai Jules Guesde 94403 Cedex, VITRY-SUR-SEINE, Vitry/Alfortville, France
| | - E Seung
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | - L Wu
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | | | | | - J Li
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA
| | - K Larabi
- Sanofi, 13 quai Jules Guesde 94403 Cedex, VITRY-SUR-SEINE, Vitry/Alfortville, France
| | - M Ahmed
- Sanofi, 50 Binney St., Cambridge, MA, 02142, USA
| | - V Pelekanou
- Sanofi, 50 Binney St., Cambridge, MA, 02142, USA.,Bayer Pharmaceuticals, Cambridge, MA, 02142, USA
| | - Z-Y Yang
- Sanofi, 270 Albany St., Cambridge, MA, 02139, USA.,Modex Therapeutics, 22 Strathmore Road, Natick, MA, 01760, USA
| | | | - S K Stamatelos
- Sanofi, 55 Corporate Dr, Bridgewater, NJ, 08807, USA. .,Bayer Pharmaceuticals, PH100 Bayer Boulevard, Whippany, NJ, 07981, USA.
| |
Collapse
|
2
|
Nanavati C, Mager DE. Network-Based Systems Analysis Explains Sequence-Dependent Synergism of Bortezomib and Vorinostat in Multiple Myeloma. AAPS JOURNAL 2021; 23:101. [PMID: 34403034 DOI: 10.1208/s12248-021-00622-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/05/2021] [Indexed: 12/22/2022]
Abstract
Bortezomib and vorinostat exhibit synergistic effects in multiple myeloma (MM) cells when given in sequence, and the purpose of this study was to evaluate the molecular determinants of the interaction using a systems pharmacology approach. A Boolean network model consisting of 79 proteins and 225 connections was developed using literature information characterizing mechanisms of drug action and intracellular signal transduction. Network visualization and structural analysis were conducted, and model simulations were compared with experimental data. Critical biomarkers, such as pNFκB, p53, cellular stress, and p21, were identified using measures of network centrality and model reduction. U266 cells were then exposed to bortezomib (3 nM) and vorinostat (2 μM) as single agents or in simultaneous and sequential (bortezomib for first 24 h, followed by addition of vorinostat for another 24 h) combinations. Temporal changes for nine of the critical proteins in the reduced Boolean model were measured over 48 h, and cellular proliferation was measured over 96 h. A mechanism-based systems model was developed that captured the biological basis of a bortezomib and vorinostat sequence-dependent pharmacodynamic interaction. The model was further extended in vivo by linking in vitro parameter values and dynamics of p21, caspase-3, and pAKT biomarkers to tumor growth in xenograft mice reported in the literature. Network-based methodologies and pharmacodynamic principles were integrated successfully to evaluate bortezomib and vorinostat interactions in a mechanistic and quantitative manner. The model can be potentially applied to evaluate their combination regimens and explore in vivo dosing regimens.
Collapse
Affiliation(s)
- Charvi Nanavati
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, 431 Pharmacy Building Buffalo, New York, 14214, USA
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, 431 Pharmacy Building Buffalo, New York, 14214, USA.
| |
Collapse
|
3
|
Zheng F, Xiao Y, Liu H, Fan Y, Dao M. Patient-Specific Organoid and Organ-on-a-Chip: 3D Cell-Culture Meets 3D Printing and Numerical Simulation. Adv Biol (Weinh) 2021; 5:e2000024. [PMID: 33856745 PMCID: PMC8243895 DOI: 10.1002/adbi.202000024] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/13/2021] [Indexed: 12/11/2022]
Abstract
The last few decades have witnessed diversified in vitro models to recapitulate the architecture and function of living organs or tissues and contribute immensely to advances in life science. Two novel 3D cell culture models: 1) Organoid, promoted mainly by the developments of stem cell biology and 2) Organ-on-a-chip, enhanced primarily due to microfluidic technology, have emerged as two promising approaches to advance the understanding of basic biological principles and clinical treatments. This review describes the comparable distinct differences between these two models and provides more insights into their complementarity and integration to recognize their merits and limitations for applicable fields. The convergence of the two approaches to produce multi-organoid-on-a-chip or human organoid-on-a-chip is emerging as a new approach for building 3D models with higher physiological relevance. Furthermore, rapid advancements in 3D printing and numerical simulations, which facilitate the design, manufacture, and results-translation of 3D cell culture models, can also serve as novel tools to promote the development and propagation of organoid and organ-on-a-chip systems. Current technological challenges and limitations, as well as expert recommendations and future solutions to address the promising combinations by incorporating organoids, organ-on-a-chip, 3D printing, and numerical simulation, are also summarized.
Collapse
Affiliation(s)
- Fuyin Zheng
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| | - Yuminghao Xiao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hui Liu
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Ming Dao
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Biological Sciences, Nanyang Technological University, Singapore, 639798, Singapore
| |
Collapse
|
4
|
Abstract
Making decisions on how best to treat cancer patients requires the integration of different data sets, including genomic profiles, tumour histopathology, radiological images, proteomic analysis and more. This wealth of biological information calls for novel strategies to integrate such information in a meaningful, predictive and experimentally verifiable way. In this Perspective we explain how executable computational models meet this need. Such models provide a means for comprehensive data integration, can be experimentally validated, are readily interpreted both biologically and clinically, and have the potential to predict effective therapies for different cancer types and subtypes. We explain what executable models are and how they can be used to represent the dynamic biological behaviours inherent in cancer, and demonstrate how such models, when coupled with automated reasoning, facilitate our understanding of the mechanisms by which oncogenic signalling pathways regulate tumours. We explore how executable models have impacted the field of cancer research and argue that extending them to represent a tumour in a specific patient (that is, an avatar) will pave the way for improved personalized treatments and precision medicine. Finally, we highlight some of the ongoing challenges in developing executable models and stress that effective cross-disciplinary efforts are key to forward progress in the field.
Collapse
Affiliation(s)
- Matthew A Clarke
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Jasmin Fisher
- Department of Biochemistry, University of Cambridge, Cambridge, UK.
- UCL Cancer Institute, University College London, London, UK.
| |
Collapse
|
5
|
van Hasselt JGC, Iyengar R. Systems Pharmacology: Defining the Interactions of Drug Combinations. Annu Rev Pharmacol Toxicol 2018; 59:21-40. [PMID: 30260737 DOI: 10.1146/annurev-pharmtox-010818-021511] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The majority of diseases are associated with alterations in multiple molecular pathways and complex interactions at the cellular and organ levels. Single-target monotherapies therefore have intrinsic limitations with respect to their maximum therapeutic benefits. The potential of combination drug therapies has received interest for the treatment of many diseases and is well established in some areas, such as oncology. Combination drug treatments may allow us to identify synergistic drug effects, reduce adverse drug reactions, and address variability in disease characteristics between patients. Identification of combination therapies remains challenging. We discuss current state-of-the-art systems pharmacology approaches to enable rational identification of combination therapies. These approaches, which include characterization of mechanisms of disease and drug action at a systems level, can enable understanding of drug interactions at the molecular, cellular, physiological, and organismal levels. Such multiscale understanding can enable precision medicine by promoting the rational development of combination therapy at the level of individual patients for many diseases.
Collapse
Affiliation(s)
- J G Coen van Hasselt
- Department of Pharmacological Sciences, Systems Biology Center, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; .,Division of Systems Biomedicine and Pharmacology, Leiden Academic Center for Drug Research, Leiden University, 2333 Leiden, Netherlands;
| | - Ravi Iyengar
- Department of Pharmacological Sciences, Systems Biology Center, Mount Sinai Institute for Systems Biomedicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| |
Collapse
|
6
|
Ramakrishnan V, Mager DE. Network-Based Analysis of Bortezomib Pharmacodynamic Heterogeneity in Multiple Myeloma Cells. J Pharmacol Exp Ther 2018; 365:734-751. [PMID: 29632237 PMCID: PMC5959840 DOI: 10.1124/jpet.118.247924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 04/05/2018] [Indexed: 12/19/2022] Open
Abstract
The objective of this study is to evaluate the heterogeneity in pharmacodynamic response in four in vitro multiple myeloma cell lines to treatment with bortezomib, and to assess whether such differences are associated with drug-induced intracellular signaling protein dynamics identified via a logic-based network modeling approach. The in vitro pharmacodynamic-efficacy of bortezomib was evaluated through concentration-effect and cell proliferation dynamical studies in U266, RPMI8226, MM.1S, and NCI-H929 myeloma cell lines. A Boolean logic-based network model incorporating intracellular protein signaling pathways relevant to myeloma cell growth, proliferation, and apoptosis was developed based on information available in the literature and used to identify key proteins regulating bortezomib pharmacodynamics. The time-course of network-identified proteins was measured using the MAGPIX protein assay system. Traditional pharmacodynamic modeling endpoints revealed variable responses of the cell lines to bortezomib treatment, classifying cell lines as more sensitive (MM.1S and NCI-H929) and less sensitive (U266 and RPMI8226). Network centrality and model reduction identified key proteins (e.g., phosphorylated nuclear factor-κB, phosphorylated protein kinase B, phosphorylated mechanistic target of rapamycin, Bcl-2, phosphorylated c-Jun N-terminal kinase, phosphorylated p53, p21, phosphorylated Bcl-2-associated death promoter, caspase 8, and caspase 9) that govern bortezomib pharmacodynamics. The corresponding relative expression (normalized to 0-hour untreated-control cells) of proteins demonstrated a greater magnitude and earlier onset of stimulation/inhibition in cells more sensitive (MM.1S and NCI-H929) to bortezomib-induced cell death at 20 nM, relative to the less sensitive cells (U266 and RPMI8226). Overall, differences in intracellular signaling appear to be associated with bortezomib pharmacodynamic heterogeneity, and key proteins may be potential biomarkers to evaluate bortezomib responses.
Collapse
Affiliation(s)
- Vidya Ramakrishnan
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, New York
| | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, New York
| |
Collapse
|
7
|
Traynard P, Tobalina L, Eduati F, Calzone L, Saez-Rodriguez J. Logic Modeling in Quantitative Systems Pharmacology. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:499-511. [PMID: 28681552 PMCID: PMC5572374 DOI: 10.1002/psp4.12225] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/01/2017] [Accepted: 06/15/2017] [Indexed: 12/12/2022]
Abstract
Here we present logic modeling as an approach to understand deregulation of signal transduction in disease and to characterize a drug's mode of action. We discuss how to build a logic model from the literature and experimental data and how to analyze the resulting model to obtain insights of relevance for systems pharmacology. Our workflow uses the free tools OmniPath (network reconstruction from the literature), CellNOpt (model fit to experimental data), MaBoSS (model analysis), and Cytoscape (visualization).
Collapse
Affiliation(s)
- Pauline Traynard
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - Luis Tobalina
- RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, Germany
| | - Federica Eduati
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Laurence Calzone
- Institut Curie, PSL Research University, Mines Paris Tech, Inserm, U900, Paris, France
| | - Julio Saez-Rodriguez
- RWTH Aachen University, Faculty of Medicine, Joint Research Centre for Computational Biomedicine, Aachen, Germany.,European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, UK
| |
Collapse
|
8
|
Kirouac DC, Schaefer G, Chan J, Merchant M, Orr C, Huang SMA, Moffat J, Liu L, Gadkar K, Ramanujan S. Clinical responses to ERK inhibition in BRAFV600E-mutant colorectal cancer predicted using a computational model. NPJ Syst Biol Appl 2017; 3:14. [PMID: 28649441 PMCID: PMC5460205 DOI: 10.1038/s41540-017-0016-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/18/2017] [Accepted: 05/04/2017] [Indexed: 12/11/2022] Open
Abstract
Approximately 10% of colorectal cancers harbor BRAFV600E mutations, which constitutively activate the MAPK signaling pathway. We sought to determine whether ERK inhibitor (GDC-0994)-containing regimens may be of clinical benefit to these patients based on data from in vitro (cell line) and in vivo (cell- and patient-derived xenograft) studies of cetuximab (EGFR), vemurafenib (BRAF), cobimetinib (MEK), and GDC-0994 (ERK) combinations. Preclinical data was used to develop a mechanism-based computational model linking cell surface receptor (EGFR) activation, the MAPK signaling pathway, and tumor growth. Clinical predictions of anti-tumor activity were enabled by the use of tumor response data from three Phase 1 clinical trials testing combinations of EGFR, BRAF, and MEK inhibitors. Simulated responses to GDC-0994 monotherapy (overall response rate = 17%) accurately predicted results from a Phase 1 clinical trial regarding the number of responding patients (2/18) and the distribution of tumor size changes ("waterfall plot"). Prospective simulations were then used to evaluate potential drug combinations and predictive biomarkers for increasing responsiveness to MEK/ERK inhibitors in these patients.
Collapse
Affiliation(s)
- Daniel C. Kirouac
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Gabriele Schaefer
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Jocelyn Chan
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Mark Merchant
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Christine Orr
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Shih-Min A. Huang
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - John Moffat
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Lichuan Liu
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Kapil Gadkar
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| | - Saroja Ramanujan
- Genentech Research & Early Development, 1 DNA Way, South San Francisco, CA 94080 USA
| |
Collapse
|
9
|
Eduati F, Doldàn-Martelli V, Klinger B, Cokelaer T, Sieber A, Kogera F, Dorel M, Garnett MJ, Blüthgen N, Saez-Rodriguez J. Drug Resistance Mechanisms in Colorectal Cancer Dissected with Cell Type-Specific Dynamic Logic Models. Cancer Res 2017; 77:3364-3375. [PMID: 28381545 DOI: 10.1158/0008-5472.can-17-0078] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/17/2017] [Accepted: 03/31/2017] [Indexed: 12/20/2022]
Abstract
Genomic features are used as biomarkers of sensitivity to kinase inhibitors used widely to treat human cancer, but effective patient stratification based on these principles remains limited in impact. Insofar as kinase inhibitors interfere with signaling dynamics, and, in turn, signaling dynamics affects inhibitor responses, we investigated associations in this study between cell-specific dynamic signaling pathways and drug sensitivity. Specifically, we measured 14 phosphoproteins under 43 different perturbed conditions (combinations of 5 stimuli and 7 inhibitors) in 14 colorectal cancer cell lines, building cell line-specific dynamic logic models of underlying signaling networks. Model parameters representing pathway dynamics were used as features to predict sensitivity to a panel of 27 drugs. Specific parameters of signaling dynamics correlated strongly with drug sensitivity for 14 of the drugs, 9 of which had no genomic biomarker. Following one of these associations, we validated a drug combination predicted to overcome resistance to MEK inhibitors by coblockade of GSK3, which was not found based on associations with genomic data. These results suggest that to better understand the cancer resistance and move toward personalized medicine, it is essential to consider signaling network dynamics that cannot be inferred from static genotypes. Cancer Res; 77(12); 3364-75. ©2017 AACR.
Collapse
Affiliation(s)
- Federica Eduati
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Victoria Doldàn-Martelli
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, United Kingdom.,Departamento de Física de la Materia Condensada, Condensed Matter Physics Center (IFIMAC) and Instituto Nicolás Cabrera, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Bertram Klinger
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Integrative Research Institute (IRI) Life Sciences and Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Thomas Cokelaer
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, United Kingdom
| | - Anja Sieber
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Integrative Research Institute (IRI) Life Sciences and Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fiona Kogera
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Mathurin Dorel
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Integrative Research Institute (IRI) Life Sciences and Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Mathew J Garnett
- Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, United Kingdom
| | - Nils Blüthgen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany. .,Integrative Research Institute (IRI) Life Sciences and Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Julio Saez-Rodriguez
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Trust Genome Campus, Hinxton, United Kingdom. .,Joint Research Centre for Computational Biomedicine (JRC-COMBINE), RWTH Aachen University, Faculty of Medicine, Aachen, Germany
| |
Collapse
|
10
|
Mistry HB, Fabre MA, Young J, Clack G, Dickinson PA. Systems Pharmacology Modeling of Prostate-Specific Antigen in Patients With Prostate Cancer Treated With an Androgen Receptor Antagonist and Down-Regulator. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2016; 5:258-63. [PMID: 27299938 PMCID: PMC4879474 DOI: 10.1002/psp4.12066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 01/29/2016] [Indexed: 11/29/2022]
Abstract
First‐in‐human (FIH) studies with AZD3514, a selective androgen receptor (AR) down‐regulator, showed decreases of >30% in the prostate‐specific antigen (PSA) in some patients. A modeling approach was adopted to understand these observations and define the optimum clinical use hypothesis for AZD3514 for clinical testing. Initial empirical modeling showed that only baseline PSA correlated significantly with this biological response, whereas drug concentration did not. To identify the mechanistic cause of this observation, a mechanism‐based model was first developed, which described the effects of AZD3514 on AR protein and PSA mRNA levels in LNCaP cells with and without dihydrotestosterone (DHT). Second, the mechanism‐based model was linked to a population pharmacokinetic (PK) model; PSA effects of clinical doses were subsequently simulated under different clinical conditions. This model was used to adjust the design of the ongoing clinical FIH study and direct the backup program.
Collapse
Affiliation(s)
- H B Mistry
- Manchester Pharmacy School, The University of Manchester, UK
| | - M-A Fabre
- Quantitiative Clinical Pharmacology, AstraZeneca, Alderley Park, UK
| | - J Young
- Goosebrook Associates Ltd, The BioHub at Alderley Park Alderley Edge, UK
| | - G Clack
- Early Clinical Development, AstraZeneca, Alderley Park, UK
| | - P A Dickinson
- Seda Pharmaceutical Development Services, The BioHub at Alderley Park Alderley Edge, UK
| |
Collapse
|
11
|
Kirouac DC, Du J, Lahdenranta J, Onsum MD, Nielsen UB, Schoeberl B, McDonagh CF. HER2+ Cancer Cell Dependence on PI3K vs. MAPK Signaling Axes Is Determined by Expression of EGFR, ERBB3 and CDKN1B. PLoS Comput Biol 2016; 12:e1004827. [PMID: 27035903 PMCID: PMC4818107 DOI: 10.1371/journal.pcbi.1004827] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/22/2016] [Indexed: 12/24/2022] Open
Abstract
Understanding the molecular pathways by which oncogenes drive cancerous cell growth, and how dependence on such pathways varies between tumors could be highly valuable for the design of anti-cancer treatment strategies. In this work we study how dependence upon the canonical PI3K and MAPK cascades varies across HER2+ cancers, and define biomarkers predictive of pathway dependencies. A panel of 18 HER2+ (ERBB2-amplified) cell lines representing a variety of indications was used to characterize the functional and molecular diversity within this oncogene-defined cancer. PI3K and MAPK-pathway dependencies were quantified by measuring in vitro cell growth responses to combinations of AKT (MK2206) and MEK (GSK1120212; trametinib) inhibitors, in the presence and absence of the ERBB3 ligand heregulin (NRG1). A combination of three protein measurements comprising the receptors EGFR, ERBB3 (HER3), and the cyclin-dependent kinase inhibitor p27 (CDKN1B) was found to accurately predict dependence on PI3K/AKT vs. MAPK/ERK signaling axes. Notably, this multivariate classifier outperformed the more intuitive and clinically employed metrics, such as expression of phospho-AKT and phospho-ERK, and PI3K pathway mutations (PIK3CA, PTEN, and PIK3R1). In both cell lines and primary patient samples, we observed consistent expression patterns of these biomarkers varies by cancer indication, such that ERBB3 and CDKN1B expression are relatively high in breast tumors while EGFR expression is relatively high in other indications. The predictability of the three protein biomarkers for differentiating PI3K/AKT vs. MAPK dependence in HER2+ cancers was confirmed using external datasets (Project Achilles and GDSC), again out-performing clinically used genetic markers. Measurement of this minimal set of three protein biomarkers could thus inform treatment, and predict mechanisms of drug resistance in HER2+ cancers. More generally, our results show a single oncogenic transformation can have differing effects on cell signaling and growth, contingent upon the molecular and cellular context. Biomarkers capable of accurately predicting patient responses to alternate therapies are critical to realizing the vision of precision medicine. Identifying such biomarkers is, however, challenging due to the inherent complexity of biological networks. Here we sought to identify molecular features that predict how a genetically defined subset of cancers (HER2+) differentially depend on two oncogenic signaling pathways, the PI3K/AKT and MAPK/ERK cascades. We find that combined measurement of three non-intuitive proteins (EGFR, ERBB3, and CDKN1B) accurately predicts cellular dependence on these signaling pathways, and responsiveness to drugs targeting their constituents. Notably, this three-biomarker model outperformed both biological intuition (phosho-AKT and phospho-ERK) and current clinical practice (PIK3CA mutations). More broadly, this exemplifies how the functional consequences of a single oncogenic driver (HER2) can depend upon molecular and cellular context. Expression of these markers also varies by indication, such that breast cancers are biased toward PI3K-dependnece, while non-breast indications (lung, ovarian, and gastric) are particularly MAPK-dependent, and thus may respond differently to therapeutic strategies developed for breast cancer. Together, we believe that our results will aid the design of novel, stratified treatment strategies for HER2+ disease.
Collapse
Affiliation(s)
- Daniel C Kirouac
- Discovery, Merrimack Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Jinyan Du
- Discovery, Merrimack Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Johanna Lahdenranta
- Discovery, Merrimack Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Matthew D Onsum
- Discovery, Merrimack Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Ulrik B Nielsen
- Discovery, Merrimack Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Birgit Schoeberl
- Discovery, Merrimack Pharmaceuticals, Cambridge, Massachusetts, United States of America
| | - Charlotte F McDonagh
- Discovery, Merrimack Pharmaceuticals, Cambridge, Massachusetts, United States of America
| |
Collapse
|
12
|
Gadkar K, Lu J, Sahasranaman S, Davis J, Mazer NA, Ramanujan S. Evaluation of HDL-modulating interventions for cardiovascular risk reduction using a systems pharmacology approach. J Lipid Res 2015; 57:46-55. [PMID: 26522778 PMCID: PMC4689335 DOI: 10.1194/jlr.m057943] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Indexed: 11/20/2022] Open
Abstract
The recent failures of cholesteryl ester transport protein inhibitor drugs to decrease CVD risk, despite raising HDL cholesterol (HDL-C) levels, suggest that pharmacologic increases in HDL-C may not always reflect elevations in reverse cholesterol transport (RCT), the process by which HDL is believed to exert its beneficial effects. HDL-modulating therapies can affect HDL properties beyond total HDL-C, including particle numbers, size, and composition, and may contribute differently to RCT and CVD risk. The lack of validated easily measurable pharmacodynamic markers to link drug effects to RCT, and ultimately to CVD risk, complicates target and compound selection and evaluation. In this work, we use a systems pharmacology model to contextualize the roles of different HDL targets in cholesterol metabolism and provide quantitative links between HDL-related measurements and the associated changes in RCT rate to support target and compound evaluation in drug development. By quantifying the amount of cholesterol removed from the periphery over the short-term, our simulations show the potential for infused HDL to treat acute CVD. For the primary prevention of CVD, our analysis suggests that the induction of ApoA-I synthesis may be a more viable approach, due to the long-term increase in RCT rate.
Collapse
Affiliation(s)
- Kapil Gadkar
- Genentech Research and Early Development, South San Francisco, CA
| | - James Lu
- Roche Pharma Research and Early Development, Clinical Pharmacology, Disease Modeling Group, Roche Innovation Center Basel, Basel, Switzerland
| | | | - John Davis
- Genentech Research and Early Development, South San Francisco, CA
| | - Norman A Mazer
- Roche Pharma Research and Early Development, Clinical Pharmacology, Disease Modeling Group, Roche Innovation Center Basel, Basel, Switzerland
| | - Saroja Ramanujan
- Genentech Research and Early Development, South San Francisco, CA
| |
Collapse
|
13
|
Oxygen-Driven Tumour Growth Model: A Pathology-Relevant Mathematical Approach. PLoS Comput Biol 2015; 11:e1004550. [PMID: 26517813 PMCID: PMC4627780 DOI: 10.1371/journal.pcbi.1004550] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 09/12/2015] [Indexed: 02/04/2023] Open
Abstract
Xenografts -as simplified animal models of cancer- differ substantially in vasculature and stromal architecture when compared to clinical tumours. This makes mathematical model-based predictions of clinical outcome challenging. Our objective is to further understand differences in tumour progression and physiology between animal models and the clinic. To achieve that, we propose a mathematical model based upon tumour pathophysiology, where oxygen -as a surrogate for endocrine delivery- is our main focus. The Oxygen-Driven Model (ODM), using oxygen diffusion equations, describes tumour growth, hypoxia and necrosis. The ODM describes two key physiological parameters. Apparent oxygen uptake rate ( kR′) represents the amount of oxygen cells seem to need to proliferate. The more oxygen they appear to need, the more the oxygen transport. kR′ gathers variability from the vasculature, stroma and tumour morphology. Proliferating rate (kp) deals with cell line specific factors to promote growth. The KH,KN describe the switch of hypoxia and necrosis. Retrospectively, using archived data, we looked at longitudinal tumour volume datasets for 38 xenografted cell lines and 5 patient-derived xenograft-like models. Exploration of the parameter space allows us to distinguish 2 groups of parameters. Group 1 of cell lines shows a spread in values of kR′ and lower kp, indicating that tumours are poorly perfused and slow growing. Group 2 share the value of the oxygen uptake rate ( kR′) and vary greatly in kp, which we interpret as having similar oxygen transport, but more tumour intrinsic variability in growth. However, the ODM has some limitations when tested in explant-like animal models, whose complex tumour-stromal morphology may not be captured in the current version of the model. Incorporation of stroma in the ODM will help explain these discrepancies. We have provided an example. The ODM is a very simple -and versatile- model suitable for the design of preclinical experiments, which can be modified and enhanced whilst maintaining confidence in its predictions. Tumour-bearing animal models of cancer are needed to discover new drugs to treat cancer. We aim in this article to capture—through mathematics- some underlying phenomena of tumour growth in animals. We propose a set of equations that, despite being very simple, describe tumour growth, hypoxia and necrosis. Cells under low oxygen levels change into a stress state called “hypoxia”, which will ultimately lead to tissue death, also known as “necrosis” and “apoptosis”. Hypoxic cells undergo a variety of changes which impact tumour growth, development, metastasis and -most importantly- response to therapy. Hence, oxygen distribution is important. We simulate oxygen profiles to locate hypoxic and necrotic tumour regions. Finally, this mathematical model allows us to compare and classify animal models from a grounded and physiological perspective, rather than a more convenient and empirical one. This will help us understand how well (or poorly) animal tumours mimic tumours in patients. The simplicity of our mathematical model allows us to obtain more information out of the same animal sets without any further experiments, hopefully saving time, money and animal usage.
Collapse
|
14
|
Stokes CL, Cirit M, Lauffenburger DA. Physiome-on-a-Chip: The Challenge of "Scaling" in Design, Operation, and Translation of Microphysiological Systems. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015; 4:559-62. [PMID: 26535154 PMCID: PMC4625858 DOI: 10.1002/psp4.12042] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 09/11/2015] [Indexed: 12/22/2022]
Abstract
Scaling of a microphysiological system (MPS) or physiome-on-a-chip is arguably two interrelated, modeling-based activities: on-platform scaling and in vitro-in vivo translation. This dual approach reduces the need to perfectly rescale and mimic in vivo physiology, an aspiration that is both extremely challenging and not substantively meaningful because of uncertain relevance of any specific physiological condition. Accordingly, this perspective offers a tractable approach for designing interacting MPSs and relating in vitro results to analogous context in vivo.
Collapse
Affiliation(s)
- C L Stokes
- Stokes Consulting Redwood City, California, USA
| | - M Cirit
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| | - D A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology Cambridge, Massachusetts, USA
| |
Collapse
|
15
|
Morra R, Shankar J, Robinson CJ, Halliwell S, Butler L, Upton M, Hay S, Micklefield J, Dixon N. Dual transcriptional-translational cascade permits cellular level tuneable expression control. Nucleic Acids Res 2015; 44:e21. [PMID: 26405200 PMCID: PMC4756846 DOI: 10.1093/nar/gkv912] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 08/28/2015] [Indexed: 12/03/2022] Open
Abstract
The ability to induce gene expression in a small molecule dependent manner has led to many applications in target discovery, functional elucidation and bio-production. To date these applications have relied on a limited set of protein-based control mechanisms operating at the level of transcription initiation. The discovery, design and reengineering of riboswitches offer an alternative means by which to control gene expression. Here we report the development and characterization of a novel tunable recombinant expression system, termed RiboTite, which operates at both the transcriptional and translational level. Using standard inducible promoters and orthogonal riboswitches, a multi-layered modular genetic control circuit was developed to control the expression of both bacteriophage T7 RNA polymerase and recombinant gene(s) of interest. The system was benchmarked against a number of commonly used E. coli expression systems, and shows tight basal control, precise analogue tunability of gene expression at the cellular level, dose-dependent regulation of protein production rates over extended growth periods and enhanced cell viability. This novel system expands the number of E. coli expression systems for use in recombinant protein production and represents a major performance enhancement over and above the most widely used expression systems.
Collapse
Affiliation(s)
- Rosa Morra
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK Faculty of Life Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Jayendra Shankar
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK Faculty of Life Sciences, University of Manchester, Manchester, M13 9PL, UK School of Chemistry, University of Manchester, Manchester, M13 9PL, UK
| | - Christopher J Robinson
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK School of Chemistry, University of Manchester, Manchester, M13 9PL, UK
| | - Samantha Halliwell
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK Faculty of Life Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Lisa Butler
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK Faculty of Life Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Mathew Upton
- School of Biomedical & Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth, PL4 8AA, UK
| | - Sam Hay
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK Faculty of Life Sciences, University of Manchester, Manchester, M13 9PL, UK
| | - Jason Micklefield
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK School of Chemistry, University of Manchester, Manchester, M13 9PL, UK SYNBIOCHEM, University of Manchester, Manchester, M13 9PL, UK
| | - Neil Dixon
- Manchester Institute of Biotechnology, University of Manchester, Manchester, M13 9PL, UK Faculty of Life Sciences, University of Manchester, Manchester, M13 9PL, UK School of Chemistry, University of Manchester, Manchester, M13 9PL, UK SYNBIOCHEM, University of Manchester, Manchester, M13 9PL, UK
| |
Collapse
|
16
|
Benson N. Network-based discovery through mechanistic systems biology. Implications for applications--SMEs and drug discovery: where the action is. DRUG DISCOVERY TODAY. TECHNOLOGIES 2015; 15:41-8. [PMID: 26464089 DOI: 10.1016/j.ddtec.2015.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 06/30/2015] [Accepted: 07/14/2015] [Indexed: 01/10/2023]
Abstract
Phase II attrition remains the most important challenge for drug discovery. Tackling the problem requires improved understanding of the complexity of disease biology. Systems biology approaches to this problem can, in principle, deliver this. This article reviews the reports of the application of mechanistic systems models to drug discovery questions and discusses the added value. Although we are on the journey to the virtual human, the length, path and rate of learning from this remain an open question. Success will be dependent on the will to invest and make the most of the insight generated along the way.
Collapse
Affiliation(s)
- Neil Benson
- Xenologiq Ltd., Unit 43, Canterbury Innovation Centre, University Road, Canterbury CT2 7FG, UK.
| |
Collapse
|
17
|
Kirouac DC, Lahdenranta J, Du J, Yarar D, Onsum MD, Nielsen UB, McDonagh CF. Model-Based Design of a Decision Tree for Treating HER2+ Cancers Based on Genetic and Protein Biomarkers. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2015. [PMID: 26225238 PMCID: PMC4394616 DOI: 10.1002/psp4.19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human cancers are incredibly diverse with regard to molecular aberrations, dependence on oncogenic signaling pathways, and responses to pharmacological intervention. We wished to assess how cellular dependence on the canonical PI3K vs. MAPK pathways within HER2+ cancers affects responses to combinations of targeted therapies, and biomarkers predictive of their activity. Through an integrative analysis of mechanistic model simulations and in vitro cell line profiling, we designed a six-arm decision tree to stratify treatment of HER2+ cancers using combinations of targeted agents. Activating mutations in the PI3K and MAPK pathways (PIK3CA and KRAS), and expression of the HER3 ligand heregulin determined sensitivity to combinations of inhibitors against HER2 (lapatinib), HER3 (MM-111), AKT (MK-2206), and MEK (GSK-1120212; trametinib), in addition to the standard of care trastuzumab (Herceptin). The strategy used to identify effective combinations and predictive biomarkers in HER2-expressing tumors may be more broadly extendable to other human cancers.
Collapse
Affiliation(s)
- D C Kirouac
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - J Lahdenranta
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - J Du
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - D Yarar
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - M D Onsum
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - U B Nielsen
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| | - C F McDonagh
- Merrimack Pharmaceuticals Cambridge, Massachusetts, USA
| |
Collapse
|
18
|
Sharan S, Woo S. Systems pharmacology approaches for optimization of antiangiogenic therapies: challenges and opportunities. Front Pharmacol 2015; 6:33. [PMID: 25750626 PMCID: PMC4335258 DOI: 10.3389/fphar.2015.00033] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 02/06/2015] [Indexed: 12/16/2022] Open
Abstract
Targeted therapies have become an important therapeutic paradigm for multiple malignancies. The rapid development of resistance to these therapies impedes the successful management of advanced cancer. Due to the redundancy in angiogenic signaling, alternative proangiogenic factors are activated upon treatment with anti-VEGF agents. Higher doses of the agents lead to greater stimulation of compensatory proangiogenic pathways that limit the therapeutic efficacy of VEGF-targeted drugs and produce escape mechanisms for tumor. Evidence suggests that dose intensity and schedules affect the dynamics of the development of this resistance. Thus, an optimal dosing regimen is crucial to maximizing the therapeutic benefit of antiangiogenic agents and limiting treatment resistance. A systems pharmacology approach using multiscale computational modeling can facilitate a mechanistic understanding of these dynamics of angiogenic biomarkers and their impacts on tumor reduction and resistance. Herein, we discuss a systems pharmacology approach integrating the biology of VEGF-targeted therapy resistance, including circulating biomarkers, and pharmacodynamics to enable the optimization of antiangiogenic therapy for therapeutic gains.
Collapse
Affiliation(s)
- Satish Sharan
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center , Oklahoma City, OK, USA
| |
Collapse
|
19
|
Visser SAG, de Alwis DP, Kerbusch T, Stone JA, Allerheiligen SRB. Implementation of quantitative and systems pharmacology in large pharma. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e142. [PMID: 25338195 PMCID: PMC4474169 DOI: 10.1038/psp.2014.40] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 07/30/2014] [Indexed: 02/04/2023]
Abstract
Quantitative and systems pharmacology concepts and tools are the foundation of the model-informed drug development paradigm at Merck for integrating knowledge, enabling decisions, and enhancing submissions. Rigorous prioritization of modeling and simulation activities has enabled key drug development decisions and led to a high return on investment through significant cost avoidance. Critical factors for the successful implementation, examples on impact on decision making with associated return of investment, and drivers for continued success are discussed.
Collapse
Affiliation(s)
- S A G Visser
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| | - D P de Alwis
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| | - T Kerbusch
- Quantitive Pharmacology and Pharmacometrics, MSD, Oss, The Netherlands
| | - J A Stone
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| | - S R B Allerheiligen
- Quantitative Pharmacology and Pharmacometrics, Merck Research Labs, Merck & Co, Rahway, New Jersey, USA
| |
Collapse
|
20
|
Dominietto M, Tsinoremas N, Capobianco E. Integrative analysis of cancer imaging readouts by networks. Mol Oncol 2014; 9:1-16. [PMID: 25263240 PMCID: PMC5528685 DOI: 10.1016/j.molonc.2014.08.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 02/01/2023] Open
Abstract
Cancer is a multifactorial and heterogeneous disease. The corresponding complexity appears at multiple levels: from the molecular and the cellular constitution to the macroscopic phenotype, and at the diagnostic and therapeutic management stages. The overall complexity can be approximated to a certain extent, e.g. characterized by a set of quantitative phenotypic observables recorded in time‐space resolved dimensions by using multimodal imaging approaches. The transition from measures to data can be made effective through various computational inference methods, including networks, which are inherently capable of mapping variables and data to node‐ and/or edge‐valued topological properties, dynamic modularity configurations, and functional motifs. We illustrate how networks can integrate imaging data to explain cancer complexity, and assess potential pre‐clinical and clinical impact. Computational Multiplexing Imaging merges imaging and networks. Networks show signatures of tumor heterogeneity and phenotypic profiles observed in‐vivo. A profile ensemble establishes a tumor fingerprint, and this constitutes a novel type of marker. Personalized treatment is embedded in a systems medicine approach.
Collapse
Affiliation(s)
- Marco Dominietto
- Biomaterial Science Center, University of Basel, Basel, Switzerland; Institute for Biomedical Engineering, ETH and University of Zurich, Zurich, Switzerland
| | | | - Enrico Capobianco
- Center for Computational Science, University of Miami, Miami, FL, USA; Laboratory of Integrative Systems Medicine, Institute of Clinical Physiology, CNR, Pisa, Italy.
| |
Collapse
|
21
|
Hu QN, Deng Z, Tu W, Yang X, Meng ZB, Deng ZX, Liu J. VNP: Interactive Visual Network Pharmacology of Diseases, Targets, and Drugs. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2014; 3:e105. [PMID: 24622768 PMCID: PMC4039393 DOI: 10.1038/psp.2014.1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/28/2013] [Indexed: 02/04/2023]
Abstract
In drug discovery, promiscuous targets, multifactorial diseases, and "dirty" drugs construct complex network relationships. Network pharmacology description and analysis not only give a systems-level understanding of drug action and disease complexity but can also help to improve the efficiency of target selection and drug design. Visual network pharmacology (VNP) is developed to visualize network pharmacology of targets, diseases, and drugs with a graph network by using disease, target or drug names, chemical structures, or protein sequence. To our knowledge, VNP is the first free interactive VNP server that should be very helpful for systems pharmacology research. VNP is freely available at http://cadd.whu.edu.cn/ditad/vnpsearch.
Collapse
Affiliation(s)
- Q-N Hu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
| | - Z Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
| | - W Tu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
| | - X Yang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
| | - Z-B Meng
- State Key Laboratory of Software Engineering (Wuhan University) and Wuhan University School of Computer Science, Wuhan, P. R. China
| | - Z-X Deng
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (Wuhan University), Ministry of Education, and Wuhan University School of Pharmaceutical Sciences, Wuhan, P. R. China
| | - J Liu
- State Key Laboratory of Software Engineering (Wuhan University) and Wuhan University School of Computer Science, Wuhan, P. R. China
| |
Collapse
|
22
|
Birtwistle MR, Mager DE, Gallo JM. Mechanistic vs. Empirical network models of drug action. CPT Pharmacometrics Syst Pharmacol 2013; 2:e72. [PMID: 24448020 PMCID: PMC4026635 DOI: 10.1038/psp.2013.51] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Declining success rates coupled with increased costs is leading to an inevitable breaking point in the drug development pipeline. Can we avoid it by incorporating the vast mechanistic understanding of drug action? A recent review highlights this dilemma and proposes "quantitative logic gate" modeling as a solution.(1) The goal of this commentary is to contrast this approach with mechanistic biochemical network models, which, although alluded to by Kiruoac and Onsum, requires a closer analysis.
Collapse
Affiliation(s)
- M R Birtwistle
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - D E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, SUNY, Buffalo, New York, USA
| | - J M Gallo
- Department of Pharmacology and Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|