151
|
Computer-aided drug repurposing for cancer therapy: Approaches and opportunities to challenge anticancer targets. Semin Cancer Biol 2019; 68:59-74. [PMID: 31562957 DOI: 10.1016/j.semcancer.2019.09.023] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 12/14/2022]
Abstract
Despite huge efforts made in academic and pharmaceutical worldwide research, current anticancer therapies achieve effective treatment in a limited number of neoplasia cases only. Oncology terms such as big killers - to identify tumours with yet a high mortality rate - or undruggable cancer targets, and chemoresistance, represent the current therapeutic debacle of cancer treatments. In addition, metastases, tumour microenvironments, tumour heterogeneity, metabolic adaptations, and immunotherapy resistance are essential features controlling tumour response to therapies, but still, lack effective therapeutics or modulators. In this scenario, where the pharmaceutical productivity and drug efficacy in oncology seem to have reached a plateau, the so-called drug repurposing - i.e. the use of old drugs, already in clinical use, for a different therapeutic indication - is an appealing strategy to improve cancer therapy. Opportunities for drug repurposing are often based on occasional observations or on time-consuming pre-clinical drug screenings that are often not hypothesis-driven. In contrast, in-silico drug repurposing is an emerging, hypothesis-driven approach that takes advantage of the use of big-data. Indeed, the extensive use of -omics technologies, improved data storage, data meaning, machine learning algorithms, and computational modeling all offer unprecedented knowledge of the biological mechanisms of cancers and drugs' modes of action, providing extensive availability for both disease-related data and drugs-related data. This offers the opportunity to generate, with time and cost-effective approaches, computational drug networks to predict, in-silico, the efficacy of approved drugs against relevant cancer targets, as well as to select better responder patients or disease' biomarkers. Here, we will review selected disease-related data together with computational tools to be exploited for the in-silico repurposing of drugs against validated targets in cancer therapies, focusing on the oncogenic signaling pathways activation in cancer. We will discuss how in-silico drug repurposing has the promise to shortly improve our arsenal of anticancer drugs and, likely, overcome certain limitations of modern cancer therapies against old and new therapeutic targets in oncology.
Collapse
|
152
|
Failli M, Paananen J, Fortino V. Prioritizing target-disease associations with novel safety and efficacy scoring methods. Sci Rep 2019; 9:9852. [PMID: 31285471 PMCID: PMC6614395 DOI: 10.1038/s41598-019-46293-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/25/2019] [Indexed: 01/24/2023] Open
Abstract
Biological target (commonly genes or proteins) identification is still largely a manual process, where experts manually try to collect and combine information from hundreds of data sources, ranging from scientific publications to omics databases. Targeting the wrong gene or protein will lead to failure of the drug development process, as well as incur delays and costs. To improve this process, different software platforms are being developed. These platforms rely strongly on efficacy estimates based on target-disease association scores created by computational methods for drug target prioritization. Here novel computational methods are presented to more accurately evaluate the efficacy and safety of potential drug targets. The proposed efficacy scores utilize existing gene expression data and tissue/disease specific networks to improve the inference of target-disease associations. Conversely, safety scores enable the identification of genes that are essential, potentially susceptible to adverse effects or carcinogenic. Benchmark results demonstrate that our transcriptome-based methods for drug target prioritization can increase the true positive rate of target-disease associations. Additionally, the proposed safety evaluation system enables accurate predictions of targets of withdrawn drugs and targets of drug trials prematurely discontinued.
Collapse
Affiliation(s)
- Mario Failli
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Jussi Paananen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
153
|
Oprea TI. Exploring the dark genome: implications for precision medicine. Mamm Genome 2019; 30:192-200. [PMID: 31270560 DOI: 10.1007/s00335-019-09809-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/15/2019] [Indexed: 01/08/2023]
Abstract
The increase in the number of both patients and healthcare practitioners who grew up using the Internet and computers (so-called "digital natives") is likely to impact the practice of precision medicine, and requires novel platforms for data integration and mining, as well as contextualized information retrieval. The "Illuminating the Druggable Genome Knowledge Management Center" (IDG KMC) quantifies data availability from a wide range of chemical, biological, and clinical resources, and has developed platforms that can be used to navigate understudied proteins (the "dark genome"), and their potential contribution to specific pathologies. Using the "Target Importance and Novelty Explorer" (TIN-X) highlights the role of LRRC10 (a dark gene) in dilated cardiomyopathy. Combining mouse and human phenotype data leads to increased strength of evidence, which is discussed for four additional dark genes: SLX4IP and its role in glucose metabolism, the role of HSF2BP in coronary artery disease, the involvement of ELFN1 in attention-deficit hyperactivity disorder and the role of VPS13D in mouse neural tube development and its confirmed role in childhood onset movement disorders. The workflow and tools described here are aimed at guiding further experimental research, particularly within the context of precision medicine.
Collapse
Affiliation(s)
- Tudor I Oprea
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA. .,UNM Comprehensive Cancer Center, Albuquerque, NM, USA. .,Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden. .,Faculty of Health and Medical Sciences, Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
154
|
Pawar G, Madden JC, Ebbrell D, Firman JW, Cronin MTD. In Silico Toxicology Data Resources to Support Read-Across and (Q)SAR. Front Pharmacol 2019; 10:561. [PMID: 31244651 PMCID: PMC6580867 DOI: 10.3389/fphar.2019.00561] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
A plethora of databases exist online that can assist in in silico chemical or drug safety assessment. However, a systematic review and grouping of databases, based on purpose and information content, consolidated in a single source, has been lacking. To resolve this issue, this review provides a comprehensive listing of the key in silico data resources relevant to: chemical identity and properties, drug action, toxicology (including nano-material toxicity), exposure, omics, pathways, Absorption, Distribution, Metabolism and Elimination (ADME) properties, clinical trials, pharmacovigilance, patents-related databases, biological (genes, enzymes, proteins, other macromolecules etc.) databases, protein-protein interactions (PPIs), environmental exposure related, and finally databases relating to animal alternatives in support of 3Rs policies. More than nine hundred databases were identified and reviewed against criteria relating to accessibility, data coverage, interoperability or application programming interface (API), appropriate identifiers, types of in vitro, in vivo,-clinical or other data recorded and suitability for modelling, read-across, or similarity searching. This review also specifically addresses the need for solutions for mapping and integration of databases into a common platform for better translatability of preclinical data to clinical data.
Collapse
Affiliation(s)
| | | | | | | | - Mark T. D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
155
|
Canver MC, Bauer DE, Maeda T, Pinello L. DrugThatGene: integrative analysis to streamline the identification of druggable genes, pathways and protein complexes from CRISPR screens. Bioinformatics 2019; 35:1981-1984. [PMID: 30395160 PMCID: PMC6546128 DOI: 10.1093/bioinformatics/bty913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/23/2018] [Accepted: 10/31/2018] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION The clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated (Cas) nuclease system has allowed for high-throughput, large scale pooled screens for functional genomic studies. To aid in the translation of functional genomics to therapeutics, we developed DrugThatGene (DTG) as a web-based application that streamlines analysis of potential therapeutic targets identified from functional genetic screens. RESULTS Starting from a gene list as input, DTG offers automated identification of small molecules along with supporting information from human genetic and other relevant databases. Furthermore, DTG aids in the identification of common biological pathways and protein complexes in conjunction with associated small molecule inhibitors. Taken together, DTG aims to expedite the identification of small molecules from the abundance of functional genetic data generated from CRISPR screens. AVAILABILITY AND IMPLEMENTATION DTG is an open-source and free software available as a website at http://drugthatgene.pinellolab.org. Source code is available at: https://github.com/pinellolab/DrugThatGene, which can be downloaded in order to run DTG locally.
Collapse
Affiliation(s)
- Matthew C Canver
- Molecular Pathology Unit, Center for Computational and Integrative Biology, Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children’s Hospital, Boston, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Boston, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Takahiro Maeda
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Luca Pinello
- Molecular Pathology Unit, Center for Computational and Integrative Biology, Center for Cancer Research, Massachusetts General Hospital, Charlestown, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
156
|
Al-Nour MY, Ibrahim MM, Elsaman T. Ellagic Acid, Kaempferol, and Quercetin from Acacia nilotica: Promising Combined Drug With Multiple Mechanisms of Action. CURRENT PHARMACOLOGY REPORTS 2019; 5:255-280. [PMID: 32226726 PMCID: PMC7100491 DOI: 10.1007/s40495-019-00181-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The pharmacological activity of Acacia nilotica's phytochemical constituents was confirmed with evidence-based studies, but the determination of exact targets that they bind and the mechanism of action were not done; consequently, we aim to identify the exact targets that are responsible for the pharmacological activity via the computational methods. Furthermore, we aim to predict the pharmacokinetics (ADME) properties and the safety profile in order to identify the best drug candidates. To achieve those goals, various computational methods were used including the ligand-based virtual screening and molecular docking. Moreover, pkCSM and SwissADME web servers were used for the prediction of pharmacokinetics and safety. The total number of the investigated compounds and targets was 25 and 61, respectively. According to the results, the pharmacological activity was attributed to the interaction with essential targets. Ellagic acid, Kaempferol, and Quercetin were the best A. nilotica's phytochemical constituents that contribute to the therapeutic activities, were non-toxic as well as non-carcinogen. The administration of Ellagic acid, Kaempferol, and Quercetin as combined drug via the novel drug delivery systems will be a valuable therapeutic choice for the treatment of recent diseases attacking the public health including cancer, multidrug-resistant bacterial infections, diabetes mellitus, and chronic inflammatory systemic disease.
Collapse
Affiliation(s)
- Mosab Yahya Al-Nour
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, Sudan
| | - Musab Mohamed Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, Sudan
| | - Tilal Elsaman
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Omdurman Islamic University, Omdurman, Sudan
| |
Collapse
|
157
|
Ryaboshapkina M, Hammar M. Tissue-specific genes as an underutilized resource in drug discovery. Sci Rep 2019; 9:7233. [PMID: 31076736 PMCID: PMC6510781 DOI: 10.1038/s41598-019-43829-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/01/2019] [Indexed: 12/25/2022] Open
Abstract
Tissue-specific genes are believed to be good drug targets due to improved safety. Here we show that this intuitive notion is not reflected in phase 1 and 2 clinical trials, despite the historic success of tissue-specific targets and their 2.3-fold overrepresentation among targets of marketed non-oncology drugs. We compare properties of tissue-specific genes and drug targets. We show that tissue-specificity of the target may also be related to efficacy of the drug. The relationship may be indirect (enrichment in Mendelian disease and PTVesc genes) or direct (elevated betweenness centrality scores for tissue-specifically produced enzymes and secreted proteins). Reduced evolutionary conservation of tissue-specific genes may represent a bottleneck for drug projects, prompting development of novel models with smaller evolutionary gap to humans. We show that the opportunities to identify tissue-specific drug targets are not exhausted and discuss potential use cases for tissue-specific genes in drug research.
Collapse
Affiliation(s)
- Maria Ryaboshapkina
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden.
| | - Mårten Hammar
- Translational Science, Cardiovascular, Renal and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
158
|
Benstead-Hume G, Chen X, Hopkins SR, Lane KA, Downs JA, Pearl FMG. Predicting synthetic lethal interactions using conserved patterns in protein interaction networks. PLoS Comput Biol 2019; 15:e1006888. [PMID: 30995217 PMCID: PMC6488098 DOI: 10.1371/journal.pcbi.1006888] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/29/2019] [Accepted: 02/18/2019] [Indexed: 11/30/2022] Open
Abstract
In response to a need for improved treatments, a number of promising novel targeted cancer therapies are being developed that exploit human synthetic lethal interactions. This is facilitating personalised medicine strategies in cancers where specific tumour suppressors have become inactivated. Mainly due to the constraints of the experimental procedures, relatively few human synthetic lethal interactions have been identified. Here we describe SLant (Synthetic Lethal analysis via Network topology), a computational systems approach to predicting human synthetic lethal interactions that works by identifying and exploiting conserved patterns in protein interaction network topology both within and across species. SLant out-performs previous attempts to classify human SSL interactions and experimental validation of the models predictions suggests it may provide useful guidance for future SSL screenings and ultimately aid targeted cancer therapy development.
Collapse
Affiliation(s)
- Graeme Benstead-Hume
- Bioinformatics Lab, School of Life Sciences, University of Sussex, Falmer, Brighton, United Kingdom
| | - Xiangrong Chen
- Bioinformatics Lab, School of Life Sciences, University of Sussex, Falmer, Brighton, United Kingdom
| | - Suzanna R. Hopkins
- Division of Cancer Biology, Institute of Cancer Research, Chester Beatty Laboratories, London, United Kingdom
| | - Karen A. Lane
- Division of Cancer Biology, Institute of Cancer Research, Chester Beatty Laboratories, London, United Kingdom
| | - Jessica A. Downs
- Division of Cancer Biology, Institute of Cancer Research, Chester Beatty Laboratories, London, United Kingdom
| | - Frances M. G. Pearl
- Bioinformatics Lab, School of Life Sciences, University of Sussex, Falmer, Brighton, United Kingdom
| |
Collapse
|
159
|
Gaspar HA, Gerring Z, Hübel C, Middeldorp CM, Derks EM, Breen G. Using genetic drug-target networks to develop new drug hypotheses for major depressive disorder. Transl Psychiatry 2019; 9:117. [PMID: 30877270 PMCID: PMC6420656 DOI: 10.1038/s41398-019-0451-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 01/28/2019] [Accepted: 02/12/2019] [Indexed: 12/25/2022] Open
Abstract
The major depressive disorder (MDD) working group of the Psychiatric Genomics Consortium (PGC) has published a genome-wide association study (GWAS) for MDD in 130,664 cases, identifying 44 risk variants. We used these results to investigate potential drug targets and repurposing opportunities. We built easily interpretable bipartite drug-target networks integrating interactions between drugs and their targets, genome-wide association statistics, and genetically predicted expression levels in different tissues, using the online tool Drug Targetor ( drugtargetor.com ). We also investigated drug-target relationships that could be impacting MDD. MAGMA was used to perform pathway analyses and S-PrediXcan to investigate the directionality of tissue-specific expression levels in patients vs. controls. Outside the major histocompatibility complex (MHC) region, 153 protein-coding genes are significantly associated with MDD in MAGMA after multiple testing correction; among these, five are predicted to be down or upregulated in brain regions and 24 are known druggable genes. Several drug classes were significantly enriched, including monoamine reuptake inhibitors, sex hormones, antipsychotics, and antihistamines, indicating an effect on MDD and potential repurposing opportunities. These findings not only require validation in model systems and clinical examination, but also show that GWAS may become a rich source of new therapeutic hypotheses for MDD and other psychiatric disorders that need new-and better-treatment options.
Collapse
Affiliation(s)
- Héléna A Gaspar
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, SE5 8AF, UK.
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, EC1V 2PD, UK.
| | - Zachary Gerring
- Translational Neurogenomics Laboratory, QIMR Berghofer Institute of Medical Research, Brisbane City, QLD 4006, Australia
| | - Christopher Hübel
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, SE5 8AF, UK
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, EC1V 2PD, UK
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Christel M Middeldorp
- Child Health Research Centre, University of Queensland, South Brisbane, QLD 4072, Australia
- Child and Youth Mental Health Service, Children's Health Queensland Hospital and Health Service, South Brisbane, QLD 4101, Australia
- Biological Psychology, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, Netherlands
| | - Eske M Derks
- Translational Neurogenomics Laboratory, QIMR Berghofer Institute of Medical Research, Brisbane City, QLD 4006, Australia
| | - Gerome Breen
- King's College London, Institute of Psychiatry, Psychology and Neuroscience, Social, Genetic and Developmental Psychiatry (SGDP) Centre, London, SE5 8AF, UK
- National Institute for Health Research Biomedical Research Centre, South London and Maudsley National Health Service Trust, London, EC1V 2PD, UK
| |
Collapse
|
160
|
Chen YA, Yogo E, Kurihara N, Ohno T, Higuchi C, Rokushima M, Mizuguchi K. Assessing drug target suitability using TargetMine. F1000Res 2019; 8:233. [PMID: 30984386 PMCID: PMC6439796 DOI: 10.12688/f1000research.18214.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/23/2019] [Indexed: 12/22/2022] Open
Abstract
In selecting drug target candidates for pharmaceutical research, the linkage to disease and the tractability of the target are two important factors that can ultimately determine the drug efficacy. Several existing resources can provide gene-disease associations, but determining whether such a list of genes are attractive drug targets often requires further information gathering and analysis. In addition, few resources provide the information required to evaluate the tractability of a target. To address these issues, we have updated TargetMine, a data warehouse for assisting target prioritization, by integrating new data sources for gene-disease associations and enhancing functionalities for target assessment. As a data mining platform that integrates a variety of data sources, including protein structures and chemical compounds, TargetMine now offers a powerful and flexible interface for constructing queries to check genetic evidence, tractability and other relevant features for the candidate genes. We demonstrate these features by using several specific examples.
Collapse
Affiliation(s)
- Yi-An Chen
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 5670085, Japan
| | - Erika Yogo
- Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Toyonaka, Osaka, 5610825, Japan
| | - Naoko Kurihara
- Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Toyonaka, Osaka, 5610825, Japan
| | - Tomoshige Ohno
- Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Toyonaka, Osaka, 5610825, Japan
| | - Chihiro Higuchi
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 5670085, Japan
| | - Masatomo Rokushima
- Shionogi Pharmaceutical Research Center, Shionogi & Co., Ltd., Toyonaka, Osaka, 5610825, Japan
| | - Kenji Mizuguchi
- National Institutes of Biomedical Innovation, Health and Nutrition, Ibaraki, Osaka, 5670085, Japan
| |
Collapse
|
161
|
Doncheva NT, Morris JH, Gorodkin J, Jensen LJ. Cytoscape StringApp: Network Analysis and Visualization of Proteomics Data. J Proteome Res 2019. [PMID: 30450911 DOI: 10.1101/438192] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
Protein networks have become a popular tool for analyzing and visualizing the often long lists of proteins or genes obtained from proteomics and other high-throughput technologies. One of the most popular sources of such networks is the STRING database, which provides protein networks for more than 2000 organisms, including both physical interactions from experimental data and functional associations from curated pathways, automatic text mining, and prediction methods. However, its web interface is mainly intended for inspection of small networks and their underlying evidence. The Cytoscape software, on the other hand, is much better suited for working with large networks and offers greater flexibility in terms of network analysis, import, and visualization of additional data. To include both resources in the same workflow, we created stringApp, a Cytoscape app that makes it easy to import STRING networks into Cytoscape, retains the appearance and many of the features of STRING, and integrates data from associated databases. Here, we introduce many of the stringApp features and show how they can be used to carry out complex network analysis and visualization tasks on a typical proteomics data set, all through the Cytoscape user interface. stringApp is freely available from the Cytoscape app store: http://apps.cytoscape.org/apps/stringapp .
Collapse
Affiliation(s)
- Nadezhda T Doncheva
- Novo Nordisk Foundation Center for Protein Research , University of Copenhagen , 2200 Copenhagen N, Denmark
- Center for Non-Coding RNA in Technology and Health , University of Copenhagen , 1870 Frederiksberg C, Denmark
- Department of Veterinary and Animal Sciences , University of Copenhagen , 1870 Frederiksberg C, Denmark
| | - John H Morris
- Resource on Biocomputing, Visualization, and Informatics , University of California , San Francisco , California 94158-2517 , United States
| | - Jan Gorodkin
- Center for Non-Coding RNA in Technology and Health , University of Copenhagen , 1870 Frederiksberg C, Denmark
- Department of Veterinary and Animal Sciences , University of Copenhagen , 1870 Frederiksberg C, Denmark
| | - Lars J Jensen
- Novo Nordisk Foundation Center for Protein Research , University of Copenhagen , 2200 Copenhagen N, Denmark
- Center for Non-Coding RNA in Technology and Health , University of Copenhagen , 1870 Frederiksberg C, Denmark
| |
Collapse
|
162
|
Vijayan V, Rouillard AD, Rajpal DK, Agarwal P. Could advances in representation learning in Artificial Intelligence provide the new paradigm for data integration in drug discovery? Expert Opin Drug Discov 2019; 14:191-194. [PMID: 30696299 DOI: 10.1080/17460441.2019.1573811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Vinaya Vijayan
- a Computational Biology , GlaxoSmithKline R&D , Collegeville , PA , USA
| | | | - Deepak K Rajpal
- a Computational Biology , GlaxoSmithKline R&D , Collegeville , PA , USA
| | - Pankaj Agarwal
- a Computational Biology , GlaxoSmithKline R&D , Collegeville , PA , USA
| |
Collapse
|
163
|
Griesenauer RH, Schillebeeckx C, Kinch MS. Assessing the public landscape of clinical-stage pharmaceuticals through freely available online databases. Drug Discov Today 2019; 24:1010-1016. [PMID: 30690196 DOI: 10.1016/j.drudis.2019.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/19/2018] [Accepted: 01/22/2019] [Indexed: 12/23/2022]
Abstract
Several public databases have emerged over the past decade to enable chemo- and bio-informatics research in the field of drug development. To a naive observer, as well as many seasoned professionals, the differences among many drug databases are unclear. We assessed the availability of all pharmaceuticals with evidence of clinical testing (i.e., been in at least a Phase I clinical trial) and highlight the major differences and similarities between public databases containing clinically tested pharmaceuticals. We review a selection of the most recent and prominent databases including: ChEMBL, CRIB NME, DrugBank, DrugCentral, PubChem, repoDB, SuperDrug2 and WITHDRAWN, and found that ∼11700 unique active pharmaceutical ingredients are available in the public domain, with evidence of clinical testing.
Collapse
Affiliation(s)
- Rebekah H Griesenauer
- Center for Research Innovation in Biotechnology, Washington University in St Louis, MO 63130, USA
| | | | - Michael S Kinch
- Center for Research Innovation in Biotechnology, Washington University in St Louis, MO 63130, USA.
| |
Collapse
|
164
|
Ursu O, Holmes J, Bologa CG, Yang JJ, Mathias SL, Stathias V, Nguyen DT, Schürer S, Oprea T. DrugCentral 2018: an update. Nucleic Acids Res 2019; 47:D963-D970. [PMID: 30371892 PMCID: PMC6323925 DOI: 10.1093/nar/gky963] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/02/2018] [Accepted: 10/26/2018] [Indexed: 01/21/2023] Open
Abstract
DrugCentral is a drug information resource (http://drugcentral.org) open to the public since 2016 and previously described in the 2017 Nucleic Acids Research Database issue. Since the 2016 release, 103 new approved drugs were updated. The following new data sources have been included: Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS), FDA Orange Book information, L1000 gene perturbation profile distance/similarity matrices and estimated protonation constants. New and existing entries have been updated with the latest information from scientific literature, drug labels and external databases. The web interface has been updated to display and query new data. The full database dump and data files are available for download from the DrugCentral website.
Collapse
Affiliation(s)
- Oleg Ursu
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Jayme Holmes
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Cristian G Bologa
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Jeremy J Yang
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Stephen L Mathias
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Vasileios Stathias
- Center for Computational Science, Miller School of Medicine, University of Miami, Coral Gables, FL 33146, USA
| | - Dac-Trung Nguyen
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Stephan Schürer
- Center for Computational Science, Miller School of Medicine, University of Miami, Coral Gables, FL 33146, USA
| | - Tudor Oprea
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| |
Collapse
|
165
|
Alaimo S, Pulvirenti A. Network-Based Drug Repositioning: Approaches, Resources, and Research Directions. Methods Mol Biol 2019; 1903:97-113. [PMID: 30547438 DOI: 10.1007/978-1-4939-8955-3_6] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The wealth of knowledge and omic data available in drug research allowed the rising of several computational methods in drug discovery field yielding a novel and exciting application called drug repositioning. Several computational methods try to make a high-level integration of all the knowledge in order to discover unknown mechanisms. In this chapter we present an in-depth review of data resources and computational models for drug repositioning.
Collapse
Affiliation(s)
- Salvatore Alaimo
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Alfredo Pulvirenti
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy.
| |
Collapse
|
166
|
Abstract
Computational prediction of the clinical success or failure of a potential drug target for therapeutic use is a challenging problem. Novel network propagation algorithms that integrate heterogeneous biological networks are proving useful for drug target identification and prioritization. These approaches typically utilize a network describing relationships between targets, a method to disseminate the relevant information through the network, and a method to elucidate new associations between targets and diseases. Here, we utilize one such network propagation-based approach, DTINet, which starts with diffusion component analysis of networks of both potential drug targets and diseases. Then an inductive matrix completion algorithm is applied to identify novel disease targets based on their network topological similarities with known disease targets with successfully launched drugs. DTINet performed well as assessed with area under the precision-recall curve (AUPR = 0.88 ± 0.007) and area under the receiver operating characteristic curve (AUROC = 0.86 ± 0.008). These metrics improved when we combined data from multiple networks in the target space but reduced significantly when we used a more conservative method to define negative controls (AUPR = 0.56 ± 0.007, AUROC = 0.57 ± 0.007). We are optimistic that integration of more relevant and cleaner datasets and networks, careful calibration of model parameters, as well as algorithmic improvements will improve prediction accuracy. However, we also recognize that predicting drug targets that are likely to be successful is an extremely challenging problem due to its complex nature and sparsity of known disease targets.
Collapse
|
167
|
Abstract
Pharmacological science is trying to establish the link between chemicals, targets, and disease-related phenotypes. A plethora of chemical proteomics and structural data have been generated, thanks to the target-based approach that has dominated drug discovery at the turn of the century. There is an invaluable source of information for in silico target profiling. Prediction is based on the principle of chemical similarity (similar drugs bind similar targets) or on first principles from the biophysics of molecular interactions. In the first case, compound comparison is made through ligand-based chemical similarity search or through classifier-based machine learning approach. The 3D techniques are based on 3D structural descriptors or energy-based scoring scheme to infer a binding affinity of a compound with its putative target. More recently, a new approach based on compound set metric has been proposed in which a query compound is compared with a whole of compounds associated with a target or a family of targets. This chapter reviews the different techniques of in silico target profiling and their main applications such as inference of unwanted targets, drug repurposing, or compound prioritization after phenotypic-based screening campaigns.
Collapse
|
168
|
Browne CM, Jiang B, Ficarro SB, Doctor ZM, Johnson JL, Card JD, Sivakumaren SC, Alexander WM, Yaron TM, Murphy CJ, Kwiatkowski NP, Zhang T, Cantley LC, Gray NS, Marto JA. A Chemoproteomic Strategy for Direct and Proteome-Wide Covalent Inhibitor Target-Site Identification. J Am Chem Soc 2018; 141:191-203. [PMID: 30518210 DOI: 10.1021/jacs.8b07911] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Despite recent clinical successes for irreversible drugs, potential toxicities mediated by unpredictable modification of off-target cysteines represents a major hurdle for expansion of covalent drug programs. Understanding the proteome-wide binding profile of covalent inhibitors can significantly accelerate their development; however, current mass spectrometry strategies typically do not provide a direct, amino acid level readout of covalent activity for complex, selective inhibitors. Here we report the development of CITe-Id, a novel chemoproteomic approach that employs covalent pharmacologic inhibitors as enrichment reagents in combination with an optimized proteomic platform to directly quantify dose-dependent binding at cysteine-thiols across the proteome. CITe-Id analysis of our irreversible CDK inhibitor THZ1 identified dose-dependent covalent modification of several unexpected kinases, including a previously unannotated cysteine (C840) on the understudied kinase PKN3. These data streamlined our development of JZ128 as a new selective covalent inhibitor of PKN3. Using JZ128 as a probe compound, we identified novel potential PKN3 substrates, thus offering an initial molecular view of PKN3 cellular activity. CITe-Id provides a powerful complement to current chemoproteomic platforms to characterize the selectivity of covalent inhibitors, identify new, pharmacologically addressable cysteine-thiols, and inform structure-based drug design programs.
Collapse
Affiliation(s)
- Christopher M Browne
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Baishan Jiang
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Scott B Ficarro
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States.,Blais Proteomics Center , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States
| | - Zainab M Doctor
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jared L Johnson
- Meyer Cancer Center , Weill Cornell Medicine and New York Presbyterian Hospital , New York , New York 10065 , United States
| | - Joseph D Card
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Blais Proteomics Center , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States
| | - Sindhu Carmen Sivakumaren
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - William M Alexander
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Blais Proteomics Center , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States
| | - Tomer M Yaron
- Meyer Cancer Center , Weill Cornell Medicine and New York Presbyterian Hospital , New York , New York 10065 , United States
| | - Charles J Murphy
- Meyer Cancer Center , Weill Cornell Medicine and New York Presbyterian Hospital , New York , New York 10065 , United States
| | - Nicholas P Kwiatkowski
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States.,Whitehead Institute for Biomedical Research , Cambridge , Massachusetts 02142 , United States
| | - Tinghu Zhang
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Lewis C Cantley
- Meyer Cancer Center , Weill Cornell Medicine and New York Presbyterian Hospital , New York , New York 10065 , United States
| | - Nathanael S Gray
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Biological Chemistry and Molecular Pharmacology , Harvard Medical School , Boston , Massachusetts 02115 , United States
| | - Jarrod A Marto
- Department of Cancer Biology , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Blais Proteomics Center , Dana-Farber Cancer Institute , Boston , Massachusetts 02215 , United States.,Department of Pathology , Brigham and Women's Hospital, Harvard Medical School , Boston , Massachusetts 02115 , United States
| |
Collapse
|
169
|
Shergalis A, Bankhead A, Luesakul U, Muangsin N, Neamati N. Current Challenges and Opportunities in Treating Glioblastoma. Pharmacol Rev 2018; 70:412-445. [PMID: 29669750 PMCID: PMC5907910 DOI: 10.1124/pr.117.014944] [Citation(s) in RCA: 504] [Impact Index Per Article: 84.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common and aggressive primary brain tumor, has a high mortality rate despite extensive efforts to develop new treatments. GBM exhibits both intra- and intertumor heterogeneity, lending to resistance and eventual tumor recurrence. Large-scale genomic and proteomic analysis of GBM tumors has uncovered potential drug targets. Effective and “druggable” targets must be validated to embark on a robust medicinal chemistry campaign culminating in the discovery of clinical candidates. Here, we review recent developments in GBM drug discovery and delivery. To identify GBM drug targets, we performed extensive bioinformatics analysis using data from The Cancer Genome Atlas project. We discovered 20 genes, BOC, CLEC4GP1, ELOVL6, EREG, ESR2, FDCSP, FURIN, FUT8-AS1, GZMB, IRX3, LITAF, NDEL1, NKX3-1, PODNL1, PTPRN, QSOX1, SEMA4F, TH, VEGFC, and C20orf166AS1 that are overexpressed in a subpopulation of GBM patients and correlate with poor survival outcomes. Importantly, nine of these genes exhibit higher expression in GBM versus low-grade glioma and may be involved in disease progression. In this review, we discuss these proteins in the context of GBM disease progression. We also conducted computational multi-parameter optimization to assess the blood-brain barrier (BBB) permeability of small molecules in clinical trials for GBM treatment. Drug delivery in the context of GBM is particularly challenging because the BBB hinders small molecule transport. Therefore, we discuss novel drug delivery methods, including nanoparticles and prodrugs. Given the aggressive nature of GBM and the complexity of targeting the central nervous system, effective treatment options are a major unmet medical need. Identification and validation of biomarkers and drug targets associated with GBM disease progression present an exciting opportunity to improve treatment of this devastating disease.
Collapse
Affiliation(s)
- Andrea Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Armand Bankhead
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Urarika Luesakul
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Nongnuj Muangsin
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| |
Collapse
|
170
|
Doncheva NT, Morris JH, Gorodkin J, Jensen LJ. Cytoscape StringApp: Network Analysis and Visualization of Proteomics Data. J Proteome Res 2018; 18:623-632. [PMID: 30450911 DOI: 10.1021/acs.jproteome.8b00702] [Citation(s) in RCA: 1166] [Impact Index Per Article: 194.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protein networks have become a popular tool for analyzing and visualizing the often long lists of proteins or genes obtained from proteomics and other high-throughput technologies. One of the most popular sources of such networks is the STRING database, which provides protein networks for more than 2000 organisms, including both physical interactions from experimental data and functional associations from curated pathways, automatic text mining, and prediction methods. However, its web interface is mainly intended for inspection of small networks and their underlying evidence. The Cytoscape software, on the other hand, is much better suited for working with large networks and offers greater flexibility in terms of network analysis, import, and visualization of additional data. To include both resources in the same workflow, we created stringApp, a Cytoscape app that makes it easy to import STRING networks into Cytoscape, retains the appearance and many of the features of STRING, and integrates data from associated databases. Here, we introduce many of the stringApp features and show how they can be used to carry out complex network analysis and visualization tasks on a typical proteomics data set, all through the Cytoscape user interface. stringApp is freely available from the Cytoscape app store: http://apps.cytoscape.org/apps/stringapp .
Collapse
Affiliation(s)
- Nadezhda T Doncheva
- Novo Nordisk Foundation Center for Protein Research , University of Copenhagen , 2200 Copenhagen N, Denmark.,Center for Non-Coding RNA in Technology and Health , University of Copenhagen , 1870 Frederiksberg C, Denmark.,Department of Veterinary and Animal Sciences , University of Copenhagen , 1870 Frederiksberg C, Denmark
| | - John H Morris
- Resource on Biocomputing, Visualization, and Informatics , University of California , San Francisco , California 94158-2517 , United States
| | - Jan Gorodkin
- Center for Non-Coding RNA in Technology and Health , University of Copenhagen , 1870 Frederiksberg C, Denmark.,Department of Veterinary and Animal Sciences , University of Copenhagen , 1870 Frederiksberg C, Denmark
| | - Lars J Jensen
- Novo Nordisk Foundation Center for Protein Research , University of Copenhagen , 2200 Copenhagen N, Denmark.,Center for Non-Coding RNA in Technology and Health , University of Copenhagen , 1870 Frederiksberg C, Denmark
| |
Collapse
|
171
|
Halu A, Wang JG, Iwata H, Mojcher A, Abib AL, Singh SA, Aikawa M, Sharma A. Context-enriched interactome powered by proteomics helps the identification of novel regulators of macrophage activation. eLife 2018; 7:37059. [PMID: 30303482 PMCID: PMC6179386 DOI: 10.7554/elife.37059] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 08/30/2018] [Indexed: 02/06/2023] Open
Abstract
The role of pro-inflammatory macrophage activation in cardiovascular disease (CVD) is a complex one amenable to network approaches. While an indispensible tool for elucidating the molecular underpinnings of complex diseases including CVD, the interactome is limited in its utility as it is not specific to any cell type, experimental condition or disease state. We introduced context-specificity to the interactome by combining it with co-abundance networks derived from unbiased proteomics measurements from activated macrophage-like cells. Each macrophage phenotype contributed to certain regions of the interactome. Using a network proximity-based prioritization method on the combined network, we predicted potential regulators of macrophage activation. Prediction performance significantly increased with the addition of co-abundance edges, and the prioritized candidates captured inflammation, immunity and CVD signatures. Integrating the novel network topology with transcriptomics and proteomics revealed top candidate drivers of inflammation. In vitro loss-of-function experiments demonstrated the regulatory role of these proteins in pro-inflammatory signaling. When human cells or tissues are injured, the body triggers a response known as inflammation to repair the damage and protect itself from further harm. However, if the same issue keeps recurring, the tissues become inflamed for longer periods of time, which may ultimately lead to health problems. This is what could be happening in cardiovascular diseases, where long-term inflammation could damage the heart and blood vessels. Many different proteins interact with each other to control inflammation; gaining an insight into the nature of these interactions could help to pinpoint the role of each molecular actor. Researchers have used a combination of unbiased, large-scale experimental and computational approaches to develop the interactome, a map of the known interactions between all proteins in humans. However, interactions between proteins can change between cell types, or during disease. Here, Halu et al. aimed to refine the human interactome and identify new proteins involved in inflammation, especially in the context of cardiovascular disease. Cells called macrophages produce signals that trigger inflammation whey they detect damage in other cells or tissues. The experiments used a technique called proteomics to measure the amounts of all the proteins in human macrophages. Combining these data with the human interactome made it possible to predict new links between proteins known to have a role in inflammation and other proteins in the interactome. Further analysis using other sets of data from macrophages helped identify two new candidate proteins – GBP1 and WARS – that may promote inflammation. Halu et al. then used a genetic approach to deactivate the genes and decrease the levels of these two proteins in macrophages, which caused the signals that encourage inflammation to drop. These findings suggest that GBP1 and WARS regulate the activity of macrophages to promote inflammation. The two proteins could therefore be used as drug targets to treat cardiovascular diseases and other disorders linked to inflammation, but further studies will be needed to precisely dissect how GBP1 and WARS work in humans.
Collapse
Affiliation(s)
- Arda Halu
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States.,Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Jian-Guo Wang
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Hiroshi Iwata
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Alexander Mojcher
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Ana Luisa Abib
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Sasha A Singh
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Amitabh Sharma
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
172
|
Palasca O, Santos A, Stolte C, Gorodkin J, Jensen LJ. TISSUES 2.0: an integrative web resource on mammalian tissue expression. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2018; 2018:4851151. [PMID: 29617745 PMCID: PMC5808782 DOI: 10.1093/database/bay003] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 01/04/2018] [Indexed: 11/13/2022]
Abstract
Physiological and molecular similarities between organisms make it possible to translate findings from simpler experimental systems—model organisms—into more complex ones, such as human. This translation facilitates the understanding of biological processes under normal or disease conditions. Researchers aiming to identify the similarities and differences between organisms at the molecular level need resources collecting multi-organism tissue expression data. We have developed a database of gene–tissue associations in human, mouse, rat and pig by integrating multiple sources of evidence: transcriptomics covering all four species and proteomics (human only), manually curated and mined from the scientific literature. Through a scoring scheme, these associations are made comparable across all sources of evidence and across organisms. Furthermore, the scoring produces a confidence score assigned to each of the associations. The TISSUES database (version 2.0) is publicly accessible through a user-friendly web interface and as part of the STRING app for Cytoscape. In addition, we analyzed the agreement between datasets, across and within organisms, and identified that the agreement is mainly affected by the quality of the datasets rather than by the technologies used or organisms compared. Database URL: http://tissues.jensenlab.org/
Collapse
Affiliation(s)
- Oana Palasca
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for non-coding RNA in Technology and Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Alberto Santos
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jan Gorodkin
- Center for non-coding RNA in Technology and Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lars Juhl Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for non-coding RNA in Technology and Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
173
|
Freudenberg JM, Dunham I, Sanseau P, Rajpal DK. Uncovering new disease indications for G-protein coupled receptors and their endogenous ligands. BMC Bioinformatics 2018; 19:345. [PMID: 30285606 PMCID: PMC6167889 DOI: 10.1186/s12859-018-2392-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/23/2018] [Indexed: 11/29/2022] Open
Abstract
Background The Open Targets Platform integrates different data sources in order to facilitate identification of potential therapeutic drug targets to treat human diseases. It currently provides evidence for nearly 2.6 million potential target-disease pairs. G-protein coupled receptors are a drug target class of high interest because of the number of successful drugs being developed against them over many years. Here we describe a systematic approach utilizing the Open Targets Platform data to uncover and prioritize potential new disease indications for the G-protein coupled receptors and their ligands. Results Utilizing the data available in the Open Targets platform, potential G-protein coupled receptor and endogenous ligand disease association pairs were systematically identified. Intriguing examples such as GPR35 for inflammatory bowel disease and CXCR4 for viral infection are used as illustrations of how a systematic approach can aid in the prioritization of interesting drug discovery hypotheses. Combining evidences for G-protein coupled receptors and their corresponding endogenous peptidergic ligands increases confidence and provides supportive evidence for potential new target-disease hypotheses. Comparing such hypotheses to the global pharma drug discovery pipeline to validate the approach showed that more than 93% of G-protein coupled receptor-disease pairs with a high overall Open Targets score involved receptors with an existing drug discovery program. Conclusions The Open Targets gene-disease score can be used to prioritize potential G-protein coupled receptors-indication hypotheses. In addition, availability of multiple different evidence types markedly increases confidence as does combining evidence from known receptor-ligand pairs. Comparing the top-ranked hypotheses to the current global pharma pipeline serves validation of our approach and identifies and prioritizes new therapeutic opportunities. Electronic supplementary material The online version of this article (10.1186/s12859-018-2392-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Ian Dunham
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.,European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Philippe Sanseau
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.,Computational Biology and Stats, Target Sciences, GSK Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Deepak K Rajpal
- Computational Biology, Target Sciences, GlaxoSmithKline, Collegeville, PA, 19426, USA.
| |
Collapse
|
174
|
Piñero J, Gonzalez-Perez A, Guney E, Aguirre-Plans J, Sanz F, Oliva B, Furlong LI. Network, Transcriptomic and Genomic Features Differentiate Genes Relevant for Drug Response. Front Genet 2018; 9:412. [PMID: 30319692 PMCID: PMC6168038 DOI: 10.3389/fgene.2018.00412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/05/2018] [Indexed: 11/13/2022] Open
Abstract
Understanding the mechanisms underlying drug therapeutic action and toxicity is crucial for the prevention and management of drug adverse reactions, and paves the way for a more efficient and rational drug design. The characterization of drug targets, drug metabolism proteins, and proteins associated to side effects according to their expression patterns, their tolerance to genomic variation and their role in cellular networks, is a necessary step in this direction. In this contribution, we hypothesize that different classes of proteins involved in the therapeutic effect of drugs and in their adverse effects have distinctive transcriptomics, genomics and network features. We explored the properties of these proteins within global and organ-specific interactomes, using multi-scale network features, evaluated their gene expression profiles in different organs and tissues, and assessed their tolerance to loss-of-function variants leveraging data from 60K subjects. We found that drug targets that mediate side effects are more central in cellular networks, more intolerant to loss-of-function variation, and show a wider breadth of tissue expression than targets not mediating side effects. In contrast, drug metabolizing enzymes and transporters are less central in the interactome, more tolerant to deleterious variants, and are more constrained in their tissue expression pattern. Our findings highlight distinctive features of proteins related to drug action, which could be applied to prioritize drugs with fewer probabilities of causing side effects.
Collapse
Affiliation(s)
- Janet Piñero
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Abel Gonzalez-Perez
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Emre Guney
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Joaquim Aguirre-Plans
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ferran Sanz
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Baldo Oliva
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laura I Furlong
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
175
|
Morfa CJ, Bassoni D, Szabo A, McAnally D, Sharir H, Hood BL, Vasile S, Wehrman T, Lamerdin J, Smith LH. A Pharmacochaperone-Based High-Throughput Screening Assay for the Discovery of Chemical Probes of Orphan Receptors. Assay Drug Dev Technol 2018; 16:384-396. [PMID: 30251873 DOI: 10.1089/adt.2018.868] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) have varying and diverse physiological roles, transmitting signals from a range of stimuli, including light, chemicals, peptides, and mechanical forces. More than 130 GPCRs are orphan receptors (i.e., their endogenous ligands are unknown), representing a large untapped reservoir of potential therapeutic targets for pharmaceutical intervention in a variety of diseases. Current deorphanization approaches are slow, laborious, and usually require some in-depth knowledge about the receptor pharmacology. In this study we describe a cell-based assay to identify small molecule probes of orphan receptors that requires no a priori knowledge of receptor pharmacology. Built upon the concept of pharmacochaperones, where cell-permeable small molecules facilitate the trafficking of mutant receptors to the plasma membrane, the simple and robust technology is readily accessible by most laboratories and is amenable to high-throughput screening. The assay consists of a target harboring a synthetic point mutation that causes retention of the target in the endoplasmic reticulum. Coupled with a beta-galactosidase enzyme-fragment complementation reporter system, the assay identifies compounds that act as pharmacochaperones causing forward trafficking of the mutant GPCR. The assay can identify compounds with varying mechanisms of action including agonists and antagonists. A universal positive control compound circumvents the need for a target-specific ligand. The veracity of the approach is demonstrated using the beta-2-adrenergic receptor. Together with other existing assay technologies to validate the signaling pathways and the specificity of ligands identified, this pharmacochaperone-based approach can accelerate the identification of ligands for these potentially therapeutically useful receptors.
Collapse
Affiliation(s)
- Camilo J Morfa
- 1 Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | | | - Andras Szabo
- 1 Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Danielle McAnally
- 1 Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Haleli Sharir
- 1 Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Becky L Hood
- 1 Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Stefan Vasile
- 1 Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| | - Tom Wehrman
- 2 Eurofins DiscoverX Corporation , Fremont, California
| | - Jane Lamerdin
- 2 Eurofins DiscoverX Corporation , Fremont, California
| | - Layton H Smith
- 1 Conrad Prebys Center for Chemical Genomics, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida
| |
Collapse
|
176
|
Zhi LQ, Yang YX, Yao SX, Qing Z, Ma JB. Identification of Novel Target for Osteosarcoma by Network Analysis. Med Sci Monit 2018; 24:5914-5924. [PMID: 30144309 PMCID: PMC6120164 DOI: 10.12659/msm.909973] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Osteosarcoma (OS) is a highly complicated bone cancer involving imbalance of signaling transduction networks in cells. Development of new anti-osteosarcoma drugs is very challenging, mainly due to lack of known key targets. Material/Method In this study, we attempted to reveal more promising targets for drug design by “Target-Pathway” network analysis, providing the new therapeutic strategy of osteosarcoma. The potential targets used for the treatment of OS were selected from 4 different sources: DrugBank, TCRD database, dbDEMC database, and recent scientific literature papers. Cytoscape was used for the establishment of the “Target-Pathway” network. Results The obtained results suggest that tankyrase 2 (TNKS2) might be a very good potential protein target for the treatment of osteosarcoma. An in vitro MTT assay proved that it is an available option against OS by targeting the TNKS2 protein. Subsequently, cell cycle and apoptosis assay by flow cytometry showed the TNKS2 inhibitor can obviously induce cell cycle arrest, apoptosis, and mitotic cell death. Conclusions Tankyrase 2 (TNKS2), a member of the multifunctional poly(ADP-ribose) polymerases (PARPs), could be a very useful protein target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Li-Qiang Zhi
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Yi-Xin Yang
- Medical Experiment Center, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China (mainland)
| | - Shu-Xin Yao
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Zhong Qing
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| | - Jian-Bing Ma
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China (mainland)
| |
Collapse
|
177
|
Floris M, Olla S, Schlessinger D, Cucca F. Genetic-Driven Druggable Target Identification and Validation. Trends Genet 2018; 34:558-570. [PMID: 29803319 PMCID: PMC6088790 DOI: 10.1016/j.tig.2018.04.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/13/2018] [Accepted: 04/23/2018] [Indexed: 12/19/2022]
Abstract
Choosing the right biological target is the critical primary decision for the development of new drugs. Systematic genetic association testing of both human diseases and quantitative traits, along with resultant findings of coincident associations between them, is becoming a powerful approach to infer drug targetable candidates and generate in vitro tests to identify compounds that can modulate them therapeutically. Here, we discuss opportunities and challenges, and infer criteria for the optimal use of genetic findings in the drug discovery pipeline.
Collapse
Affiliation(s)
- Matteo Floris
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy; IRGB-CNR, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | - Stefania Olla
- IRGB-CNR, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | - David Schlessinger
- Laboratory of Genetics, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Francesco Cucca
- Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy; IRGB-CNR, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy.
| |
Collapse
|
178
|
Sinkala M, Mulder N, Martin DP. Integrative landscape of dysregulated signaling pathways of clinically distinct pancreatic cancer subtypes. Oncotarget 2018; 9:29123-29139. [PMID: 30018740 PMCID: PMC6044387 DOI: 10.18632/oncotarget.25632] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022] Open
Abstract
Despite modern therapeutic advances, the survival prospects of pancreatic cancer patients have remained poor. Besides being highly metastatic, pancreatic cancer is challenging to treat because it is caused by a heterogeneous array of somatic mutations that impact a variety of signaling pathways and cellular regulatory systems. Here we use publicly available transcriptomic, copy number alteration and mutation profiling datasets from pancreatic cancer patients together with data on disease outcomes to show that the three major pancreatic cancer subtypes each display distinctive aberrations in cell signaling and metabolic pathways. Importantly, patients afflicted with these different pancreatic cancer subtypes also exhibit distinctive survival profiles. Within these patients, we find that dysregulation of the phosphoinositide 3-kinase and mitogen-activated protein kinase pathways, and p53 mediated disruptions of cell cycle processes are apparently drivers of disease. Further, we identify for the first time the molecular perturbations of signalling networks that are likely the primary causes of oncogenesis in each of the three pancreatic cancer subtypes.
Collapse
Affiliation(s)
- Musalula Sinkala
- University of Cape Town, School of Health Sciences, Department of Integrative Biomedical Sciences, Computational Biology Division, Observatory, 7925, South Africa
| | - Nicola Mulder
- University of Cape Town, School of Health Sciences, Department of Integrative Biomedical Sciences, Computational Biology Division, Observatory, 7925, South Africa
| | - Darren Patrick Martin
- University of Cape Town, School of Health Sciences, Department of Integrative Biomedical Sciences, Computational Biology Division, Observatory, 7925, South Africa
| |
Collapse
|
179
|
Oprea TI, Bologa CG, Brunak S, Campbell A, Gan GN, Gaulton A, Gomez SM, Guha R, Hersey A, Holmes J, Jadhav A, Jensen LJ, Johnson GL, Karlson A, Leach AR, Ma’ayan A, Malovannaya A, Mani S, Mathias SL, McManus MT, Meehan TF, von Mering C, Muthas D, Nguyen DT, Overington JP, Papadatos G, Qin J, Reich C, Roth BL, Schürer SC, Simeonov A, Sklar LA, Southall N, Tomita S, Tudose I, Ursu O, Vidovic D, Waller A, Westergaard D, Yang JJ, Zahoránszky-Köhalmi G. Unexplored therapeutic opportunities in the human genome. Nat Rev Drug Discov 2018; 17:317-332. [PMID: 29472638 PMCID: PMC6339563 DOI: 10.1038/nrd.2018.14] [Citation(s) in RCA: 209] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A large proportion of biomedical research and the development of therapeutics is focused on a small fraction of the human genome. In a strategic effort to map the knowledge gaps around proteins encoded by the human genome and to promote the exploration of currently understudied, but potentially druggable, proteins, the US National Institutes of Health launched the Illuminating the Druggable Genome (IDG) initiative in 2014. In this article, we discuss how the systematic collection and processing of a wide array of genomic, proteomic, chemical and disease-related resource data by the IDG Knowledge Management Center have enabled the development of evidence-based criteria for tracking the target development level (TDL) of human proteins, which indicates a substantial knowledge deficit for approximately one out of three proteins in the human proteome. We then present spotlights on the TDL categories as well as key drug target classes, including G protein-coupled receptors, protein kinases and ion channels, which illustrate the nature of the unexplored opportunities for biomedical research and therapeutic development.
Collapse
Affiliation(s)
- Tudor I. Oprea
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
- UNM Comprehensive Cancer Center, Albuquerque, NM, USA
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Cristian G. Bologa
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Søren Brunak
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Anna Gaulton
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Shawn M. Gomez
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Rajarshi Guha
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Anne Hersey
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Jayme Holmes
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Lars Juhl Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gary L. Johnson
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Anneli Karlson
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Present addresses: SciBite Limited, BioData Innovation Centre, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Andrew R. Leach
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Avi Ma’ayan
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Subramani Mani
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Stephen L. Mathias
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | | | - Terrence F. Meehan
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Daniel Muthas
- Respiratory, Inflammation and Autoimmunity Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D Gothenburg, Mölndal, Sweden
| | - Dac-Trung Nguyen
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - John P. Overington
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Medicines Discovery Catapult, Alderley Edge, UK
| | - George Papadatos
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- GlaxoSmithKline, Stevenage, UK
| | - Jun Qin
- Baylor College of Medicine, Houston, TX, USA
| | | | - Bryan L. Roth
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Stephan C. Schürer
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Anton Simeonov
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Larry A. Sklar
- UNM Comprehensive Cancer Center, Albuquerque, NM, USA
- Center for Molecular Discovery, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM, USA
- Department of Pathology, University of New Mexico, Albuquerque, NM, USA
| | - Noel Southall
- National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, MD, USA
| | - Susumu Tomita
- Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Ilinca Tudose
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Google Germany GmbH, München, Germany
| | - Oleg Ursu
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Dušica Vidovic
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Anna Waller
- Center for Molecular Discovery, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - David Westergaard
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jeremy J. Yang
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Gergely Zahoránszky-Köhalmi
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM, USA
- NIH-NCATS, Rockville, MD, USA
| |
Collapse
|
180
|
Pogodin PV, Lagunin AA, Rudik AV, Filimonov DA, Druzhilovskiy DS, Nicklaus MC, Poroikov VV. How to Achieve Better Results Using PASS-Based Virtual Screening: Case Study for Kinase Inhibitors. Front Chem 2018; 6:133. [PMID: 29755970 PMCID: PMC5935003 DOI: 10.3389/fchem.2018.00133] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/09/2018] [Indexed: 12/16/2022] Open
Abstract
Discovery of new pharmaceutical substances is currently boosted by the possibility of utilization of the Synthetically Accessible Virtual Inventory (SAVI) library, which includes about 283 million molecules, each annotated with a proposed synthetic one-step route from commercially available starting materials. The SAVI database is well-suited for ligand-based methods of virtual screening to select molecules for experimental testing. In this study, we compare the performance of three approaches for the analysis of structure-activity relationships that differ in their criteria for selecting of "active" and "inactive" compounds included in the training sets. PASS (Prediction of Activity Spectra for Substances), which is based on a modified Naïve Bayes algorithm, was applied since it had been shown to be robust and to provide good predictions of many biological activities based on just the structural formula of a compound even if the information in the training set is incomplete. We used different subsets of kinase inhibitors for this case study because many data are currently available on this important class of drug-like molecules. Based on the subsets of kinase inhibitors extracted from the ChEMBL 20 database we performed the PASS training, and then applied the model to ChEMBL 23 compounds not yet present in ChEMBL 20 to identify novel kinase inhibitors. As one may expect, the best prediction accuracy was obtained if only the experimentally confirmed active and inactive compounds for distinct kinases in the training procedure were used. However, for some kinases, reasonable results were obtained even if we used merged training sets, in which we designated as inactives the compounds not tested against the particular kinase. Thus, depending on the availability of data for a particular biological activity, one may choose the first or the second approach for creating ligand-based computational tools to achieve the best possible results in virtual screening.
Collapse
Affiliation(s)
- Pavel V. Pogodin
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Alexey A. Lagunin
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
- Department of Bioinformatics, Medical-Biological Department, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anastasia V. Rudik
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
| | - Dmitry A. Filimonov
- Department of Bioinformatics, Institute of Biomedical Chemistry, Moscow, Russia
| | | | - Mark C. Nicklaus
- Computer-Aided Drug Design Group, Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, NIH, NCI-Frederick, Frederick, MD, United States
| | | |
Collapse
|
181
|
Huang LC, Ross KE, Baffi TR, Drabkin H, Kochut KJ, Ruan Z, D'Eustachio P, McSkimming D, Arighi C, Chen C, Natale DA, Smith C, Gaudet P, Newton AC, Wu C, Kannan N. Integrative annotation and knowledge discovery of kinase post-translational modifications and cancer-associated mutations through federated protein ontologies and resources. Sci Rep 2018; 8:6518. [PMID: 29695735 PMCID: PMC5916945 DOI: 10.1038/s41598-018-24457-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 03/23/2018] [Indexed: 11/09/2022] Open
Abstract
Many bioinformatics resources with unique perspectives on the protein landscape are currently available. However, generating new knowledge from these resources requires interoperable workflows that support cross-resource queries. In this study, we employ federated queries linking information from the Protein Kinase Ontology, iPTMnet, Protein Ontology, neXtProt, and the Mouse Genome Informatics to identify key knowledge gaps in the functional coverage of the human kinome and prioritize understudied kinases, cancer variants and post-translational modifications (PTMs) for functional studies. We identify 32 functional domains enriched in cancer variants and PTMs and generate mechanistic hypotheses on overlapping variant and PTM sites by aggregating information at the residue, protein, pathway and species level from these resources. We experimentally test the hypothesis that S768 phosphorylation in the C-helix of EGFR is inhibitory by showing that oncogenic variants altering S768 phosphorylation increase basal EGFR activity. In contrast, oncogenic variants altering conserved phosphorylation sites in the ‘hydrophobic motif’ of PKCβII (S660F and S660C) are loss-of-function in that they reduce kinase activity and enhance membrane translocation. Our studies provide a framework for integrative, consistent, and reproducible annotation of the cancer kinomes.
Collapse
Affiliation(s)
- Liang-Chin Huang
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Karen E Ross
- Protein Information Resource (PIR), Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | - Timothy R Baffi
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | | | - Krzysztof J Kochut
- Department of Computer Science, University of Georgia, Athens, GA, 30602, USA
| | - Zheng Ruan
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA
| | - Peter D'Eustachio
- Department of Biochemistry & Molecular Pharmacology, NYU School of Medicine, New York, NY, 10016, USA
| | - Daniel McSkimming
- Genome, Environment, and Microbiome (GEM) Center of Excellence, University at Buffalo, Buffalo, NY, 14203, USA
| | - Cecilia Arighi
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, 19711, USA
| | - Chuming Chen
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, 19711, USA
| | - Darren A Natale
- Protein Information Resource (PIR), Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20007, USA
| | | | - Pascale Gaudet
- SIB Swiss Institute of Bioinformatics, Lausanne, 1015, Switzerland
| | - Alexandra C Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Cathy Wu
- Protein Information Resource (PIR), Department of Biochemistry and Molecular & Cellular Biology, Georgetown University Medical Center, Washington, DC, 20007, USA.,Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, 19711, USA
| | - Natarajan Kannan
- Institute of Bioinformatics, University of Georgia, Athens, GA, 30602, USA.
| |
Collapse
|
182
|
Cannon DC, Yang JJ, Mathias SL, Ursu O, Mani S, Waller A, Schürer SC, Jensen LJ, Sklar LA, Bologa CG, Oprea TI. TIN-X: target importance and novelty explorer. Bioinformatics 2018; 33:2601-2603. [PMID: 28398460 PMCID: PMC5870731 DOI: 10.1093/bioinformatics/btx200] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 04/06/2017] [Indexed: 11/14/2022] Open
Abstract
Motivation The increasing amount of peer-reviewed manuscripts requires the development of specific mining tools to facilitate the visual exploration of evidence linking diseases and proteins. Results We developed TIN-X, the Target Importance and Novelty eXplorer, to visualize the association between proteins and diseases, based on text mining data processed from scientific literature. In the current implementation, TIN-X supports exploration of data for G-protein coupled receptors, kinases, ion channels, and nuclear receptors. TIN-X supports browsing and navigating across proteins and diseases based on ontology classes, and displays a scatter plot with two proposed new bibliometric statistics: Importance and Novelty. Availability and Implementation http://www.newdrugtargets.org. Contact cbologa@salud.unm.edu.
Collapse
Affiliation(s)
- Daniel C Cannon
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Jeremy J Yang
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Stephen L Mathias
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Oleg Ursu
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Subramani Mani
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Anna Waller
- UNM Center for Molecular Discovery, University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Stephan C Schürer
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Lars Juhl Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen N 2200, Denmark
| | - Larry A Sklar
- UNM Center for Molecular Discovery, University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA.,Department of Pathology, University of New Mexico, NM 87131, USA
| | - Cristian G Bologa
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| | - Tudor I Oprea
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA
| |
Collapse
|
183
|
Collins KA, Stuhlmiller TJ, Zawistowski JS, East MP, Pham TT, Hall CR, Goulet DR, Bevill SM, Angus SP, Velarde SH, Sciaky N, Oprea TI, Graves LM, Johnson GL, Gomez SM. Proteomic analysis defines kinase taxonomies specific for subtypes of breast cancer. Oncotarget 2018; 9:15480-15497. [PMID: 29643987 PMCID: PMC5884642 DOI: 10.18632/oncotarget.24337] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/19/2018] [Indexed: 01/10/2023] Open
Abstract
Multiplexed small molecule inhibitors covalently bound to Sepharose beads (MIBs) were used to capture functional kinases in luminal, HER2-enriched and triple negative (basal-like and claudin-low) breast cancer cell lines and tumors. Kinase MIB-binding profiles at baseline without perturbation proteomically distinguished the four breast cancer subtypes. Understudied kinases, whose disease associations and pharmacology are generally unexplored, were highly represented in MIB-binding taxonomies and are integrated into signaling subnetworks with kinases that have been previously well characterized in breast cancer. Computationally it was possible to define subtypes using profiles of less than 50 of the more than 300 kinases bound to MIBs that included understudied as well as metabolic and lipid kinases. Furthermore, analysis of MIB-binding profiles established potential functional annotations for these understudied kinases. Thus, comprehensive MIBs-based capture of kinases provides a unique proteomics-based method for integration of poorly characterized kinases of the understudied kinome into functional subnetworks in breast cancer cells and tumors that is not possible using genomic strategies. The MIB-binding profiles readily defined subtype-selective differential adaptive kinome reprogramming in response to targeted kinase inhibition, demonstrating how MIB profiles can be used in determining dynamic kinome changes that result in subtype selective phenotypic state changes.
Collapse
Affiliation(s)
- Kyla A.L. Collins
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Timothy J. Stuhlmiller
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Jon S. Zawistowski
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Michael P. East
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Trang T. Pham
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Claire R. Hall
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27514, USA
| | - Daniel R. Goulet
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Samantha M. Bevill
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Steven P. Angus
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Sara H. Velarde
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Noah Sciaky
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Tudor I. Oprea
- Translational Informatics Division, School of Medicine, University of New Mexico, Albuquerque, NM 87106, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM 87131, USA
| | - Lee M. Graves
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Gary L. Johnson
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Shawn M. Gomez
- Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill, NC 27514, USA
| |
Collapse
|
184
|
Tan SK, Jermakowicz A, Mookhtiar AK, Nemeroff CB, Schürer SC, Ayad NG. Drug Repositioning in Glioblastoma: A Pathway Perspective. Front Pharmacol 2018; 9:218. [PMID: 29615902 PMCID: PMC5864870 DOI: 10.3389/fphar.2018.00218] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/27/2018] [Indexed: 12/27/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most malignant primary adult brain tumor. The current standard of care is surgical resection, radiation, and chemotherapy treatment, which extends life in most cases. Unfortunately, tumor recurrence is nearly universal and patients with recurrent glioblastoma typically survive <1 year. Therefore, new therapies and therapeutic combinations need to be developed that can be quickly approved for use in patients. However, in order to gain approval, therapies need to be safe as well as effective. One possible means of attaining rapid approval is repurposing FDA approved compounds for GBM therapy. However, candidate compounds must be able to penetrate the blood-brain barrier (BBB) and therefore a selection process has to be implemented to identify such compounds that can eliminate GBM tumor expansion. We review here psychiatric and non-psychiatric compounds that may be effective in GBM, as well as potential drugs targeting cell death pathways. We also discuss the potential of data-driven computational approaches to identify compounds that induce cell death in GBM cells, enabled by large reference databases such as the Library of Integrated Network Cell Signatures (LINCS). Finally, we argue that identifying pathways dysregulated in GBM in a patient specific manner is essential for effective repurposing in GBM and other gliomas.
Collapse
Affiliation(s)
- Sze Kiat Tan
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, Miami Project to Cure Paralysis, Sylvester Comprehensive Cancer Center, University of Miami Brain Tumor Initiative, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Anna Jermakowicz
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, Miami Project to Cure Paralysis, Sylvester Comprehensive Cancer Center, University of Miami Brain Tumor Initiative, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Adnan K Mookhtiar
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, Miami Project to Cure Paralysis, Sylvester Comprehensive Cancer Center, University of Miami Brain Tumor Initiative, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Charles B Nemeroff
- Department of Psychiatry and Behavioral Sciences and Center on Aging, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Stephan C Schürer
- Department of Molecular Pharmacology, Center for Computational Sciences, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nagi G Ayad
- Department of Psychiatry and Behavioral Sciences, Center for Therapeutic Innovation, Miami Project to Cure Paralysis, Sylvester Comprehensive Cancer Center, University of Miami Brain Tumor Initiative, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
185
|
|
186
|
Fotis C, Antoranz A, Hatziavramidis D, Sakellaropoulos T, Alexopoulos LG. Network-based technologies for early drug discovery. Drug Discov Today 2017; 23:626-635. [PMID: 29294361 DOI: 10.1016/j.drudis.2017.12.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 11/22/2017] [Accepted: 12/20/2017] [Indexed: 02/07/2023]
Abstract
Although the traditional drug discovery approach has led to the development of many successful drugs, the attrition rates remain high. Recent advances in systems-oriented approaches (systems-biology and/or pharmacology) and 'omics technologies has led to a plethora of new computational tools that promise to enable a more-informed and successful implementation of the reductionist, one drug for one target for one disease, approach. These tools, based on biomolecular pathways and interaction networks, offer a systematic approach to unravel the mechanism(s) of a disease and link them to the chemical space and network footprint of a drug. Drug discovery can draw upon this holistic approach to identify the most-promising targets and compounds during the early phases of development.
Collapse
Affiliation(s)
- Chris Fotis
- National Technical University of Athens, Athens, Greece
| | - Asier Antoranz
- National Technical University of Athens, Athens, Greece; Protavio Ltd, Cambridge, UK
| | | | | | | |
Collapse
|
187
|
Blucher AS, Choonoo G, Kulesz-Martin M, Wu G, McWeeney SK. Evidence-Based Precision Oncology with the Cancer Targetome. Trends Pharmacol Sci 2017; 38:1085-1099. [PMID: 28964549 PMCID: PMC5759325 DOI: 10.1016/j.tips.2017.08.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 08/17/2017] [Accepted: 08/29/2017] [Indexed: 11/16/2022]
Abstract
A core tenet of precision oncology is the rational choice of drugs to interact with patient-specific biological targets of interest, but it is currently difficult for researchers to obtain consistent and well-supported target information for pharmaceutical drugs. We review current drug-target interaction resources and critically assess how supporting evidence is handled. We introduce the concept of a unified Cancer Targetome to aggregate drug-target interactions in an evidence-based framework. We discuss current unmet needs and the implications for evidence-based clinical omics. The focus of this review is precision oncology but the discussion is highly relevant to targeted therapies in any area.
Collapse
Affiliation(s)
- Aurora S Blucher
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Gabrielle Choonoo
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Molly Kulesz-Martin
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Department of Dermatology and Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR, USA
| | - Guanming Wu
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Shannon K McWeeney
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA; Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
188
|
Lin Y, Mehta S, Küçük-McGinty H, Turner JP, Vidovic D, Forlin M, Koleti A, Nguyen DT, Jensen LJ, Guha R, Mathias SL, Ursu O, Stathias V, Duan J, Nabizadeh N, Chung C, Mader C, Visser U, Yang JJ, Bologa CG, Oprea TI, Schürer SC. Drug target ontology to classify and integrate drug discovery data. J Biomed Semantics 2017; 8:50. [PMID: 29122012 PMCID: PMC5679337 DOI: 10.1186/s13326-017-0161-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 10/17/2017] [Indexed: 11/12/2022] Open
Abstract
Background One of the most successful approaches to develop new small molecule therapeutics has been to start from a validated druggable protein target. However, only a small subset of potentially druggable targets has attracted significant research and development resources. The Illuminating the Druggable Genome (IDG) project develops resources to catalyze the development of likely targetable, yet currently understudied prospective drug targets. A central component of the IDG program is a comprehensive knowledge resource of the druggable genome. Results As part of that effort, we have developed a framework to integrate, navigate, and analyze drug discovery data based on formalized and standardized classifications and annotations of druggable protein targets, the Drug Target Ontology (DTO). DTO was constructed by extensive curation and consolidation of various resources. DTO classifies the four major drug target protein families, GPCRs, kinases, ion channels and nuclear receptors, based on phylogenecity, function, target development level, disease association, tissue expression, chemical ligand and substrate characteristics, and target-family specific characteristics. The formal ontology was built using a new software tool to auto-generate most axioms from a database while supporting manual knowledge acquisition. A modular, hierarchical implementation facilitate ontology development and maintenance and makes use of various external ontologies, thus integrating the DTO into the ecosystem of biomedical ontologies. As a formal OWL-DL ontology, DTO contains asserted and inferred axioms. Modeling data from the Library of Integrated Network-based Cellular Signatures (LINCS) program illustrates the potential of DTO for contextual data integration and nuanced definition of important drug target characteristics. DTO has been implemented in the IDG user interface Portal, Pharos and the TIN-X explorer of protein target disease relationships. Conclusions DTO was built based on the need for a formal semantic model for druggable targets including various related information such as protein, gene, protein domain, protein structure, binding site, small molecule drug, mechanism of action, protein tissue localization, disease association, and many other types of information. DTO will further facilitate the otherwise challenging integration and formal linking to biological assays, phenotypes, disease models, drug poly-pharmacology, binding kinetics and many other processes, functions and qualities that are at the core of drug discovery. The first version of DTO is publically available via the website http://drugtargetontology.org/, Github (http://github.com/DrugTargetOntology/DTO), and the NCBO Bioportal (http://bioportal.bioontology.org/ontologies/DTO). The long-term goal of DTO is to provide such an integrative framework and to populate the ontology with this information as a community resource. Electronic supplementary material The online version of this article (10.1186/s13326-017-0161-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yu Lin
- Center for Computational Science, University of Miami, Coral Gables, FL, USA
| | - Saurabh Mehta
- Center for Computational Science, University of Miami, Coral Gables, FL, USA.,Department of Applied Chemistry, Delhi Technological University, Delhi, India
| | - Hande Küçük-McGinty
- Center for Computational Science, University of Miami, Coral Gables, FL, USA.,Department of Computer Science, University of Miami, Coral Gables, FL, USA
| | - John Paul Turner
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Dusica Vidovic
- Center for Computational Science, University of Miami, Coral Gables, FL, USA.,Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Michele Forlin
- Center for Computational Science, University of Miami, Coral Gables, FL, USA.,Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Amar Koleti
- Center for Computational Science, University of Miami, Coral Gables, FL, USA
| | - Dac-Trung Nguyen
- National Center for Advancing Translational Science, Rockville, MD, USA
| | - Lars Juhl Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rajarshi Guha
- National Center for Advancing Translational Science, Rockville, MD, USA
| | - Stephen L Mathias
- Department of Internal Medicine, Translational Informatics Division, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Oleg Ursu
- Department of Internal Medicine, Translational Informatics Division, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Vasileios Stathias
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Jianbin Duan
- Center for Computational Science, University of Miami, Coral Gables, FL, USA.,Department of Computer Science, University of Miami, Coral Gables, FL, USA
| | - Nooshin Nabizadeh
- Center for Computational Science, University of Miami, Coral Gables, FL, USA
| | - Caty Chung
- Center for Computational Science, University of Miami, Coral Gables, FL, USA
| | - Christopher Mader
- Center for Computational Science, University of Miami, Coral Gables, FL, USA
| | - Ubbo Visser
- Department of Computer Science, University of Miami, Coral Gables, FL, USA
| | - Jeremy J Yang
- Department of Internal Medicine, Translational Informatics Division, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Cristian G Bologa
- Department of Internal Medicine, Translational Informatics Division, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Tudor I Oprea
- Department of Internal Medicine, Translational Informatics Division, University of New Mexico School of Medicine, Albuquerque, NM, USA.
| | - Stephan C Schürer
- Center for Computational Science, University of Miami, Coral Gables, FL, USA. .,Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
189
|
Sam E, Athri P. Web-based drug repurposing tools: a survey. Brief Bioinform 2017; 20:299-316. [DOI: 10.1093/bib/bbx125] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Elizabeth Sam
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| | - Prashanth Athri
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| |
Collapse
|
190
|
Benstead-Hume G, Wooller SK, Pearl FM. Computational Approaches to Identify Genetic Interactions for Cancer Therapeutics. J Integr Bioinform 2017; 14:/j/jib.2017.14.issue-3/jib-2017-0027/jib-2017-0027.xml. [PMID: 28941356 PMCID: PMC6042820 DOI: 10.1515/jib-2017-0027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/28/2017] [Accepted: 08/10/2017] [Indexed: 12/17/2022] Open
Abstract
The development of improved cancer therapies is frequently cited as an urgent unmet medical need. Here we describe how genetic interactions are being therapeutically exploited to identify novel targeted treatments for cancer. We discuss the current methodologies that use 'omics data to identify genetic interactions, in particular focusing on synthetic sickness lethality (SSL) and synthetic dosage lethality (SDL). We describe the experimental and computational approaches undertaken both in humans and model organisms to identify these interactions. Finally we discuss some of the identified targets with licensed drugs, inhibitors in clinical trials or with compounds under development.
Collapse
|
191
|
Boezio B, Audouze K, Ducrot P, Taboureau O. Network-based Approaches in Pharmacology. Mol Inform 2017; 36. [PMID: 28692140 DOI: 10.1002/minf.201700048] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/21/2017] [Indexed: 12/23/2022]
Abstract
In drug discovery, network-based approaches are expected to spotlight our understanding of drug action across multiple layers of information. On one hand, network pharmacology considers the drug response in the context of a cellular or phenotypic network. On the other hand, a chemical-based network is a promising alternative for characterizing the chemical space. Both can provide complementary support for the development of rational drug design and better knowledge of the mechanisms underlying the multiple actions of drugs. Recent progress in both concepts is discussed here. In addition, a network-based approach using drug-target-therapy data is introduced as an example.
Collapse
Affiliation(s)
- Baptiste Boezio
- Université Paris Diderot - Inserm UMR-S973, MTi, 75205, Paris Cedex 13, 75013, Paris, France
| | - Karine Audouze
- Université Paris Diderot - Inserm UMR-S973, MTi, 75205, Paris Cedex 13, 75013, Paris, France
| | - Pierre Ducrot
- Institut de Recherche Servier, 125 Chemin de Ronde, 78290, Croissy-sur-Seine, France
| | - Olivier Taboureau
- Université Paris Diderot - Inserm UMR-S973, MTi, 75205, Paris Cedex 13, 75013, Paris, France
| |
Collapse
|
192
|
Bioinformatics in translational drug discovery. Biosci Rep 2017; 37:BSR20160180. [PMID: 28487472 PMCID: PMC6448364 DOI: 10.1042/bsr20160180] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 05/04/2017] [Accepted: 05/08/2017] [Indexed: 12/31/2022] Open
Abstract
Bioinformatics approaches are becoming ever more essential in translational drug discovery both in academia and within the pharmaceutical industry. Computational exploitation of the increasing volumes of data generated during all phases of drug discovery is enabling key challenges of the process to be addressed. Here, we highlight some of the areas in which bioinformatics resources and methods are being developed to support the drug discovery pipeline. These include the creation of large data warehouses, bioinformatics algorithms to analyse 'big data' that identify novel drug targets and/or biomarkers, programs to assess the tractability of targets, and prediction of repositioning opportunities that use licensed drugs to treat additional indications.
Collapse
|
193
|
Nowotka MM, Gaulton A, Mendez D, Bento AP, Hersey A, Leach A. Using ChEMBL web services for building applications and data processing workflows relevant to drug discovery. Expert Opin Drug Discov 2017; 12:757-767. [PMID: 28602100 DOI: 10.1080/17460441.2017.1339032] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
INTRODUCTION ChEMBL is a manually curated database of bioactivity data on small drug-like molecules, used by drug discovery scientists. Among many access methods, a REST API provides programmatic access, allowing the remote retrieval of ChEMBL data and its integration into other applications. This approach allows scientists to move from a world where they go to the ChEMBL web site to search for relevant data, to one where ChEMBL data can be simply integrated into their everyday tools and work environment. Areas covered: This review highlights some of the audiences who may benefit from using the ChEMBL API, and the goals they can address, through the description of several use cases. The examples cover a team communication tool (Slack), a data analytics platform (KNIME), batch job management software (Luigi) and Rich Internet Applications. Expert opinion: The advent of web technologies, cloud computing and micro services oriented architectures have made REST APIs an essential ingredient of modern software development models. The widespread availability of tools consuming RESTful resources have made them useful for many groups of users. The ChEMBL API is a valuable resource of drug discovery bioactivity data for professional chemists, chemistry students, data scientists, scientific and web developers.
Collapse
Affiliation(s)
- Michał M Nowotka
- a European Molecular Biology Laboratory - European Bioinformatics Institute , Wellcome Genome Campus , Hinxton , UK
| | - Anna Gaulton
- a European Molecular Biology Laboratory - European Bioinformatics Institute , Wellcome Genome Campus , Hinxton , UK
| | - David Mendez
- a European Molecular Biology Laboratory - European Bioinformatics Institute , Wellcome Genome Campus , Hinxton , UK
| | - A Patricia Bento
- a European Molecular Biology Laboratory - European Bioinformatics Institute , Wellcome Genome Campus , Hinxton , UK
| | - Anne Hersey
- a European Molecular Biology Laboratory - European Bioinformatics Institute , Wellcome Genome Campus , Hinxton , UK
| | - Andrew Leach
- a European Molecular Biology Laboratory - European Bioinformatics Institute , Wellcome Genome Campus , Hinxton , UK
| |
Collapse
|
194
|
Reisdorf WC, Chhugani N, Sanseau P, Agarwal P. Harnessing public domain data to discover and validate therapeutic targets. Expert Opin Drug Discov 2017; 12:687-693. [DOI: 10.1080/17460441.2017.1329296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- William C. Reisdorf
- Computational Biology, Target Sciences, GlaxoSmithKline R&D, King of Prussia, PA, USA
| | - Neha Chhugani
- Jacobs School of Engineering, University of California San Diego, Belle Mead, NJ, USA
| | - Philippe Sanseau
- Computational Biology, Target Sciences, GlaxoSmithKline R&D, Hertfordshire, UK
| | - Pankaj Agarwal
- Computational Biology, Target Sciences, GlaxoSmithKline R&D, King of Prussia, PA, USA
| |
Collapse
|
195
|
Gaulton A, Hersey A, Nowotka M, Bento AP, Chambers J, Mendez D, Mutowo P, Atkinson F, Bellis LJ, Cibrián-Uhalte E, Davies M, Dedman N, Karlsson A, Magariños MP, Overington JP, Papadatos G, Smit I, Leach AR. The ChEMBL database in 2017. Nucleic Acids Res 2016; 45:D945-D954. [PMID: 27899562 PMCID: PMC5210557 DOI: 10.1093/nar/gkw1074] [Citation(s) in RCA: 1405] [Impact Index Per Article: 175.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 10/21/2016] [Accepted: 10/30/2016] [Indexed: 11/14/2022] Open
Abstract
ChEMBL is an open large-scale bioactivity database (https://www.ebi.ac.uk/chembl), previously described in the 2012 and 2014 Nucleic Acids Research Database Issues. Since then, alongside the continued extraction of data from the medicinal chemistry literature, new sources of bioactivity data have also been added to the database. These include: deposited data sets from neglected disease screening; crop protection data; drug metabolism and disposition data and bioactivity data from patents. A number of improvements and new features have also been incorporated. These include the annotation of assays and targets using ontologies, the inclusion of targets and indications for clinical candidates, addition of metabolic pathways for drugs and calculation of structural alerts. The ChEMBL data can be accessed via a web-interface, RDF distribution, data downloads and RESTful web-services.
Collapse
Affiliation(s)
- Anna Gaulton
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Anne Hersey
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Michał Nowotka
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - A Patrícia Bento
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Jon Chambers
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - David Mendez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Prudence Mutowo
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Francis Atkinson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Louisa J Bellis
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Elena Cibrián-Uhalte
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Mark Davies
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Nathan Dedman
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Anneli Karlsson
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - María Paula Magariños
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK.,Open Targets, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - John P Overington
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - George Papadatos
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Ines Smit
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Andrew R Leach
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| |
Collapse
|