129 results on '"John P. Overington"'
Search Results
2. Target Identification of Mycobacterium tuberculosis Phenotypic Hits Using a Concerted Chemogenomic, Biophysical, and Structural Approach
- Author
-
Grace Mugumbate, Vitor Mendes, Michal Blaszczyk, Mohamad Sabbah, George Papadatos, Joel Lelievre, Lluis Ballell, David Barros, Chris Abell, Tom L. Blundell, and John P. Overington
- Subjects
Mycobacterium tuberculosis ,phenotypic hits ,target identification ,drug resistance ,EthR ,InhA ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Mycobacterium phenotypic hits are a good reservoir for new chemotypes for the treatment of tuberculosis. However, the absence of defined molecular targets and modes of action could lead to failure in drug development. Therefore, a combination of ligand-based and structure-based chemogenomic approaches followed by biophysical and biochemical validation have been used to identify targets for Mycobacterium tuberculosis phenotypic hits. Our approach identified EthR and InhA as targets for several hits, with some showing dual activity against these proteins. From the 35 predicted EthR inhibitors, eight exhibited an IC50 below 50 μM against M. tuberculosis EthR and three were confirmed to be also simultaneously active against InhA. Further hit validation was performed using X-ray crystallography yielding eight new crystal structures of EthR inhibitors. Although the EthR inhibitors attain their activity against M. tuberculosis by hitting yet undefined targets, these results provide new lead compounds that could be further developed to be used to potentiate the effect of EthA activated pro-drugs, such as ethionamide, thus enhancing their bactericidal effect.
- Published
- 2017
- Full Text
- View/download PDF
3. ChEMBL Beaker: A Lightweight Web Framework Providing Robust and Extensible Cheminformatics Services
- Author
-
Michał Nowotka, Mark Davies, George Papadatos, and John P. Overington
- Subjects
web services ,framework ,server ,REST ,API ,open source ,Technology ,Science (General) ,Q1-390 - Abstract
ChEMBL Beaker is an open source web framework, exposing a versatile chemistry-focused API (Application Programming Interface) to support the development of new cheminformatics applications. This paper describes the current functionality offered by Beaker and outlines the future technology roadmap.
- Published
- 2014
- Full Text
- View/download PDF
4. MyChEMBL: A Virtual Platform for Distributing Cheminformatics Tools and Open Data
- Author
-
Mark Davies, Michał Nowotka, George Papadatos, Francis Atkinson, Gerard J. P. van Westen, Nathan Dedman, Rodrigo Ochoa, and John P. Overington
- Subjects
virtual machine ,ChEMBL ,open data ,RDKit ,web services ,Technology ,Science (General) ,Q1-390 - Abstract
MyChEMBL is an open virtual platform which provides a free, secure, standardised and easy to use chemoinformatics environment for bioactivity data mining, machine learning, application development, learning and teaching. The main technical features of myChEMBL along with its applications and future plans are discussed here.
- Published
- 2014
- Full Text
- View/download PDF
5. Data from Repurposing Vandetanib plus Everolimus for the Treatment of ACVR1-Mutant Diffuse Intrinsic Pontine Glioma
- Author
-
Chris Jones, Fernando Carceller, Angel M. Carcaboso, Sabine Mueller, Andres Morales La Madrid, Ofelia Cruz, Michael Hubank, Jacques Grill, Jeremy Pryce, Ashirwad Merve, Safa Al-Sarraj, Bassel Zebian, Andrew Mackinnon, David Sheppard, Anne Phelan, John P. Overington, Alan Mackay, Sara Temelso, Florence I. Raynaud, Akos Pal, Adam Donovan, Ruth Ruddle, Daniel P. Smith, Elizabeth A. Corley, Valeria Molinari, Cinzia Lavarino, Reda Stankunaite, Nagore Olaciregui, Peter J. Richardson, and Diana M. Carvalho
- Abstract
Somatic mutations in ACVR1 are found in a quarter of children with diffuse intrinsic pontine glioma (DIPG), but there are no ACVR1 inhibitors licensed for the disease. Using an artificial intelligence–based platform to search for approved compounds for ACVR1-mutant DIPG, the combination of vandetanib and everolimus was identified as a possible therapeutic approach. Vandetanib, an inhibitor of VEGFR/RET/EGFR, was found to target ACVR1 (Kd = 150 nmol/L) and reduce DIPG cell viability in vitro but has limited ability to cross the blood–brain barrier. In addition to mTOR, everolimus inhibited ABCG2 (BCRP) and ABCB1 (P-gp) transporters and was synergistic in DIPG cells when combined with vandetanib in vitro. This combination was well tolerated in vivo and significantly extended survival and reduced tumor burden in an orthotopic ACVR1-mutant patient-derived DIPG xenograft model. Four patients with ACVR1-mutant DIPG were treated with vandetanib plus an mTOR inhibitor, informing the dosing and toxicity profile of this combination for future clinical studies.Significance:Twenty-five percent of patients with the incurable brainstem tumor DIPG harbor somatic activating mutations in ACVR1, but there are no approved drugs targeting the receptor. Using artificial intelligence, we identify and validate, both experimentally and clinically, the novel combination of vandetanib and everolimus in these children based on both signaling and pharmacokinetic synergies.This article is highlighted in the In This Issue feature, p. 275
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Figures S1-S3 from Repurposing Vandetanib plus Everolimus for the Treatment of ACVR1-Mutant Diffuse Intrinsic Pontine Glioma
- Author
-
Chris Jones, Fernando Carceller, Angel M. Carcaboso, Sabine Mueller, Andres Morales La Madrid, Ofelia Cruz, Michael Hubank, Jacques Grill, Jeremy Pryce, Ashirwad Merve, Safa Al-Sarraj, Bassel Zebian, Andrew Mackinnon, David Sheppard, Anne Phelan, John P. Overington, Alan Mackay, Sara Temelso, Florence I. Raynaud, Akos Pal, Adam Donovan, Ruth Ruddle, Daniel P. Smith, Elizabeth A. Corley, Valeria Molinari, Cinzia Lavarino, Reda Stankunaite, Nagore Olaciregui, Peter J. Richardson, and Diana M. Carvalho
- Abstract
Supplementary Figures S1-S3
- Published
- 2023
- Full Text
- View/download PDF
7. Scientific Lenses to Support Multiple Views over Linked Chemistry Data.
- Author
-
Colin R. Batchelor, Christian Y. A. Brenninkmeijer, Christine Chichester, Mark Davies, Daniela Digles, Ian Dunlop, Chris T. A. Evelo, Anna Gaulton, Carole A. Goble, Alasdair J. G. Gray, Paul Groth, Lee Harland, Karen Karapetyan, Antonis Loizou, John P. Overington, Steve Pettifer, Jon Steele, Robert Stevens 0001, Valery Tkachenko, Andra Waagmeester, Antony J. Williams, and Egon L. Willighagen
- Published
- 2014
- Full Text
- View/download PDF
8. Repurposing Vandetanib plus Everolimus for the Treatment of ACVR1-Mutant Diffuse Intrinsic Pontine Glioma
- Author
-
Andrew D MacKinnon, Sabine Mueller, Sara Temelso, Ashirwad Merve, Alan Mackay, Reda Stankunaite, Chris Jones, Daniel P. Smith, Safa Al-Sarraj, Andres Morales La Madrid, Jacques Grill, Adam Donovan, Ofelia Cruz, Mike Hubank, David Sheppard, Cinzia Lavarino, Anne Phelan, Bassel Zebian, Valeria Molinari, Angel M. Carcaboso, Diana Carvalho, Jeremy Pryce, Elizabeth A Corley, Fernando Carceller, Ruth Ruddle, Nagore G. Olaciregui, Akos Pal, Florence I. Raynaud, Peter John Richardson, and John P. Overington
- Subjects
Everolimus ,Abcg2 ,biology ,business.industry ,ACVR1 ,Vandetanib ,In vitro ,Oncology ,In vivo ,biology.protein ,Cancer research ,Medicine ,Viability assay ,business ,PI3K/AKT/mTOR pathway ,medicine.drug - Abstract
Somatic mutations in ACVR1 are found in a quarter of children with diffuse intrinsic pontine glioma (DIPG), but there are no ACVR1 inhibitors licensed for the disease. Using an artificial intelligence–based platform to search for approved compounds for ACVR1-mutant DIPG, the combination of vandetanib and everolimus was identified as a possible therapeutic approach. Vandetanib, an inhibitor of VEGFR/RET/EGFR, was found to target ACVR1 (Kd = 150 nmol/L) and reduce DIPG cell viability in vitro but has limited ability to cross the blood–brain barrier. In addition to mTOR, everolimus inhibited ABCG2 (BCRP) and ABCB1 (P-gp) transporters and was synergistic in DIPG cells when combined with vandetanib in vitro. This combination was well tolerated in vivo and significantly extended survival and reduced tumor burden in an orthotopic ACVR1-mutant patient-derived DIPG xenograft model. Four patients with ACVR1-mutant DIPG were treated with vandetanib plus an mTOR inhibitor, informing the dosing and toxicity profile of this combination for future clinical studies. Significance: Twenty-five percent of patients with the incurable brainstem tumor DIPG harbor somatic activating mutations in ACVR1, but there are no approved drugs targeting the receptor. Using artificial intelligence, we identify and validate, both experimentally and clinically, the novel combination of vandetanib and everolimus in these children based on both signaling and pharmacokinetic synergies. This article is highlighted in the In This Issue feature, p. 275
- Published
- 2021
- Full Text
- View/download PDF
9. Artificial intelligence, drug repurposing and peer review
- Author
-
Sagie Davidovich, Tudor I. Oprea, Alex Zhavoronkov, John P. Overington, Jeremy M Levin, Thomas Clozel, Charles R. Cantor, Evelyne Bischof, and Quentin Vanhaelen
- Subjects
0303 health sciences ,2019-20 coronavirus outbreak ,Coronavirus disease 2019 (COVID-19) ,Computer science ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Biomedical Engineering ,MEDLINE ,Bioengineering ,Context (language use) ,Applied Microbiology and Biotechnology ,Data science ,03 medical and health sciences ,Drug repositioning ,0302 clinical medicine ,Pandemic ,Molecular Medicine ,Computational analysis ,030217 neurology & neurosurgery ,030304 developmental biology ,Biotechnology - Abstract
Can traditional computational analysis and machine learning help compensate for inadequate peer review of drug-repurposing papers in the context of an infodemic?
- Published
- 2020
- Full Text
- View/download PDF
10. Semantic-Web Access to Patent Annotations.
- Author
-
Anna Gaulton, Lee Harland, Mark Davies, George Papadatos, Antonis Loizou, Nathan Dedman, Daniela Digles, Stian Soiland-Reyes, Valery Tkachenko, Stefan Senger, John P. Overington, and Nick Lynch
- Published
- 2015
11. Investigating the Potential Anti-Viral Effects of Proton Pump Inhibitors on Influenza: Intention-to-Treat Trial Emulation Using Electronic Health Records
- Author
-
Caroline Dale, Rohan Takhar, Michail Katsoulis, Valerie Kuan Po Ai, Sheng-Chia Chung, Rui Providencia, John P. Overington, and Reecha Sofat
- Subjects
History ,Polymers and Plastics ,Business and International Management ,Industrial and Manufacturing Engineering - Published
- 2022
- Full Text
- View/download PDF
12. Improving the odds of drug development success through human genomics: modelling study
- Author
-
John P. Overington, Spiros Denaxas, David Prieto, Felix A. Kruger, Raymond J. MacAllister, Aroon D. Hingorani, Harry Hemingway, Anna Gaulton, Reecha Sofat, Valerie Kuan, Sandesh Chopade, Juan P Casas, and Chris Finan
- Subjects
False discovery rate ,Druggability ,lcsh:Medicine ,Genome-wide association study ,Genomics ,Disease ,Computational biology ,Biology ,Article ,External validity ,03 medical and health sciences ,0302 clinical medicine ,Medical research ,Drug Development ,Target identification ,Humans ,lcsh:Science ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,lcsh:R ,3. Good health ,Drug development ,lcsh:Q ,030217 neurology & neurosurgery ,Type I and type II errors ,Genome-Wide Association Study - Abstract
Lack of efficacy in the intended disease indication is the major cause of clinical phase drug development failure. Explanations could include the poor external validity of pre-clinical (cell, tissue, and animal) models of human disease and the high false discovery rate (FDR) in preclinical science. FDR is related to the proportion of true relationships available for discovery (γ), and the type 1 (false-positive) and type 2 (false negative) error rates of the experiments designed to uncover them. We estimated the FDR in preclinical science, its effect on drug development success rates, and improvements expected from use of human genomics rather than preclinical studies as the primary source of evidence for drug target identification. Calculations were based on a sample space defined by all human diseases – the ‘disease-ome’ – represented as columns; and all protein coding genes – ‘the protein-coding genome’– represented as rows, producing a matrix of unique gene- (or protein-) disease pairings. We parameterised the space based on 10,000 diseases, 20,000 protein-coding genes, 100 causal genes per disease and 4000 genes encoding druggable targets, examining the effect of varying the parameters and a range of underlying assumptions, on the inferences drawn. We estimated γ, defined mathematical relationships between preclinical FDR and drug development success rates, and estimated improvements in success rates based on human genomics (rather than orthodox preclinical studies). Around one in every 200 protein-disease pairings was estimated to be causal (γ = 0.005) giving an FDR in preclinical research of 92.6%, which likely makes a major contribution to the reported drug development failure rate of 96%. Observed success rate was only slightly greater than expected for a random pick from the sample space. Values for γ back-calculated from reported preclinical and clinical drug development success rates were also close to the a priori estimates. Substituting genome wide (or druggable genome wide) association studies for preclinical studies as the major information source for drug target identification was estimated to reverse the probability of late stage failure because of the more stringent type 1 error rate employed and the ability to interrogate every potential druggable target in the same experiment. Genetic studies conducted at much larger scale, with greater resolution of disease end-points, e.g. by connecting genomics and electronic health record data within healthcare systems has the potential to produce radical improvement in drug development success rate.
- Published
- 2019
- Full Text
- View/download PDF
13. Brain, a Library for the OWL2 EL profile.
- Author
-
Samuel Croset, John P. Overington, and Dietrich Rebholz-Schuhmann
- Published
- 2013
14. The Comparison of Structures and Sequences: Alignment, Searching and the Detection of Common Folds.
- Author
-
Mark S. Johnson, John P. Overington, Yvonne Edwards, Alex C. W. May, and Michael A. Rodionov
- Published
- 1994
15. EXTH-46. ARTIFICIAL INTELLIGENCE-BASED IDENTIFICATION OF COMBINED VANDETANIB AND EVEROLIMUS IN THE TREATMENT OF ACVR1-MUTANT DIFFUSE INTRINSIC PONTINE GLIOMA
- Author
-
Andrew D MacKinnon, Chris Jones, Andres Morales La Madrid, Ofelia Cruz, Peter D. Richardson, Nagore G. Olaciregui, Akos Pal, Cinzia Lavarino, Sabine Mueller, Valeria Molinari, Diana Carvalho, Adam Donovan, Reda Stankunaite, Angel M. Carcaboso, Elizabeth A Corley, Dave Sheppard, Fernando Carceller, Ruth Ruddle, Mike Hubank, Florence I. Raynaud, Anne Phelan, and John P. Overington
- Subjects
Cancer Research ,Everolimus ,Oncology ,business.industry ,Mutant ,Cancer research ,Medicine ,Neurology (clinical) ,ACVR1 ,business ,Vandetanib ,Preclinical Experimental Therapeutics ,medicine.drug - Abstract
Somatic mutations in ACVR1, encoding the serine/threonine kinase ALK2 receptor, are found in a quarter of children with the currently incurable brain tumour diffuse intrinsic pontine glioma (DIPG). Treatment of ACVR1-mutant DIPG patient-derived models with multiple inhibitor chemotypes leads to a reduction in cell viability in vitro and extended survival in orthotopic xenografts in vivo, though there are currently no specific ACVR1 inhibitors licensed for DIPG. Using an Artificial Intelligence-based platform to search for approved compounds which could be used to treat ACVR1-mutant DIPG, the combination of vandetanib and everolimus was identified as a possible therapeutic approach. Vandetanib, an approved inhibitor of VEGFR/RET/EGFR, was found to target ACVR1 (Kd=150nM) and reduce DIPG cell viability in vitro, but has been trialed in DIPG patients with limited success, in part due to an inability to cross the blood-brain-barrier. In addition to mTOR, everolimus inhibits both ABCG2 (BCRP) and ABCB1 (P-gp) transporter, and was synergistic in DIPG cells when combined with vandetanib in vitro. This combination is well-tolerated in vivo, and significantly extended survival and reduced tumour burden in an orthotopic ACVR1-mutant patient-derived DIPG xenograft model. Based on these preclinical data, three patients with ACVR1-mutant DIPG were treated with vandetanib and everolimus. These cases may inform on the dosing and the toxicity profile of this combination for future clinical studies. This bench-to-bedside approach represents a rapidly translatable therapeutic strategy in children with ACVR1 mutant DIPG.
- Published
- 2020
16. Artificial intelligence, drug repurposing and peer review
- Author
-
Jeremy M, Levin, Tudor I, Oprea, Sagie, Davidovich, Thomas, Clozel, John P, Overington, Quentin, Vanhaelen, Charles R, Cantor, Evelyne, Bischof, and Alex, Zhavoronkov
- Subjects
Artificial Intelligence ,Drug Repositioning ,Computational Biology ,Humans ,Pandemics ,COVID-19 Drug Treatment - Published
- 2020
17. Setting our sights on infectious diseases
- Author
-
Jen Southern, Maria Jose Lafuente-Monasterio, Pottage John C, Caitlin Taylor, William W. Hope, Srinivasa P. S. Rao, Didier Leroy, Jennifer Keiser, Sarah Cook, Clifton E. Barry, John P. Overington, Paul Herrling, Shyam Sundar, Olena Moshynets, David Horn, Lynn L. Silver, Valerie Mizrahi, Manu De Rycker, Isabela Ribeiro, Thomas Spangenberg, Ujjini H. Manjunatha, Nathalie Gobeau, Paul D. Leeson, Jacquin C. Niles, Nicholas J. White, Kevin D. Read, Susan Wyllie, Timothy J. Miles, Richard Amewu, Ian H. Gilbert, Frederick S. Buckner, Wesley C. Van Voorhis, James S. McCarthy, Paul G. Wyatt, Jennifer Herrmann, Michael A. J. Ferguson, Bree B. Aldridge, and HIPS, Helmholtz-Institut für Pharmazeutische Forschung Saarland, Universitätscampus E8.1 66123 Saarbrücken, Germany.
- Subjects
0301 basic medicine ,030106 microbiology ,education ,MEDLINE ,Drug Evaluation, Preclinical ,HIV Infections ,Disease ,Communicable Diseases ,03 medical and health sciences ,Viewpoint ,Drug Discovery ,Medicine ,Humans ,Poverty ,Medical education ,business.industry ,Congresses as Topic ,Combined Modality Therapy ,United Kingdom ,3. Good health ,Sight ,030104 developmental biology ,Infectious Diseases ,Low and middle income countries ,Communicable Disease Control ,Neglected tropical diseases ,business - Abstract
In May 2019, the Wellcome Centre for Anti-Infectives Research (; W; CAIR) at the University of Dundee, UK, held an international conference with the aim of discussing some key questions around discovering new medicines for infectious diseases and a particular focus on diseases affecting Low and Middle Income Countries. There is an urgent need for new drugs to treat most infectious diseases. We were keen to see if there were lessons that we could learn across different disease areas and between the preclinical and clinical phases with the aim of exploring how we can improve and speed up the drug discovery, translational, and clinical development processes. We started with an introductory session on the current situation and then worked backward from clinical development to combination therapy, pharmacokinetic/pharmacodynamic (PK/PD) studies, drug discovery pathways, and new starting points and targets. This Viewpoint aims to capture some of the learnings.
- Published
- 2020
18. Brain, Biomedical Knowledge Manipulation.
- Author
-
Samuel Croset, John P. Overington, and Dietrich Rebholz-Schuhmann
- Published
- 2013
19. The Functional Therapeutic Chemical Classification System.
- Author
-
Samuel Croset, John P. Overington, and Dietrich Rebholz-Schuhmann
- Published
- 2013
20. Pharos: Collating protein information to shed light on the druggable genome
- Author
-
Rajarshi Guha, Anne Hersey, Nicolas F. Fernandez, Anna Waller, Lars Juhl Jensen, Juhee Patel, John P. Overington, Larry A. Sklar, Geetha Mandava, Dac-Trung Nguyen, Ajit Jadhav, Søren Brunak, Saurabh Mehta, Jeremy J. Yang, Subramani Mani, Noel Southall, Timothy Sheils, Guixia Liu, Dusica Vidovic, Cristian Bologa, Anneli Karlsson, Anna Gaulton, Avi Ma'ayan, Stephan C. Schürer, Tudor I. Oprea, Jayme Holmes, Oleg Ursu, Andrew D. Rouillard, Stephen L. Mathias, and Anton Simeonov
- Subjects
0301 basic medicine ,Interface (Java) ,Druggability ,Genomics ,Biology ,Web Browser ,Genome ,World Wide Web ,03 medical and health sciences ,0302 clinical medicine ,Resource (project management) ,Databases, Genetic ,Drug Discovery ,Genetics ,Database Issue ,Cluster Analysis ,Humans ,Use case ,Obesity ,Computational Biology ,Search Engine ,030104 developmental biology ,Pharmacogenetics ,User interface ,Relevant information ,030217 neurology & neurosurgery ,Software - Abstract
The 'druggable genome' encompasses several protein families, but only a subset of targets within them have attracted significant research attention and thus have information about them publicly available. The Illuminating the Druggable Genome (IDG) program was initiated in 2014, has the goal of developing experimental techniques and a Knowledge Management Center (KMC) that would collect and organize information about protein targets from four families, representing the most common druggable targets with an emphasis on understudied proteins. Here, we describe two resources developed by the KMC: the Target Central Resource Database (TCRD) which collates many heterogeneous gene/protein datasets and Pharos (https://pharos.nih.gov), a multimodal web interface that presents the data from TCRD. We briefly describe the types and sources of data considered by the KMC and then highlight features of the Pharos interface designed to enable intuitive access to the IDG knowledgebase. The aim of Pharos is to encourage 'serendipitous browsing', whereby related, relevant information is made easily discoverable. We conclude by describing two use cases that highlight the utility of Pharos and TCRD.
- Published
- 2016
21. Drug discovery and development in the era of Big Data
- Author
-
John P. Overington, Pat Walters, Jürgen Bajorath, and Jeremy L. Jenkins
- Subjects
0301 basic medicine ,Pharmacology ,business.industry ,Drug discovery ,Computer science ,Big data ,MEDLINE ,Data science ,03 medical and health sciences ,030104 developmental biology ,Informatics ,Drug Discovery ,Molecular Medicine ,business - Published
- 2016
- Full Text
- View/download PDF
22. Crowdsourcing Twitter annotations to identify first-hand experiences of prescription drug use
- Author
-
John P. Overington, Nestor Alvaro, Christoph Lofi, Mike Conway, Nigel Collier, and Son Doan
- Subjects
Prescription drug ,Twitter ,Bayesian probability ,Health Informatics ,Fleiss' kappa ,Machine learning ,computer.software_genre ,Crowdsourcing ,Drug Prescriptions ,Pharmacovigilance ,Feature (machine learning) ,Humans ,Medicine ,Social media ,Set (psychology) ,business.industry ,Natural language processing ,3. Good health ,Computer Science Applications ,Identification (information) ,Artificial intelligence ,business ,Social Media ,computer - Abstract
Display Omitted Study on the automatic identification of first-hand drug intake reports in Twitter.Crowd-sourced judgements compared against expert judgements show moderate agreement.The experimental set up used 6 machine learning models with 7 different feature sets.Bayesian Generalized Linear Model performs the best (F1=0.64 and Informedness=0.43). Self-reported patient data has been shown to be a valuable knowledge source for post-market pharmacovigilance. In this paper we propose using the popular micro-blogging service Twitter to gather evidence about adverse drug reactions (ADRs) after firstly having identified micro-blog messages (also know as tweets) that report first-hand experience. In order to achieve this goal we explore machine learning with data crowdsourced from laymen annotators. With the help of lay annotators recruited from CrowdFlower we manually annotated 1548 tweets containing keywords related to two kinds of drugs: SSRIs (eg. Paroxetine), and cognitive enhancers (eg. Ritalin). Our results show that inter-annotator agreement (Fleiss kappa) for crowdsourcing ranks in moderate agreement with a pair of experienced annotators (Spearmans Rho=0.471). We utilized the gold standard annotations from CrowdFlower for automatically training a range of supervised machine learning models to recognize first-hand experience. F-Score values are reported for 6 of these techniques with the Bayesian Generalized Linear Model being the best (F-Score=0.64 and Informedness=0.43) when combined with a selected set of features obtained by using information gain criteria.
- Published
- 2015
- Full Text
- View/download PDF
23. Unexplored therapeutic opportunities in the human genome
- Author
-
Christian Reich, Christian von Mering, Søren Brunak, Jeremy J. Yang, Anna Malovannaya, Lars Juhl Jensen, Rajarshi Guha, Avi Ma'ayan, Jun Qin, Gary L. Johnson, Susumu Tomita, Daniel Muthas, Michael T. McManus, Anton Simeonov, David Westergaard, Noel Southall, Jayme Holmes, Andrew R. Leach, Ajit Jadhav, John P. Overington, George Papadatos, Dusica Vidovic, Cristian Bologa, Allen Campbell, Stephan C. Schürer, Stephen L. Mathias, Gergely Zahoránszky-Köhalmi, Gregory N. Gan, Tudor I. Oprea, Shawn M. Gomez, Ilinca Tudose, Anne Hersey, Subramani Mani, Bryan L. Roth, Dac-Trung Nguyen, Terrence F. Meehan, Anneli Karlson, Oleg Ursu, Anna Waller, Larry A. Sklar, and Anna Gaulton
- Subjects
0301 basic medicine ,Pharmacology ,Extramural ,Drug target ,Druggability ,General Medicine ,Computational biology ,Biology ,Genome ,Article ,03 medical and health sciences ,030104 developmental biology ,Drug Discovery ,Human proteome project ,Human genome ,Knowledge deficit ,Human proteins - 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. © 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
- Published
- 2018
24. Drug Target Commons: A Community Effort to Build a Consensus Knowledge Base for Drug-Target Interactions
- Author
-
John P. Overington, Prson Gautam, Balaguru Ravikumar, Markus Vähä-Koskela, Abhishekh Gupta, Alok Jaiswal, Liye He, Suleiman A. Khan, Gopal Peddinti, Brinton Seashore-Ludlow, Andrew R. Leach, Laxman Yetukuri, Zaid Alam, Mehreen Ali, Arjan J. van Adrichem, Gretchen A. Repasky, Elina Parri, Krister Wennerberg, Anne Hersey, Anna-Lena Gustavsson, Ella Karjalainen, Tero Aittokallio, Anni Rebane, Ziaurrehman Tanoli, Janica Wakkinen, Jing Tang, Institute for Molecular Medicine Finland, Medicum, University of Helsinki, Computational Systems Medicine, Krister Wennerberg / Principal Investigator, Tero Aittokallio / Principal Investigator, and Bioinformatics
- Subjects
0301 basic medicine ,community effort ,Standardization ,INFORMATION ,Knowledge Bases ,bioassay annotation ,Clinical Biochemistry ,open data ,Reuse ,Biochemistry ,0302 clinical medicine ,Drug Discovery ,BINDING ,TOOL ,Drug Interactions ,drug repurposing ,Drug discovery ,cheminformatics ,CANCER ,Drug repositioning ,Open data ,Pharmaceutical Preparations ,Knowledge base ,Cheminformatics ,030220 oncology & carcinogenesis ,Molecular Medicine ,Consensus ,DATABASE ,chemical biology ,drug repositioning ,Biology ,Article ,drug discovery ,PROBES ,03 medical and health sciences ,Humans ,Molecular Biology ,data curation ,Pharmacology ,ta112 ,Data curation ,business.industry ,ta111 ,Drug Repositioning ,ta1182 ,Data science ,crowd sourcing ,030104 developmental biology ,DISCOVERY ,1182 Biochemistry, cell and molecular biology ,3111 Biomedicine ,business - Abstract
Summary Knowledge of the full target space of bioactive substances, approved and investigational drugs as well as chemical probes, provides important insights into therapeutic potential and possible adverse effects. The existing compound-target bioactivity data resources are often incomparable due to non-standardized and heterogeneous assay types and variability in endpoint measurements. To extract higher value from the existing and future compound target-profiling data, we implemented an open-data web platform, named Drug Target Commons (DTC), which features tools for crowd-sourced compound-target bioactivity data annotation, standardization, curation, and intra-resource integration. We demonstrate the unique value of DTC with several examples related to both drug discovery and drug repurposing applications and invite researchers to join this community effort to increase the reuse and extension of compound bioactivity data., Graphical Abstract, Highlights • DTC is a crowd-sourcing-based web platform to annotate drug-target bioactivity data • The open environment improves data harmonization for drug repurposing applications • DTC offers a comprehensive, reproducible, and sustainable bioactivity knowledge base, Tang et al. launches a novel crowd-sourcing effort to standardize the collection, management, curation, and annotation of the notoriously heterogeneous compound-target bioactivity measurements. The web-based community platform aims to provide the most comprehensive, reproducible, and sustainable bioactivity knowledge base for the end users.
- Published
- 2018
- Full Text
- View/download PDF
25. SureChEMBL: a large-scale, chemically annotated patent document database
- Author
-
John P. Overington, Anne Hersey, Sean A. Irvine, Nicholas T. Goncharoff, James Siddle, Nathan Dedman, Joe Pettersson, Richard Koks, Jon Chambers, George Papadatos, Anna Gaulton, and Mark Davies
- Subjects
0301 basic medicine ,Structure (mathematical logic) ,Database ,Interface (Java) ,Scale (chemistry) ,Biology ,computer.software_genre ,Pipeline (software) ,Patents as Topic ,Set (abstract data type) ,03 medical and health sciences ,030104 developmental biology ,Resource (project management) ,Pharmaceutical Preparations ,Chemical science ,Genetics ,Data Mining ,Database Issue ,computer ,Patent document ,Databases, Chemical - Abstract
SureChEMBL is a publicly available large-scale resource containing compounds extracted from the full text, images and attachments of patent documents. The data are extracted from the patent literature according to an automated text and image-mining pipeline on a daily basis. SureChEMBL provides access to a previously unavailable, open and timely set of annotated compound-patent associations, complemented with sophisticated combined structure and keyword-based search capabilities against the compound repository and patent document corpus; given the wealth of knowledge hidden in patent documents, analysis of SureChEMBL data has immediate applications in drug discovery, medicinal chemistry and other commercial areas of chemical science. Currently, the database contains 17 million compounds extracted from 14 million patent documents. Access is available through a dedicated web-based interface and data downloads at: https://www.surechembl.org/.
- Published
- 2015
- Full Text
- View/download PDF
26. The relationship between target-class and the physicochemical properties of antibacterial drugs
- Author
-
John P. Overington and Grace Mugumbate
- Subjects
Drug targets ,Physicochemical properties ,Clinical Biochemistry ,Pharmaceutical Science ,Computational biology ,Pharmacology ,Biochemistry ,Article ,Polar surface area ,Bacterial protein ,Bacterial Proteins ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Binding site ,Molecular Biology ,ComputingMethodologies_COMPUTERGRAPHICS ,Medicine(all) ,Bacteria ,Drug discovery ,Chemistry ,Organic Chemistry ,Bacterial Infections ,chEMBL ,Ribosome ,Chemical space ,Anti-Bacterial Agents ,3. Good health ,Mechanism of action ,Molecular Medicine ,Antibacterials ,medicine.symptom - Abstract
Graphical abstract, The discovery of novel mechanism of action (MOA) antibacterials has been associated with the concept that antibacterial drugs occupy a differentiated region of physicochemical space compared to human-targeted drugs. With, in broad terms, antibacterials having higher molecular weight, lower log P and higher polar surface area (PSA). By analysing the physicochemical properties of about 1700 approved drugs listed in the ChEMBL database, we show, that antibacterials for whose targets are riboproteins (i.e., composed of a complex of RNA and protein) fall outside the conventional human ‘drug-like’ chemical space; whereas antibacterials that modulate bacterial protein targets, generally comply with the ‘rule-of-five’ guidelines for classical oral human drugs. Our analysis suggests a strong target-class association for antibacterials—either protein-targeted or riboprotein-targeted. There is much discussion in the literature on the failure of screening approaches to deliver novel antibacterial lead series, and linkage of this poor success rate for antibacterials with the chemical space properties of screening collections. Our analysis suggests that consideration of target-class may be an underappreciated factor in antibacterial lead discovery, and that in fact bacterial protein-targets may well have similar binding site characteristics to human protein targets, and questions the assumption that larger, more polar compounds are a key part of successful future antibacterial discovery.
- Published
- 2015
- Full Text
- View/download PDF
27. ChEMBL web services: streamlining access to drug discovery data and utilities
- Author
-
Mark Davies, Michal Nowotka, John P. Overington, Nathan Dedman, George Papadatos, Anna Gaulton, Louisa J. Bellis, and Francis Atkinson
- Subjects
Internet ,Service (systems architecture) ,business.industry ,Drug discovery ,Resource (Windows) ,Biology ,computer.software_genre ,chEMBL ,3. Good health ,Systems Integration ,World Wide Web ,User-Computer Interface ,Workflow ,Cheminformatics ,Drug Discovery ,Genetics ,Web Server issue ,The Internet ,Web service ,business ,computer ,Databases, Chemical - Abstract
ChEMBL is now a well-established resource in the fields of drug discovery and medicinal chemistry research. The ChEMBL database curates and stores standardized bioactivity, molecule, target and drug data extracted from multiple sources, including the primary medicinal chemistry literature. Programmatic access to ChEMBL data has been improved by a recent update to the ChEMBL web services (version 2.0.x, https://www.ebi.ac.uk/chembl/api/data/docs), which exposes significantly more data from the underlying database and introduces new functionality. To complement the data-focused services, a utility service (version 1.0.x, https://www.ebi.ac.uk/chembl/api/utils/docs), which provides RESTful access to commonly used cheminformatics methods, has also been concurrently developed. The ChEMBL web services can be used together or independently to build applications and data processing workflows relevant to drug discovery and chemical biology.
- Published
- 2015
- Full Text
- View/download PDF
28. Computational and Practical Aspects of Drug Repositioning
- Author
-
John P. Overington and Tudor I. Oprea
- Subjects
Translational bioinformatics ,Computer science ,Drug discovery ,Drug Evaluation, Preclinical ,Drug Repositioning ,MEDLINE ,Computational Biology ,Original Articles ,Bioinformatics ,Evidence level ,Probability of success ,Antimalarials ,Drug repositioning ,Drug Therapy ,Risk analysis (engineering) ,Drug Discovery ,Dysentery, Amebic ,Animals ,Humans ,Molecular Medicine ,Observational study ,Drug Approval ,Barriers to entry - Abstract
The concept of the hypothesis-driven or observational-based expansion of the therapeutic application of drugs is very seductive. This is due to a number of factors, such as lower cost of development, higher probability of success, near-term clinical potential, patient and societal benefit, and also the ability to apply the approach to rare, orphan, and underresearched diseases. Another highly attractive aspect is that the "barrier to entry" is low, at least in comparison to a full drug discovery operation. The availability of high-performance computing, and databases of various forms have also enhanced the ability to pose reasonable and testable hypotheses for drug repurposing, rescue, and repositioning. In this article we discuss several factors that are currently underdeveloped, or could benefit from clearer definition in articles presenting such work. We propose a classification scheme-drug repositioning evidence level (DREL)-for all drug repositioning projects, according to the level of scientific evidence. DREL ranges from zero, which refers to predictions that lack any experimental support, to four, which refers to drugs approved for the new indication. We also present a set of simple concepts that can allow rapid and effective filtering of hypotheses, leading to a focus on those that are most likely to lead to practical safe applications of an existing drug. Some promising repurposing leads for malaria (DREL-1) and amoebic dysentery (DREL-2) are discussed.
- Published
- 2015
- Full Text
- View/download PDF
29. Flipping the odds of drug development success through human genomics
- Author
-
Aroon D. Hingorani, Raymond J. MacAllister, Anna Gaulton, Valerie Kuan, John P. Overington, David Prieto-Merino, Reecha Sofat, Chris Finan, Harry Hemingway, Juan P. Casas, Sandesh Chopade, Felix A. Kruger, and Spiros Denaxas
- Subjects
Drug development ,Druggability ,Genomics ,Human genomics ,Disease ,Biology ,Bioinformatics ,ENCODE ,Odds ,Pharmacological action - Abstract
Drug development depends on accurately identifying molecular targets that both play a causal role in a disease and are amenable to pharmacological action by small molecule drugs or bio-therapeutics, such as monoclonal antibodies.Errors in drug target specification contribute to the extremely high rates of drug development failure.Integrating knowledge of genes that encode druggable targets with those that influence susceptibility to common disease has the potential to radically improve the probability of drug development success.
- Published
- 2017
- Full Text
- View/download PDF
30. PCSK9 monoclonal antibodies for the primary and secondary prevention of cardiovascular disease
- Author
-
Amand F Schmidt, John-Paul L Carter, Lucy S Pearce, John T Wilkins, John P Overington, Aroon D Hingorani, and JP Casas
- Subjects
Time Factors ,Myocardial Infarction ,ISCHEMIC-HEART-DISEASE ,030204 cardiovascular system & hematology ,Cholinergic Antagonists ,law.invention ,0302 clinical medicine ,Randomized controlled trial ,law ,STATISTICS NOTES ,Cause of Death ,Secondary Prevention ,Pharmacology (medical) ,030212 general & internal medicine ,Randomized Controlled Trials as Topic ,AMG 145 ,Anticholesteremic Agents ,PCSK9 Inhibitors ,Absolute risk reduction ,Antibodies, Monoclonal ,RANDOMIZED CONTROLLED-TRIAL ,Middle Aged ,Primary Prevention ,Stroke ,Cardiovascular Diseases ,Meta-analysis ,Proprotein Convertase 9 ,medicine.drug ,Medicine General & Introductory Medical Sciences ,medicine.medical_specialty ,Antibodies, Monoclonal, Humanized ,Placebo ,HETEROZYGOUS FAMILIAL HYPERCHOLESTEROLEMIA ,03 medical and health sciences ,Ezetimibe ,Internal medicine ,medicine ,Humans ,Alirocumab ,LIPOPROTEIN CHOLESTEROL LEVELS ,business.industry ,SUBTILISIN/KEXIN TYPE 9 ,Cholesterol, LDL ,Odds ratio ,GLOBAL BURDEN ,RISK PATIENTS ,Clinical trial ,INHIBITOR ALIROCUMAB ,Hydroxymethylglutaryl-CoA Reductase Inhibitors ,business - Abstract
BACKGROUND: Despite the availability of effective drug therapies that reduce low‐density lipoprotein (LDL)‐cholesterol (LDL‐C), cardiovascular disease (CVD) remains an important cause of mortality and morbidity. Therefore, additional LDL‐C reduction may be warranted, especially for people who are unresponsive to, or unable to take, existing LDL‐C‐reducing therapies. By inhibiting the proprotein convertase subtilisin/kexin type 9 (PCSK9) enzyme, monoclonal antibodies (PCSK9 inhibitors) reduce LDL‐C and CVD risk. OBJECTIVES: Primary To quantify the effects of PCSK9 inhibitors on CVD, all‐cause mortality, myocardial infarction, and stroke, compared to placebo or active treatment(s) for primary and secondary prevention. Secondary To quantify the safety of PCSK9 inhibitors, with specific focus on the incidence of influenza, hypertension, type 2 diabetes, and cancer, compared to placebo or active treatment(s) for primary and secondary prevention. SEARCH METHODS: We identified studies by systematically searching CENTRAL, MEDLINE, Embase, and Web of Science in December 2019. We also searched ClinicalTrials.gov and the International Clinical Trials Registry Platform in August 2020 and screened the reference lists of included studies. This is an update of the review first published in 2017. SELECTION CRITERIA: All parallel‐group and factorial randomised controlled trials (RCTs) with a follow‐up of at least 24 weeks and adult participants with or without a history of CVD were eligible if they compared PCSK9 inhibitors alirocumab or evolocumab to placebo or active treatments such as statins, ezetimibe, or a combination of these. DATA COLLECTION AND ANALYSIS: Two review authors independently reviewed and extracted data. Where data were available, we calculated pooled effect estimates. We used GRADE to assess certainty of evidence and in 'Summary of findings' tables. MAIN RESULTS: We included 24 studies with data on 60,997 participants. Eighteen trials randomised participants to alirocumab and six to evolocumab. All participants received background lipid‐lowering treatment or lifestyle counselling. Six alirocumab studies used an active treatment comparison group (the remaining used placebo), compared to three evolocumab active comparison trials. Follow‐up ranged from 6 to 36 months for the comparisons with placebo and from 6 to 12 months for comparisons with active treatment. Most of the available studies preferentially enrolled people with either established CVD or at a high risk already, and evidence in low‐ to medium‐risk settings is minimal. Alirocumab compared with placebo decreased the risk of CVD events, with an absolute risk difference (RD) of –2% (odds ratio (OR) 0.87, 95% confidence interval (CI) 0.80 to 0.94; 10 studies, 23,868 participants; high‐certainty evidence), decreased the risk of mortality (RD –1%; OR 0.83, 95% CI 0.72 to 0.96; 12 studies, 24,797 participants; high‐certainty evidence), and MI (RD –2%; OR 0.86, 95% CI 0.79 to 0.94; 9 studies, 23,352 participants; high‐certainty evidence) and for any stroke (RD 0%; OR 0.73, 95% CI 0.58 to 0.91; 8 studies, 22,835 participants; high‐certainty evidence). Alirocumab compared with ezetimibe and statins: for CVD, the RD was 1% (OR 1.37, 95% CI 0.65 to 2.87; 3 studies, 1379 participants; low‐certainty evidence); for mortality, RD was –1% (OR 0.51, 95% CI 0.18 to 1.40; 5 studies, 1333 participants; low‐certainty evidence); for MI, RD was 1% (OR 1.45, 95% CI 0.64 to 3.28, 5 studies, 1734 participants; low‐certainty evidence); and for any stroke, RD was less than 1% (OR 0.85, 95% CI 0.13 to 5.61; 5 studies, 1734 participants; low‐certainty evidence). Evolocumab compared with placebo: for CVD, the RD was –2% (OR 0.84, 95% CI 0.78 to 0.91; 3 studies, 29,432 participants; high‐certainty evidence); for mortality, RD was less than 1% (OR 1.04, 95% CI 0.91 to 1.19; 3 studies, 29,432 participants; high‐certainty evidence); for MI, RD was –1% (OR 0.72, 95% CI 0.64 to 0.82; 3 studies, 29,432 participants; high‐certainty evidence); and for any stroke RD was less than –1% (OR 0.79, 95% CI 0.65 to 0.94; 2 studies, 28,531 participants; high‐certainty evidence). Evolocumab compared with ezetimibe and statins: for any CVD event RD was less than –1% (OR 0.66, 95% CI 0.14 to 3.04; 1 study, 218 participants; very low‐certainty evidence); for all‐cause mortality, the RD was less than 1% (OR 0.43, 95% CI 0.14 to 1.30; 3 studies, 5223 participants; very low‐certainty evidence); and for MI, RD was less than 1% (OR 0.66, 95% CI 0.23 to 1.85; 3 studies, 5003 participants; very low‐certainty evidence). There were insufficient data on any stroke. AUTHORS' CONCLUSIONS: The evidence for the clinical endpoint effects of evolocumab and alirocumab versus placebo were graded as high. There is a strong evidence base for the benefits of PCSK9 monoclonal antibodies to people who might not be eligible for other lipid‐lowering drugs, or to people who cannot meet their lipid goals on more traditional therapies, which was the main patient population of the available trials. The evidence base of PCSK9 inhibitors compared with ezetimibe and statins is much weaker (low very‐ to low‐certainty evidence) and it is unclear whether evolocumab or alirocumab might be effectively used as replacement therapies. Finally, there is very limited evidence on any potential safety issues of both evolocumab and alirocumab. While the current evidence synthesis does not reveal any adverse signals, neither does it provide evidence against such signals. This suggests careful consideration of alternative lipid lowering treatments before prescribing PCSK9 inhibitors.
- Published
- 2017
- Full Text
- View/download PDF
31. Rational design of non-resistant targeted cancer therapies
- Author
-
Marc A. Marti-Renom, John P. Overington, Bissan Al-Lazikani, and Francisco Martínez-Jiménez
- Subjects
0301 basic medicine ,Lung Neoplasms ,Cancer therapy ,Colorectal cancer ,Antineoplastic Agents ,Drug resistance ,Computational biology ,Models, Biological ,Article ,03 medical and health sciences ,Gefitinib ,Drug Delivery Systems ,Carcinoma, Non-Small-Cell Lung ,Medicine ,Humans ,Point Mutation ,EGFR inhibitors ,Multidisciplinary ,business.industry ,Drug discovery ,Point mutation ,Melanoma ,Cancer ,medicine.disease ,Neoplasm Proteins ,030104 developmental biology ,Adenocarcinoma of lung ,Adenocarcinoma ,business ,medicine.drug - Abstract
Drug resistance is one of the major problems in targeted cancer therapy. A major cause of resistance is changes in the amino acids that form the drug-target binding site. Despite of the numerous efforts made to individually understand and overcome these mutations, there is a lack of comprehensive analysis of the mutational landscape that can prospectively estimate drug-resistance mutations. Here we describe and computationally validate a framework that combines the cancer-specific likelihood with the resistance impact to enable the detection of single point mutations with the highest chance to be responsible of resistance to a particular targeted cancer therapy. Moreover, for these treatment-threatening mutations, the model proposes alternative therapies overcoming the resistance. We exemplified the applicability of the model using EGFR-gefitinib treatment for Lung Adenocarcinoma (LUAD) and Lung Squamous Cell Cancer (LSCC) and the ERK2-VTX11e treatment for melanoma and colorectal cancer. Our model correctly identified the phenotype known resistance mutations, including the classic EGFR-T790M and the ERK2-P58L/S/T mutations. Moreover, the model predicted new previously undescribed mutations as potentially responsible of drug resistance. Finally, we provided a map of the predicted sensitivity of alternative ERK2 and EGFR inhibitors, with a particular highlight of two molecules with a low predicted resistance impact. The project was supported by the Spanish MINECO to M.A.M.-R. (BFU2010-19310). We also acknowledge the support of the Spanish Ministry of Economy and Competitiveness, Centro de Excelencia Severo Ochoa 2013-2017 (SEV-2012-0208) and the CERCA Programme of the Generalitat de Catalunya.
- Published
- 2017
- Full Text
- View/download PDF
32. ChEMBL Beaker: A Lightweight Web Framework Providing Robust and Extensible Cheminformatics Services
- Author
-
John P. Overington, Michal Nowotka, Mark Davies, and George Papadatos
- Subjects
Engineering ,jel:A00 ,jel:C00 ,computer.software_genre ,Extensibility ,lcsh:Technology ,World Wide Web ,open source ,Beaker ,framework ,Management of Technology and Innovation ,server ,lcsh:Science (General) ,jel:Z00 ,Application programming interface ,business.industry ,Web application framework ,lcsh:T ,REST ,web services ,API ,chEMBL ,Cheminformatics ,Technology roadmap ,Web service ,Hardware_CONTROLSTRUCTURESANDMICROPROGRAMMING ,business ,computer ,lcsh:Q1-390 - Abstract
ChEMBL Beaker is an open source web framework, exposing a versatile chemistry-focused API (Application Programming Interface) to support the development of new cheminformatics applications. This paper describes the current functionality offered by Beaker and outlines the future technology roadmap.
- Published
- 2014
33. An atlas of genetic influences on human blood metabolites
- Author
-
Elin Grundberg, Nicole Soranzo, John P. Overington, Jeff K. Trimmer, Rita Santos, Vicky Wang, Eric B. Fauman, Lu Chen, Ana M. Valdes, Sally John, Idil Erte, Robert P. Mohney, Craig L. Hyde, Klaudia Walter, Cristina Menni, Gabi Kastenmüller, Melanie Waldenberger, Fabian J. Theis, Louella Vasquez, Michael V. Milburn, M. Julia Brosnan, Jan Krumsiek, Li Xi, Tsun-Po Yang, J. Brent Richards, Christian Gieger, Vincenzo Forgetta, Phoebe M. Roberts, Karsten Suhre, Matthias Arnold, Jie Huang, So-Youn Shin, Daniel Ziemek, Ann-Kristin Petersen, and Tim D. Spector
- Subjects
Adult ,Male ,Adolescent ,Genotype ,Genome-wide association study ,Disease ,Computational biology ,Biology ,Cohort Studies ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Metabolomics ,Germany ,Genetic variation ,Genetics ,Data Mining ,Humans ,Aged ,030304 developmental biology ,Aged, 80 and over ,Internet ,0303 health sciences ,Gene Expression Profiling ,Great Britain ,Computational Biology ,Genetic Variation ,Middle Aged ,Heritability ,Twin study ,United Kingdom ,3. Good health ,Europe ,Gene expression profiling ,Blood ,Drug development ,Genetic Loci ,Female ,Blood Chemical Analysis ,Metabolism, Inborn Errors ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Genome-wide association scans with high-throughput metabolic profiling provide unprecedented insights into how genetic variation influences metabolism and complex disease. Here we report the most comprehensive exploration of genetic loci influencing human metabolism thus far, comprising 7,824 adult individuals from 2 European population studies. We report genome-wide significant associations at 145 metabolic loci and their biochemical connectivity with more than 400 metabolites in human blood. We extensively characterize the resulting in vivo blueprint of metabolism in human blood by integrating it with information on gene expression, heritability and overlap with known loci for complex disorders, inborn errors of metabolism and pharmacological targets. We further developed a database and web-based resources for data mining and results visualization. Our findings provide new insights into the role of inherited variation in blood metabolic diversity and identify potential new opportunities for drug development and for understanding disease.
- Published
- 2014
- Full Text
- View/download PDF
34. ‘Big data’ in pharmaceutical science: challenges and opportunities
- Author
-
Al G Dossetter, Hugh Laverty, Gerhard F. Ecker, and John P. Overington
- Subjects
Pharmacology ,Engineering ,Drug Industry ,business.industry ,Chemistry, Pharmaceutical ,Drug Design ,Drug Discovery ,Big data ,Humans ,Molecular Medicine ,business ,Data science ,Databases, Chemical - Abstract
Future Medicinal Chemistry invited a selection of experts to express their views on the current impact of big data in drug discovery and design, as well as speculate on future developments in the field. The topics discussed include the challenges of implementing big data technologies, maintaining the quality and privacy of data sets, and how the industry will need to adapt to welcome the big data era. Their enlightening responses provide a snapshot of the many and varied contributions being made by big data to the advancement of pharmaceutical science.
- Published
- 2014
- Full Text
- View/download PDF
35. The ChEMBL database: a taster for medicinal chemists
- Author
-
John P. Overington and George Papadatos
- Subjects
Pharmacology ,Information retrieval ,Databases, Factual ,Computer science ,Chemistry, Pharmaceutical ,chEMBL ,computer.software_genre ,Structure-Activity Relationship ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,Chemogenomics ,Molecular Medicine ,Data mining ,Protein Kinase Inhibitors ,computer - Published
- 2014
- Full Text
- View/download PDF
36. ADME SARfari: comparative genomics of drug metabolizing systems
- Author
-
John P. Overington, Lourdes Cucurull-Sanchez, Peter J. Eddershaw, George Papadatos, Samiul Hasan, Mark Davies, Phil Jeffrey, Anne Hersey, Matthew D. Hall, and Nathan Dedman
- Subjects
Statistics and Probability ,Drug ,media_common.quotation_subject ,In silico ,Databases and Ontologies ,Genomics ,Computational biology ,Biology ,Bioinformatics ,030226 pharmacology & pharmacy ,Biochemistry ,Polymorphism, Single Nucleotide ,03 medical and health sciences ,0302 clinical medicine ,Dogs ,Animals ,Humans ,Computer Simulation ,Pharmacokinetics ,Tissue Distribution ,Tissue distribution ,Molecular Biology ,030304 developmental biology ,ADME ,media_common ,Comparative genomics ,0303 health sciences ,Internet ,Proteins ,Small molecule ,Applications Notes ,Computer Science Applications ,Computational Mathematics ,Computational Theory and Mathematics ,Pharmacogenetics ,Drug metabolism ,Software - Abstract
Motivation: ADME SARfari is a freely available web resource that enables comparative analyses of drug-disposition genes. It does so by integrating a number of publicly available data sources, which have subsequently been used to build data mining services, predictive tools and visualizations for drug metabolism researchers. The data include the interactions of small molecules with ADME (absorption, distribution, metabolism and excretion) proteins responsible for the metabolism and transport of molecules; available pharmacokinetic (PK) data; protein sequences of ADME-related molecular targets for pre-clinical model species and human; alignments of the orthologues including information on known SNPs (Single Nucleotide Polymorphism) and information on the tissue distribution of these proteins. In addition, in silico models have been developed, which enable users to predict which ADME relevant protein targets a novel compound is likely to interact with. Availability and implementation: https://www.ebi.ac.uk/chembl/admesarfari Contact: jpo@ebi.ac.uk Supplementary information: Supplementary data are available at Bioinformatics online.
- Published
- 2015
37. The ChEMBL bioactivity database: an update
- Author
-
Yvonne Light, Lora Mak, Felix A. Kruger, John P. Overington, Rita Santos, Anne Hersey, A. Patrícia Bento, Mark Davies, George Papadatos, Michal Nowotka, Louisa J. Bellis, Anna Gaulton, Jon Chambers, and Shaun McGlinchey
- Subjects
Interface (Java) ,Biology ,Ligands ,computer.software_genre ,01 natural sciences ,03 medical and health sciences ,Resource (project management) ,Drug Discovery ,Genetics ,Humans ,RDF ,030304 developmental biology ,Internet ,0303 health sciences ,Binding Sites ,Database ,business.industry ,Proteins ,computer.file_format ,chEMBL ,0104 chemical sciences ,3. Good health ,010404 medicinal & biomolecular chemistry ,VI. Genomic variation, diseases and drugs ,Pharmaceutical Preparations ,Data model ,The Internet ,Web service ,BindingDB ,business ,computer ,Databases, Chemical - Abstract
ChEMBL is an open large-scale bioactivity database (https://www.ebi.ac.uk/chembl), previously described in the 2012 Nucleic Acids Research Database Issue. Since then, a variety of new data sources and improvements in functionality have contributed to the growth and utility of the resource. In particular, more comprehensive tracking of compounds from research stages through clinical development to market is provided through the inclusion of data from United States Adopted Name applications; a new richer data model for representing drug targets has been developed; and a number of methods have been put in place to allow users to more easily identify reliable data. Finally, access to ChEMBL is now available via a new Resource Description Framework format, in addition to the web-based interface, data downloads and web services.
- Published
- 2013
- Full Text
- View/download PDF
38. The functional therapeutic chemical classification system
- Author
-
John P. Overington, Samuel Croset, and Dietrich Rebholz-Schuhmann
- Subjects
Statistics and Probability ,Polypharmacology ,Computer science ,Databases and Ontologies ,Chemical classification ,computer.software_genre ,Biochemistry ,chemistry.chemical_compound ,Alzheimer Disease ,Knowledge integration ,Humans ,Molecular Biology ,Internet ,Information retrieval ,Drug Repositioning ,Ontology language ,chEMBL ,Original Papers ,Computer Science Applications ,Computational Mathematics ,Drug repositioning ,Pharmaceutical Preparations ,Computational Theory and Mathematics ,chemistry ,Semantic technology ,Data mining ,computer - Abstract
Motivation: Drug repositioning is the discovery of new indications for compounds that have already been approved and used in a clinical setting. Recently, some computational approaches have been suggested to unveil new opportunities in a systematic fashion, by taking into consideration gene expression signatures or chemical features for instance. We present here a novel method based on knowledge integration using semantic technologies, to capture the functional role of approved chemical compounds. Results: In order to computationally generate repositioning hypotheses, we used the Web Ontology Language to formally define the semantics of over 20 000 terms with axioms to correctly denote various modes of action (MoA). Based on an integration of public data, we have automatically assigned over a thousand of approved drugs into these MoA categories. The resulting new resource is called the Functional Therapeutic Chemical Classification System and was further evaluated against the content of the traditional Anatomical Therapeutic Chemical Classification System. We illustrate how the new classification can be used to generate drug repurposing hypotheses, using Alzheimers disease as a use-case. Availability: https://www.ebi.ac.uk/chembl/ftc; https://github.com/loopasam/ftc. Contact: croset@ebi.ac.uk Supplementary information: Supplementary data are available at Bioinformatics online.
- Published
- 2013
- Full Text
- View/download PDF
39. The druggable genome and support for target identification and validation in drug development
- Author
-
Ryan Kelley, John P. Overington, R. Thomas Lumbers, Aroon D. Hingorani, Felix A. Kruger, Chris Finan, Anneli Karlsson, Rita Santos, Tina Shah, Luana Galver, Jorgen Engmann, Juan P. Casas, and Anna Gaulton
- Subjects
0301 basic medicine ,Drug ,media_common.quotation_subject ,Druggability ,Single-nucleotide polymorphism ,Genome-wide association study ,Disease ,Computational biology ,Biology ,Bioinformatics ,Polymorphism, Single Nucleotide ,Genome ,Linkage Disequilibrium ,Article ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,Mendelian randomization ,Drug Discovery ,Humans ,Molecular Targeted Therapy ,Genotyping ,Repurposing ,030304 developmental biology ,media_common ,0303 health sciences ,Genome, Human ,Drug Repositioning ,Reproducibility of Results ,General Medicine ,030104 developmental biology ,Phenotype ,Drug development ,Genetic Loci ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Target identification (identifying the correct drug targets for each disease) and target validation (demonstrating the effect of target perturbation on disease biomarkers and disease end-points) are essential steps in drug development. We showed previously that biomarker and disease endpoint associations of single nucleotide polymorphisms (SNPs) in a gene encoding a drug target accurately depict the effect of modifying the same target with a pharmacological agent; others have shown that genomic support for a target is associated with a higher rate of drug development success. To delineate drug development (including repurposing) opportunities arising from this paradigm, we connected complex disease- and biomarker-associated loci from genome wide association studies (GWAS) to an updated set of genes encoding druggable human proteins, to compounds with bioactivity against these targets and, where these were licensed drugs, to clinical indications. We used this set of genes to inform the design of a new genotyping array, to enable druggable genome-wide association studies for drug target selection and validation in human disease.
- Published
- 2016
- Full Text
- View/download PDF
40. Structural and Functional View of Polypharmacology
- Author
-
Aurelio Moya García, Tolulope Adeyelu, Felix A Kruger, Natalie L. Dawson, Jon G. Lees, John P. Overington, Christine Orengo, and Juan A.G. Ranea
- Subjects
Drug ,Protein family ,Polypharmacology ,media_common.quotation_subject ,Protein domain ,Druggability ,lcsh:Medicine ,Computational biology ,Bioinformatics ,Article ,Functional networks ,03 medical and health sciences ,0302 clinical medicine ,Sequence Analysis, Protein ,Drug Discovery ,Humans ,Medicine ,lcsh:Science ,Databases, Protein ,030304 developmental biology ,media_common ,0303 health sciences ,Binding Sites ,business.industry ,lcsh:R ,3. Good health ,030220 oncology & carcinogenesis ,Drug side effects ,lcsh:Q ,Identification (biology) ,business ,Algorithms ,Protein Binding - Abstract
Protein domains mediate drug-protein interactions and this principle can guide the design of multi-target drugs i.e. polypharmacology. In this study, we associate multi-target drugs with CATH functional families through the overrepresentation of targets of those drugs in CATH functional families. Thus, we identify CATH functional families that are currently enriched in drugs (druggable CATH functional families) and we use the network properties of these druggable protein families to analyse their association with drug side effects. Analysis of selected druggable CATH functional families, enriched in drug targets, show that relatives exhibit highly conserved drug binding sites. Furthermore, relatives within druggable CATH functional families occupy central positions in a human protein functional network, cluster together forming network neighbourhoods and are less likely to be within proteins associated with drug side effects. Our results demonstrate that CATH functional families can be used to identify drug-target interactions, opening a new research direction in target identification.
- Published
- 2016
- Full Text
- View/download PDF
41. Comprehensive characterization of the Published Kinase Inhibitor Set
- Author
-
David H. Drewry, Eidarus Salah, Joel Morris, John P. Overington, Jowita Mikolajczyk, Francis Atkinson, Alexander Tropsha, Karen Lackey, Jonathan M. Elkins, Kamal R. Abdul Azeez, Mark Kunkel, Nikolai Sepetov, Daniel J. Price, Xi Ping Huang, Eric C. Polley, Timothy M. Willson, Beverly A. Teicher, Vita Fedele, Susanne Müller, Ayman Al Haj Zen, William J. Zuercher, Eugene N. Muratov, Sergei Romanov, Paul Bamborough, Bryan L. Roth, Stefan Knapp, Denis Fourches, and M. Szklarz
- Subjects
0301 basic medicine ,Glycosylation ,Kinase ,Drug discovery ,Angiogenesis ,Phosphotransferases ,Biomedical Engineering ,Bioengineering ,Computational biology ,Biology ,Applied Microbiology and Biotechnology ,law.invention ,03 medical and health sciences ,030104 developmental biology ,Biochemistry ,law ,Cancer cell ,Recombinant DNA ,Molecular Medicine ,Kinome ,Receptor ,Protein kinase A ,Protein Kinase Inhibitors ,Biotechnology - Abstract
Despite the success of protein kinase inhibitors as approved therapeutics, drug discovery has focused on a small subset of kinase targets. Here we provide a thorough characterization of the Published Kinase Inhibitor Set (PKIS), a set of 367 small-molecule ATP-competitive kinase inhibitors that was recently made freely available with the aim of expanding research in this field and as an experiment in open-source target validation. We screen the set in activity assays with 224 recombinant kinases and 24 G protein-coupled receptors and in cellular assays of cancer cell proliferation and angiogenesis. We identify chemical starting points for designing new chemical probes of orphan kinases and illustrate the utility of these leads by developing a selective inhibitor for the previously untargeted kinases LOK and SLK. Our cellular screens reveal compounds that modulate cancer cell growth and angiogenesis in vitro. These reagents and associated data illustrate an efficient way forward to increasing understanding of the historically untargeted kinome.
- Published
- 2016
- Full Text
- View/download PDF
42. Improved large-scale prediction of growth inhibition patterns using the NCI60 cancer cell line panel
- Author
-
Michael Nilges, Andreas Bender, Thérèse E. Malliavin, John P. Overington, Isidro Cortes-Ciriano, Guillaume Bouvier, Gerard J. P. van Westen, Bioinformatique structurale - Structural Bioinformatics, Institut Pasteur [Paris] (IP)-Centre National de la Recherche Scientifique (CNRS), Leiden Academic Center for Drug Research, European Bioinformatics Institute [Hinxton] (EMBL-EBI), EMBL Heidelberg, The Unilever Centre for Molecular Science Informatics - Department of Chemistry [Cambridge, UK], University of Cambridge [UK] (CAM), G.J.P.v.W. and J.P.O. thank the EMBL member states, The Wellcome Trust (WT086151/Z/08/Z) and Marie Curie (COFUND) for funding. I.C.C., T.M. and M.N. thank the CNRS and Institut Pasteur for support. A.B. thanks Unilever and the European Research Commission (Starting Grant ERC-2013-StG 336159 MIXTURE) for funding. M.N. thanks the Investissement d’Avenir Bioinformatics Program (Grant Bip:Bip) and the European Research Commission (Advanced Grant ERC-2011-StG 294809 BayCellS) for funding., ANR-10-BINF-0003,Bip:Bip,Paradigme d'inference bayesienne pour la Biologie structurale in silico(2010), European Project: 336159,EC:FP7:ERC,ERC-2013-StG,MIXTURE(2014), European Project: 294809,EC:FP7:ERC,ERC-2011-ADG_20110310,BAYCELLS(2012), and Institut Pasteur [Paris]-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Statistics and Probability ,Support Vector Machine ,Somatic cell ,Biology ,Models, Biological ,Biochemistry ,chemistry.chemical_compound ,Text mining ,Cell Line, Tumor ,Neoplasms ,microRNA ,Humans ,Bioassay ,proteochemometrics ,Databases, Protein ,Molecular Biology ,Exome sequencing ,Cell Proliferation ,Genetics ,Cell growth ,business.industry ,Systems Biology ,Computational Biology ,bioinformatics ,Original Papers ,3. Good health ,Computer Science Applications ,Computational Mathematics ,Computational Theory and Mathematics ,chemistry ,Pharmacogenetics ,Cell culture ,Biological Assay ,Growth inhibition ,[INFO.INFO-BI]Computer Science [cs]/Bioinformatics [q-bio.QM] ,business - Abstract
Motivation: Recent large-scale omics initiatives have catalogued the somatic alterations of cancer cell line panels along with their pharmacological response to hundreds of compounds. In this study, we have explored these data to advance computational approaches that enable more effective and targeted use of current and future anticancer therapeutics. Results: We modelled the 50% growth inhibition bioassay end-point (GI50) of 17 142 compounds screened against 59 cancer cell lines from the NCI60 panel (941 831 data-points, matrix 93.08% complete) by integrating the chemical and biological (cell line) information. We determine that the protein, gene transcript and miRNA abundance provide the highest predictive signal when modelling the GI50 endpoint, which significantly outperformed the DNA copy-number variation or exome sequencing data (Tukey’s Honestly Significant Difference, P Contact: terez@pasteur.fr; ab454@ac.cam.uk Supplementary information: Supplementary data are available at Bioinformatics online.
- Published
- 2016
- Full Text
- View/download PDF
43. The EBI enzyme portal
- Author
-
Maria Jesus Martin, Henning Hermjakob, John P. Overington, Julius O.B. Jacobsen, Christoph Steinbeck, Jennifer A. Cham, Francis Rowland, Julia D. Fischer, Bijay Jassal, Claire O'Donovan, Hong Cao, Rodrigo Lopez, Paula de Matos, Janet M. Thornton, Joseph Onwubiko, Gemma L. Holliday, Syed Asad Rahman, Mickael Goujon, Sameer Velankar, Gerard J. Kleywegt, and Rafael Alcántara
- Subjects
Internet ,0303 health sciences ,Protein Conformation ,Drug discovery ,Process (engineering) ,business.industry ,End user ,030302 biochemistry & molecular biology ,Articles ,Biology ,Bioinformatics ,Enzymes ,World Wide Web ,User-Computer Interface ,03 medical and health sciences ,Genetics ,Disease ,The Internet ,Information discovery ,Databases, Protein ,business ,030304 developmental biology - Abstract
The availability of comprehensive information about enzymes plays an important role in answering questions relevant to interdisciplinary fields such as biochemistry, enzymology, biofuels, bioengineering and drug discovery. At the EMBL European Bioinformatics Institute, we have developed an enzyme portal (http://www.ebi.ac.uk/enzymeportal) to provide this wealth of information on enzymes from multiple in-house resources addressing particular data classes: protein sequence and structure, reactions, pathways and small molecules. The fact that these data reside in separate databases makes information discovery cumbersome. The main goal of the portal is to simplify this process for end users.
- Published
- 2012
- Full Text
- View/download PDF
44. Minimum information about a bioactive entity (MIABE)
- Author
-
Ian Dix, Christoph Steinbeck, Robert C. Glen, Dominic Clark, Kim E. Hammond-Kosack, Romeena K. Mann, John P. Overington, Janet M. Thornton, Christopher Southan, Anna Gaulton, Mark Forster, Christopher Larminie, Peter Murray-Rust, David S. Wishart, Steve Bryant, Elizabeth Calder, Elena Lo Piparo, Henning Hermjakob, Ola Engkvist, Martin Grigorov, Bissan Al-Lazikani, Sandra Orchard, Nick Lynch, Andrew L. Hopkins, Michael K. Gilson, and Lee Harland
- Subjects
Drug Industry ,Bioactive molecules ,media_common.quotation_subject ,Information Dissemination ,Druggability ,Guidelines as Topic ,Ontology (information science) ,Toxicology ,computer.software_genre ,01 natural sciences ,03 medical and health sciences ,Resource (project management) ,Terminology as Topic ,Drug Discovery ,Animals ,Humans ,Pharmacokinetics ,Quality (business) ,Computational analysis ,Pesticides ,030304 developmental biology ,media_common ,Pharmacology ,0303 health sciences ,Chemistry, Physical ,Drug discovery ,Communication ,Data Collection ,General Medicine ,Data science ,0104 chemical sciences ,3. Good health ,010404 medicinal & biomolecular chemistry ,Pharmaceutical Preparations ,Chemical Industry ,Drug Design ,Data mining ,computer ,Biomarkers - Abstract
Bioactive molecules such as drugs, pesticides and food additives are produced in large numbers by many commercial and academic groups around the world. Enormous quantities of data are generated on the biological properties and quality of these molecules. Access to such data - both on licensed and commercially available compounds, and also on those that fail during development - is crucial for understanding how improved molecules could be developed. For example, computational analysis of aggregated data on molecules that are investigated in drug discovery programmes has led to a greater understanding of the properties of successful drugs. However, the information required to perform these analyses is rarely published, and when it is made available it is often missing crucial data or is in a format that is inappropriate for efficient data-mining. Here, we propose a solution: the definition of reporting guidelines for bioactive entities - the Minimum Information About a Bioactive Entity (MIABE) - which has been developed by representatives of pharmaceutical companies, data resource providers and academic groups.
- Published
- 2011
- Full Text
- View/download PDF
45. Chemogenomics Approaches for Receptor Deorphanization and Extensions of the Chemogenomics Concept to Phenotypic Space
- Author
-
John P. Overington, Jörg K. Wegner, Olaf O. van den Hoven, Andreas Bender, Ad P. IJzerman, Julio E. Peironcely, Gerard J. P. van Westen, Eelke van der Horst, David R. Spring, Warren R. J. D. Galloway, and Herman W. T. van Vlijmen
- Subjects
Virtual screening ,Relation (database) ,Proteins ,Bayes Theorem ,Genomics ,General Medicine ,Computational biology ,Space (commercial competition) ,Biology ,Ligands ,Bioinformatics ,Receptors, G-Protein-Coupled ,Bayes' theorem ,chemistry.chemical_compound ,Phenotype ,chemistry ,Drug Design ,Drug Discovery ,Chemogenomics ,Chemical genetics ,Analysis method - Abstract
Chemogenomic approaches, which link ligand chemistry to bioactivity against targets (and, by extension, to phenotypes) are becoming more and more important due to the increasing number of bioactivity data available both in proprietary databases as well as in the public domain. In this article we review chemogenomics approaches applied in four different domains: Firstly, due to the relationship between protein targets from which an approximate relation between their respective bioactive ligands can be inferred, we investigate the extent to which chemogenomics approaches can be applied to receptor deorphanization. In this case it was found that by using knowledge about active compounds of related proteins, in 93% of all cases enrichment better than random could be obtained. Secondly, we analyze different chemin-formatics analysis methods with respect to their behavior in chemogenomics studies, such as subgraph mining and Baye-sian models. Thirdly, we illustrate how chemogenomics, in its particular flavor of 'proteochemometrics', can be applied to extrapolate bioactivity predictions from given data points to related targets. Finally, we extend the concept of 'chemoge-nomics' approaches, relating ligand chemistry to bioactivity against related targets, into phenotypic space which then falls into the area of 'chemical genomics' and 'chemical genetics'; given that this is very often the desired endpoint of approaches in not only the pharmaceutical industry, but also in academic probe discovery, this is often the endpoint the experimental scientist is most interested in. © 2011 Bentham Science Publishers.
- Published
- 2011
- Full Text
- View/download PDF
46. Rapid Analysis of Pharmacology for Infectious Diseases
- Author
-
John P. Overington, Andrew L. Hopkins, Harvey Rubin, Stephen K. Boyer, G. Richard J. Bickerton, and Ian M. Carruthers
- Subjects
Druggability ,Genomics ,Drug resistance ,Computational biology ,Biology ,Bioinformatics ,Communicable Diseases ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,target identification ,Drug Discovery ,Pandemic ,Chemogenomics ,Animals ,Humans ,druggable genome ,Epidemics ,030304 developmental biology ,0303 health sciences ,Genome ,030306 microbiology ,Drug discovery ,General Medicine ,3. Good health ,chemistry ,Infectious disease (medical specialty) ,Informatics ,Drug Design - Abstract
Pandemic, epidemic and endemic infectious diseases are united by a common problem: how do we rapidly and cost-effectively identify potential pharmacological interventions to treat infections? Given the large number of emerging and neglected infectious diseases and the fact that they disproportionately afflict the poorest members of the global society, new ways of thinking are required to develop high productivity discovery systems that can be applied to a large number of pathogens. The growing availability of parasite genome data provides the basis for developing methods to prioritize, a priori potential drug targets and analyze the pharmacological landscape of an infectious disease. Thus the overall objective of infectious disease informatics is to enable the rapid generation of plausible, novel medical hypotheses of test-able pharmacological experiments, by uncovering undiscovered relationships in the wealth of biomedical literature and databases that were collected for other purposes. In particular our goal is to identify potential drug targets present in a pathogen genome and prioritize which pharmacological experiments are most likely to discover drug-like lead compounds rapidly against a pathogen (i.e. which specific compounds and drug targets should be screened, in which assays and where they can be sourced). An integral part of the challenge is the development and integration of methods to predict druggability, essentiality, synthetic lethality and polypharmocology in pathogen genomes, while simultaneously integrating the inevitable issues of chemical tractability and the potential for acquired drug resistance from the start.
- Published
- 2011
- Full Text
- View/download PDF
47. PPDMs—a resource for mapping small molecule bioactivities from ChEMBL to Pfam-A protein domains
- Author
-
John P. Overington, Felix A. Kruger, Michal Nowotka, and Anna Gaulton
- Subjects
Statistics and Probability ,Protein domain ,Computational biology ,Biology ,computer.software_genre ,Biochemistry ,Domain (software engineering) ,Small Molecule Libraries ,03 medical and health sciences ,0302 clinical medicine ,Protein sequencing ,Drug Discovery ,Humans ,Databases, Protein ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Drug discovery ,Proteins ,chEMBL ,Applications Notes ,Structural Bioinformatics ,Computer Science Applications ,Protein Structure, Tertiary ,Schema (genetic algorithms) ,Computational Mathematics ,Computational Theory and Mathematics ,Data mining ,Small molecule binding ,computer ,030217 neurology & neurosurgery ,Databases, Chemical ,Software - Abstract
Summary: PPDMs is a resource that maps small molecule bioactivities to protein domains from the Pfam-A collection of protein families. Small molecule bioactivities mapped to protein domains add important precision to approaches that use protein sequence searches alignments to assist applications in computational drug discovery and systems and chemical biology. We have previously proposed a mapping heuristic for a subset of bioactivities stored in ChEMBL with the Pfam-A domain most likely to mediate small molecule binding. We have since refined this mapping using a manual procedure. Here, we present a resource that provides up-to-date mappings and the possibility to review assigned mappings as well as to participate in their assignment and curation. We also describe how mappings provided through the PPDMs resource are made accessible through the main schema of the ChEMBL database. Availability and implementation: The PPDMs resource and curation interface is available at https://www.ebi.ac.uk/chembl/research/ppdms/pfam_maps. The source-code for PPDMs is available under the Apache license at https://github.com/chembl/pfam_maps. Source code is available at https://github.com/chembl/pfam_map_loader to demonstrate the integration process with the main schema of ChEMBL. Contact: jpo@ebi.ac.uk
- Published
- 2014
48. Ligand efficiency indices for an effective mapping of chemico-biological space: the concept of an atlas-like representation
- Author
-
Cele Abad-Zapatero, Ognjen Perišić, John Wass, John P. Overington, A. Patrícia Bento, Michael E. Johnson, and Bissan Al-Lazikani
- Subjects
Protein Tyrosine Phosphatase, Non-Receptor Type 1 ,Pharmacology ,Physics ,Binding Sites ,Ligand efficiency ,Ligand ,Stereochemistry ,Drug discovery ,Atlas (topology) ,Core Binding Factors ,Molecular Conformation ,Natural coordinate system ,Ligands ,Chemical space ,Polar surface area ,Molecular Weight ,Structure-Activity Relationship ,Drug Delivery Systems ,HIV Protease ,Drug Discovery ,Polar ,Molecular Targeted Therapy ,Biological system ,Software - Abstract
We propose a numerical framework that permits an effective atlas-like representation of chemico-biological space based on a series of Cartesian planes mapping the ligands with the corresponding targets connected by an affinity parameter (K(i) or related). The numerical framework is derived from the concept of ligand efficiency indices, which provide a natural coordinate system combining the potency toward the target (biological space) with the physicochemical properties of the ligand (chemical space). This framework facilitates navigation in the multidimensional drug discovery space using map-like representations based on pairs of combined variables related to the efficiency of the ligands per Dalton (molecular weight or number of non-hydrogen atoms) and per unit of polar surface area (or number of polar atoms).
- Published
- 2010
- Full Text
- View/download PDF
49. Role of open chemical data in aiding drug discovery and design
- Author
-
John P. Overington and Anna Gaulton
- Subjects
Pharmacology ,Databases, Factual ,Computer science ,Drug discovery ,Drug Design ,Drug Discovery ,Molecular Medicine ,Chemical data ,Data science - Published
- 2010
- Full Text
- View/download PDF
50. Erratum: Unexplored therapeutic opportunities in the human genome
- Author
-
Gary L. Johnson, Susumu Tomita, Dusica Vidovic, Larry A. Sklar, Cristian Bologa, Ilinca Tudose, Anneli Karlson, Bryan L. Roth, Terrence F. Meehan, Anna Gaulton, Daniel Muthas, Jun Qin, Subramani Mani, Gregory N. Gan, Anton Simeonov, Anna Waller, Søren Brunak, Oleg Ursu, Anna Malovannaya, George Papadatos, Steven L. Mathias, Anne Hersey, Christian Reich, Christian von Mering, David Westergaard, Noel Southall, Jayme Holmes, Lars Juhl Jensen, Shawn M. Gomez, Dac-Trung Nguyen, Avi Ma'ayan, Stephan C. Schürer, Tudor I. Oprea, Allen Campbell, Rajarshi Guha, Gergely Zahoránszky-Köhalmi, Ajit Jadhav, Andrew R. Leach, Jeremy J. Yang, Michael T. McManus, and John P. Overington
- Subjects
0301 basic medicine ,Pharmacology ,Data set ,03 medical and health sciences ,030104 developmental biology ,Information retrieval ,Computer science ,Drug discovery ,Drug Discovery ,Table (database) ,Human genome ,General Medicine - Abstract
Nature Reviews Drug Discovery (2018); 10.1038/nrd.2018.14 In the version of this article that was originally published online, an older version of the data set categorizing proteins into target development levels was used to create Figure 1 than the version used to create Table 1, and data from Figure 1 were referred to at several points in the text of the article.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.