1
|
A Fast Parameter Identification Framework for Personalized Pharmacokinetics. Sci Rep 2019; 9:14143. [PMID: 31578414 PMCID: PMC6775128 DOI: 10.1038/s41598-019-50810-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/19/2019] [Indexed: 11/08/2022] Open
Abstract
This paper introduces a novel framework for fast parameter identification of personalized pharmacokinetic problems. Given one sample observation of a new subject, the framework predicts the parameters of the subject based on prior knowledge from a pharmacokinetic database. The feasibility of this framework was demonstrated by developing a new algorithm based on the Cluster Newton method, namely the constrained Cluster Newton method, where the initial points of the parameters are constrained by the database. The algorithm was tested with the compartmental model of propofol on a database of 59 subjects. The average overall absolute percentage error based on constrained Cluster Newton method is 12.10% with the threshold approach, and 13.42% with the nearest-neighbor approach. The average computation time of one estimation is 13.10 seconds. Using parallel computing, the average computation time is reduced to 1.54 seconds, achieved with 12 parallel workers. The results suggest that the proposed framework can effectively improve the prediction accuracy of the pharmacokinetic parameters with limited observations in comparison to the conventional methods. Computation cost analyses indicate that the proposed framework can take advantage of parallel computing and provide solutions within practical response times, leading to fast and accurate parameter identification of pharmacokinetic problems.
Collapse
|
2
|
Natsiavas P, Malousi A, Bousquet C, Jaulent MC, Koutkias V. Computational Advances in Drug Safety: Systematic and Mapping Review of Knowledge Engineering Based Approaches. Front Pharmacol 2019; 10:415. [PMID: 31156424 PMCID: PMC6533857 DOI: 10.3389/fphar.2019.00415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/02/2019] [Indexed: 12/12/2022] Open
Abstract
Drug Safety (DS) is a domain with significant public health and social impact. Knowledge Engineering (KE) is the Computer Science discipline elaborating on methods and tools for developing “knowledge-intensive” systems, depending on a conceptual “knowledge” schema and some kind of “reasoning” process. The present systematic and mapping review aims to investigate KE-based approaches employed for DS and highlight the introduced added value as well as trends and possible gaps in the domain. Journal articles published between 2006 and 2017 were retrieved from PubMed/MEDLINE and Web of Science® (873 in total) and filtered based on a comprehensive set of inclusion/exclusion criteria. The 80 finally selected articles were reviewed on full-text, while the mapping process relied on a set of concrete criteria (concerning specific KE and DS core activities, special DS topics, employed data sources, reference ontologies/terminologies, and computational methods, etc.). The analysis results are publicly available as online interactive analytics graphs. The review clearly depicted increased use of KE approaches for DS. The collected data illustrate the use of KE for various DS aspects, such as Adverse Drug Event (ADE) information collection, detection, and assessment. Moreover, the quantified analysis of using KE for the respective DS core activities highlighted room for intensifying research on KE for ADE monitoring, prevention and reporting. Finally, the assessed use of the various data sources for DS special topics demonstrated extensive use of dominant data sources for DS surveillance, i.e., Spontaneous Reporting Systems, but also increasing interest in the use of emerging data sources, e.g., observational healthcare databases, biochemical/genetic databases, and social media. Various exemplar applications were identified with promising results, e.g., improvement in Adverse Drug Reaction (ADR) prediction, detection of drug interactions, and novel ADE profiles related with specific mechanisms of action, etc. Nevertheless, since the reviewed studies mostly concerned proof-of-concept implementations, more intense research is required to increase the maturity level that is necessary for KE approaches to reach routine DS practice. In conclusion, we argue that efficiently addressing DS data analytics and management challenges requires the introduction of high-throughput KE-based methods for effective knowledge discovery and management, resulting ultimately, in the establishment of a continuous learning DS system.
Collapse
Affiliation(s)
- Pantelis Natsiavas
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece.,Sorbonne Université, INSERM, Univ Paris 13, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France
| | - Andigoni Malousi
- Laboratory of Biological Chemistry, Department of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Cédric Bousquet
- Sorbonne Université, INSERM, Univ Paris 13, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France.,Public Health and Medical Information Unit, University Hospital of Saint-Etienne, Saint-Étienne, France
| | - Marie-Christine Jaulent
- Sorbonne Université, INSERM, Univ Paris 13, Laboratoire d'Informatique Médicale et d'Ingénierie des Connaissances pour la e-Santé, LIMICS, Paris, France
| | - Vassilis Koutkias
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| |
Collapse
|
3
|
Herrero-Zazo M, Segura-Bedmar I, Hastings J, Martínez P. DINTO: Using OWL Ontologies and SWRL Rules to Infer Drug–Drug Interactions and Their Mechanisms. J Chem Inf Model 2015; 55:1698-707. [DOI: 10.1021/acs.jcim.5b00119] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- María Herrero-Zazo
- Department of Computer Science, Universidad Carlos III de Madrid , Leganés 28911, Madrid, Spain
| | - Isabel Segura-Bedmar
- Department of Computer Science, Universidad Carlos III de Madrid , Leganés 28911, Madrid, Spain
| | - Janna Hastings
- Cheminformatics and Metabolism, European Bioinformatics Institute (EMBL-EBI) , Hinxton, CB10 1SD, U.K
| | - Paloma Martínez
- Department of Computer Science, Universidad Carlos III de Madrid , Leganés 28911, Madrid, Spain
| |
Collapse
|
4
|
Brochhausen M, Schneider J, Malone D, Empey PE, Hogan WR, Boyce RD. Towards a foundational representation of potential drug-drug interaction knowledge. CEUR WORKSHOP PROCEEDINGS 2014; 1309:16-31. [PMID: 33139970 PMCID: PMC7603806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Inadequate representation of evidence and knowledge about potential drug-drug interactions is a major factor underlying disagreements among sources of drug information that are used by clinicians. In this paper we describe the initial steps toward developing a foundational domain representation that allows tracing the evidence underlying potential drug-drug interaction knowledge. The new representation includes biological and biomedical entities represented in existing ontologies and terminologies to foster integration of data from relevant fields such as physiology, anatomy, and laboratory sciences.
Collapse
Affiliation(s)
- Mathias Brochhausen
- Division of Biomedical Informatics, University of Arkansas for Medical Sciences, 4301 W. Markham St., #782, Little Rock, AR, 72205-7199
| | - Jodi Schneider
- Web-instrumented man-machine interactions, communities and semantics group, INRIA Sophia Antipolis - Méditerranée, France
| | - Daniel Malone
- College of Pharmacy, University of Arizona, Tucson, AZ, USA
| | - Philip E Empey
- Department of Pharmacy and Therapeutics, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - William R Hogan
- Department of Health Outcomes and Policy, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Richard D Boyce
- Department of Biomedical Informatics, University of Pittsburgh,, Pittsburgh, USA
| |
Collapse
|
5
|
Ji Z, Su J, Liu C, Wang H, Huang D, Zhou X. Integrating genomics and proteomics data to predict drug effects using binary linear programming. PLoS One 2014; 9:e102798. [PMID: 25036040 PMCID: PMC4103865 DOI: 10.1371/journal.pone.0102798] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 06/24/2014] [Indexed: 12/29/2022] Open
Abstract
The Library of Integrated Network-Based Cellular Signatures (LINCS) project aims to create a network-based understanding of biology by cataloging changes in gene expression and signal transduction that occur when cells are exposed to a variety of perturbations. It is helpful for understanding cell pathways and facilitating drug discovery. Here, we developed a novel approach to infer cell-specific pathways and identify a compound's effects using gene expression and phosphoproteomics data under treatments with different compounds. Gene expression data were employed to infer potential targets of compounds and create a generic pathway map. Binary linear programming (BLP) was then developed to optimize the generic pathway topology based on the mid-stage signaling response of phosphorylation. To demonstrate effectiveness of this approach, we built a generic pathway map for the MCF7 breast cancer cell line and inferred the cell-specific pathways by BLP. The first group of 11 compounds was utilized to optimize the generic pathways, and then 4 compounds were used to identify effects based on the inferred cell-specific pathways. Cross-validation indicated that the cell-specific pathways reliably predicted a compound's effects. Finally, we applied BLP to re-optimize the cell-specific pathways to predict the effects of 4 compounds (trichostatin A, MS-275, staurosporine, and digoxigenin) according to compound-induced topological alterations. Trichostatin A and MS-275 (both HDAC inhibitors) inhibited the downstream pathway of HDAC1 and caused cell growth arrest via activation of p53 and p21; the effects of digoxigenin were totally opposite. Staurosporine blocked the cell cycle via p53 and p21, but also promoted cell growth via activated HDAC1 and its downstream pathway. Our approach was also applied to the PC3 prostate cancer cell line, and the cross-validation analysis showed very good accuracy in predicting effects of 4 compounds. In summary, our computational model can be used to elucidate potential mechanisms of a compound's efficacy.
Collapse
Affiliation(s)
- Zhiwei Ji
- Division of Radiologic Sciences - Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, United States of America; School of Electronics and Information Engineering, Tongji University, Shanghai, P.R. China
| | - Jing Su
- Division of Radiologic Sciences - Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, United States of America
| | - Chenglin Liu
- Division of Radiologic Sciences - Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, United States of America
| | - Hongyan Wang
- Division of Radiologic Sciences - Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, United States of America
| | - Deshuang Huang
- School of Electronics and Information Engineering, Tongji University, Shanghai, P.R. China
| | - Xiaobo Zhou
- Division of Radiologic Sciences - Center for Bioinformatics and Systems Biology, Wake Forest School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina, United States of America
| |
Collapse
|
6
|
Yoshida K, Maeda K, Kusuhara H, Konagaya A. Estimation of feasible solution space using Cluster Newton Method: application to pharmacokinetic analysis of irinotecan with physiologically-based pharmacokinetic models. BMC SYSTEMS BIOLOGY 2013; 7 Suppl 3:S3. [PMID: 24555857 PMCID: PMC3852065 DOI: 10.1186/1752-0509-7-s3-s3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Background To facilitate new drug development, physiologically-based pharmacokinetic (PBPK) modeling methods receive growing attention as a tool to fully understand and predict complex pharmacokinetic phenomena. As the number of parameters to reproduce physiological functions tend to be large in PBPK models, efficient parameter estimation methods are essential. We have successfully applied a recently developed algorithm to estimate a feasible solution space, called Cluster Newton Method (CNM), to reveal the cause of irinotecan pharmacokinetic alterations in two cancer patient groups. Results After improvements in the original CNM algorithm to maintain parameter diversities, a feasible solution space was successfully estimated for 55 or 56 parameters in the irinotecan PBPK model, within ten iterations, 3000 virtual samples, and in 15 minutes (Intel Xeon E5-1620 3.60GHz × 1 or Intel Core i7-870 2.93GHz × 1). Control parameters or parameter correlations were clarified after the parameter estimation processes. Possible causes in the irinotecan pharmacokinetic alterations were suggested, but they were not conclusive. Conclusions Application of CNM achieved a feasible solution space by solving inverse problems of a system containing ordinary differential equations (ODEs). This method may give us reliable insights into other complicated phenomena, which have a large number of parameters to estimate, under limited information. It is also helpful to design prospective studies for further investigation of phenomena of interest.
Collapse
|
7
|
Vilar S, Uriarte E, Santana L, Tatonetti NP, Friedman C. Detection of drug-drug interactions by modeling interaction profile fingerprints. PLoS One 2013; 8:e58321. [PMID: 23520498 PMCID: PMC3592896 DOI: 10.1371/journal.pone.0058321] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Accepted: 02/01/2013] [Indexed: 11/19/2022] Open
Abstract
Drug-drug interactions (DDIs) constitute an important problem in postmarketing pharmacovigilance and in the development of new drugs. The effectiveness or toxicity of a medication could be affected by the co-administration of other drugs that share pharmacokinetic or pharmacodynamic pathways. For this reason, a great effort is being made to develop new methodologies to detect and assess DDIs. In this article, we present a novel method based on drug interaction profile fingerprints (IPFs) with successful application to DDI detection. IPFs were generated based on the DrugBank database, which provided 9,454 well-established DDIs as a primary source of interaction data. The model uses IPFs to measure the similarity of pairs of drugs and generates new putative DDIs from the non-intersecting interactions of a pair. We described as part of our analysis the pharmacological and biological effects associated with the putative interactions; for example, the interaction between haloperidol and dicyclomine can cause increased risk of psychosis and tardive dyskinesia. First, we evaluated the method through hold-out validation and then by using four independent test sets that did not overlap with DrugBank. Precision for the test sets ranged from 0.4–0.5 with more than two fold enrichment factor enhancement. In conclusion, we demonstrated the usefulness of the method in pharmacovigilance as a DDI predictor, and created a dataset of potential DDIs, highlighting the etiology or pharmacological effect of the DDI, and providing an exploratory tool to facilitate decision support in DDI detection and patient safety.
Collapse
Affiliation(s)
- Santiago Vilar
- Department of Biomedical Informatics, Columbia University Medical Center, New York, New York, United States of America.
| | | | | | | | | |
Collapse
|
8
|
Luciano JS, Andersson B, Batchelor C, Bodenreider O, Clark T, Denney CK, Domarew C, Gambet T, Harland L, Jentzsch A, Kashyap V, Kos P, Kozlovsky J, Lebo T, Marshall SM, McCusker JP, McGuinness DL, Ogbuji C, Pichler E, Powers RL, Prud'hommeaux E, Samwald M, Schriml L, Tonellato PJ, Whetzel PL, Zhao J, Stephens S, Dumontier M. The Translational Medicine Ontology and Knowledge Base: driving personalized medicine by bridging the gap between bench and bedside. J Biomed Semantics 2011; 2 Suppl 2:S1. [PMID: 21624155 DOI: 10.1186/2041-1480-2-s2-s1] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Translational medicine requires the integration of knowledge using heterogeneous data from health care to the life sciences. Here, we describe a collaborative effort to produce a prototype Translational Medicine Knowledge Base (TMKB) capable of answering questions relating to clinical practice and pharmaceutical drug discovery. RESULTS We developed the Translational Medicine Ontology (TMO) as a unifying ontology to integrate chemical, genomic and proteomic data with disease, treatment, and electronic health records. We demonstrate the use of Semantic Web technologies in the integration of patient and biomedical data, and reveal how such a knowledge base can aid physicians in providing tailored patient care and facilitate the recruitment of patients into active clinical trials. Thus, patients, physicians and researchers may explore the knowledge base to better understand therapeutic options, efficacy, and mechanisms of action. CONCLUSIONS This work takes an important step in using Semantic Web technologies to facilitate integration of relevant, distributed, external sources and progress towards a computational platform to support personalized medicine. AVAILABILITY TMO can be downloaded from http://code.google.com/p/translationalmedicineontology and TMKB can be accessed at http://tm.semanticscience.org/sparql.
Collapse
|
9
|
Caccia S, Garattini S, Pasina L, Nobili A. Predicting the clinical relevance of drug interactions from pre-approval studies. Drug Saf 2009; 32:1017-39. [PMID: 19810775 DOI: 10.2165/11316630-000000000-00000] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Drug interactions (DIs) may result in adverse drug events that could be prevented, but in many cases the available information on potential DIs is not easily transferable to clinical practice. The majority of studies date from preclinical or premarketing phases, using animals or human-derived sources that may not accurately reflect the growing clinical complexity of high-risk populations, such as the elderly, women, children, patients with chronic disease, polytherapy and impaired organ functions. Thus, at the time of approval of a new drug the information in the summary of product characteristics refers to potential DIs, but lacks specific management recommendations and is of limited clinical utility. Therefore, we set out to review in vitro and in vivo methods to predict and quantify potential DIs, to see whether these studies could help the physician tackle daily problems of the assessment and choice of combined drug therapies, and to propose, from a clinical point of view, how premarketing studies could be improved so as to help the physician at the patient's bedside. Preclinical and premarketing study design needs to be improved to make information easily accessible and clinically transferable. Studies should also take into account appropriate sample size, duration, co-morbidity, number of coadministered drugs, within- and between-subject variability, specific at-risk populations and/or drugs with a relatively narrow therapeutic window, and clinical endpoints. After premarketing development in situations where there is potential high risk of serious adverse events, specific phase IV studies (and/or active pharmacovigilance studies) should be required to monitor and quantitatively assess their clinical impact.
Collapse
Affiliation(s)
- Silvio Caccia
- Laboratory of Drug Metabolism, 'Mario Negri' Institute for Pharmacological Research, Milan, Italy
| | | | | | | |
Collapse
|
10
|
Lu G, Ni J. Highlighting computations in bioscience and bioinformatics: review of the Symposium of Computations in Bioinformatics and Bioscience (SCBB07). BMC Bioinformatics 2008; 9 Suppl 6:S1. [PMID: 18541044 PMCID: PMC2423432 DOI: 10.1186/1471-2105-9-s6-s1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The Second Symposium on Computations in Bioinformatics and Bioscience (SCBB07) was held in Iowa City, Iowa, USA, on August 13–15, 2007. This annual event attracted dozens of bioinformatics professionals and students, who are interested in solving emerging computational problems in bioscience, from China, Japan, Taiwan and the United States. The Scientific Committee of the symposium selected 18 peer-reviewed papers for publication in this supplemental issue of BMC Bioinformatics. These papers cover a broad spectrum of topics in computational biology and bioinformatics, including DNA, protein and genome sequence analysis, gene expression and microarray analysis, computational proteomics and protein structure classification, systems biology and machine learning.
Collapse
Affiliation(s)
- Guoqing Lu
- Department of Biology, University of Nebraska, Omaha, NE 68182, USA.
| | | |
Collapse
|