1
|
Zhang Y, Sui X, Pan F, Yu K, Li K, Tian S, Erdengasileng A, Han Q, Wang W, Wang J, Wang J, Sun D, Chung H, Zhou J, Zhou E, Lee B, Zhang P, Qiu X, Zhao T, Zhang J. A comprehensive large scale biomedical knowledge graph for AI powered data driven biomedical research. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.10.13.562216. [PMID: 38168218 PMCID: PMC10760044 DOI: 10.1101/2023.10.13.562216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
To address the rapid growth of scientific publications and data in biomedical research, knowledge graphs (KGs) have become a critical tool for integrating large volumes of heterogeneous data to enable efficient information retrieval and automated knowledge discovery (AKD). However, transforming unstructured scientific literature into KGs remains a significant challenge, with previous methods unable to achieve human-level accuracy. In this study, we utilized an information extraction pipeline that won first place in the LitCoin NLP Challenge (2022) to construct a large-scale KG named iKraph using all PubMed abstracts. The extracted information matches human expert annotations and significantly exceeds the content of manually curated public databases. To enhance the KG's comprehensiveness, we integrated relation data from 40 public databases and relation information inferred from high-throughput genomics data. This KG facilitates rigorous performance evaluation of AKD, which was infeasible in previous studies. We designed an interpretable, probabilistic-based inference method to identify indirect causal relations and applied it to real-time COVID-19 drug repurposing from March 2020 to May 2023. Our method identified 600-1400 candidate drugs per month, with one-third of those discovered in the first two months later supported by clinical trials or PubMed publications. These outcomes are very challenging to attain through alternative approaches that lack a thorough understanding of the existing literature. A cloud-based platform (https://biokde.insilicom.com) was developed for academic users to access this rich structured data and associated tools.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Statistics, Florida State University, Tallahassee, FL 32306
- Insilicom LLC, Tallahassee, FL 32303
| | - Xin Sui
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | - Feng Pan
- Insilicom LLC, Tallahassee, FL 32303
| | | | - Keqiao Li
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | - Shubo Tian
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | | | - Qing Han
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | - Wanjing Wang
- Department of Statistics, Florida State University, Tallahassee, FL 32306
| | | | - Jian Wang
- 977 Wisteria Ter., Sunnyvale, CA 94086
| | | | | | - Jun Zhou
- Insilicom LLC, Tallahassee, FL 32303
| | - Eric Zhou
- Insilicom LLC, Tallahassee, FL 32303
| | - Ben Lee
- Insilicom LLC, Tallahassee, FL 32303
| | - Peili Zhang
- Forward Informatics, Winchester, Massachusetts, 01890
| | - Xing Qiu
- Department of Biostatistics and Computational Biology, University of Rochester, Rochester, NY 14642
| | - Tingting Zhao
- Insilicom LLC, Tallahassee, FL 32303
- Department of Geography, Florida State University, Tallahassee, FL 32306
| | - Jinfeng Zhang
- Department of Statistics, Florida State University, Tallahassee, FL 32306
- Insilicom LLC, Tallahassee, FL 32303
| |
Collapse
|
2
|
Biswal S, Mallick B. Unlocking the potential of signature-based drug repurposing for anticancer drug discovery. Arch Biochem Biophys 2024; 761:110150. [PMID: 39265695 DOI: 10.1016/j.abb.2024.110150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Cancer is the leading cause of death worldwide and is often associated with tumor relapse even after chemotherapeutics. This reveals malignancy is a complex process, and high-throughput omics strategies in recent years have contributed significantly in decoding the molecular mechanisms of these complex events in cancer. Further, the omics studies yield a large volume of cancer-specific molecular signatures that promote the discovery of cancer therapy drugs by a method termed signature-based drug repurposing. The drug repurposing method identifies new uses for approved drugs beyond their intended initial therapeutic use, and there are several approaches to it. In this review, we discuss signature-based drug repurposing in cancer, how cancer omics have revolutionized this method of drug discovery, and how one can use the cancer signature data for repurposed drug identification by providing a step-by-step procedural handout. This modern approach maximizes the use of existing therapeutic agents for cancer therapy or combination therapy to overcome chemotherapeutics resistance, making it a pragmatic and efficient alternative to traditional resource-intensive and time-consuming methods.
Collapse
Affiliation(s)
- Sruti Biswal
- RNAi and Functional Genomics Lab., Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India
| | - Bibekanand Mallick
- RNAi and Functional Genomics Lab., Department of Life Science, National Institute of Technology Rourkela, Rourkela, 769008, Odisha, India.
| |
Collapse
|
3
|
Israr J, Alam S, Kumar A. System biology approaches for drug repurposing. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 205:221-245. [PMID: 38789180 DOI: 10.1016/bs.pmbts.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Drug repurposing, or drug repositioning, refers to the identification of alternative therapeutic applications for established medications that go beyond their initial indications. This strategy has becoming increasingly popular since it has the potential to significantly reduce the overall costs of drug development by around $300 million. System biology methodologies have been employed to facilitate medication repurposing, encompassing computational techniques such as signature matching and network-based strategies. These techniques utilize pre-existing drug-related data types and databases to find prospective repurposed medications that have minimal or acceptable harmful effects on patients. The primary benefit of medication repurposing in comparison to drug development lies in the fact that approved pharmaceuticals have already undergone multiple phases of clinical studies, thereby possessing well-established safety and pharmacokinetic properties. Utilizing system biology methodologies in medication repurposing offers the capacity to expedite the discovery of viable candidates for drug repurposing and offer novel perspectives for structure-based drug design.
Collapse
Affiliation(s)
- Juveriya Israr
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Barabanki, Uttar Pradesh, India; Department of Biotechnology Era University, Lucknow, Uttar Pradesh, India
| | - Shabroz Alam
- Department of Biotechnology Era University, Lucknow, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Mandhana, Kanpur, Uttar Pradesh, India.
| |
Collapse
|
4
|
González-Durruthy M, Rial R, Ruso JM. Decoding the conformational binding of drug mixtures on ovalbumin: An integrated multimodal network. Int J Biol Macromol 2024; 261:129866. [PMID: 38302030 DOI: 10.1016/j.ijbiomac.2024.129866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
This research addresses the crucial necessity for a deeper understanding of the binding interactions between surfactants and proteins, with a specific focus on ovalbumin. Considering ovalbumin's role in diverse biochemical processes, it remains a subject of significant interest for drug discovery and design. To fill existing knowledge gaps, we investigated the binding interaction between dicloxacillin and cetyltrimethylammonium bromide (CTAB) on ovalbumin, employing a comprehensive approach that combines computational modeling with experimental validations. Using the ezPocket tool, the computational phase predicted ten relevant binding sites on ovalbumin's surface. The isobologram combination index (CI) heatmap strongly suggested a complex interplay of antagonistic and synergistic effects. Besides, a conformational drug-drug interaction network was proposed to explore the stability of the surfactant mixture within specific binding sites of ovalbumin, revealing a dynamic landscape of suggested antagonist effects. Experimental validations through UV-vis, Fluorescence, and circular dichroism (CD) spectroscopy further corroborated the computational findings, confirming the formation of stable complexes. Finally, this study not only advances our comprehension of ovalbumin's interactions with surfactants but also offers a multidimensional perspective and an advanced methodological framework for efficient therapeutic strategies, opening new avenues for future applications in drug development and applied biochemistry.
Collapse
Affiliation(s)
- Michael González-Durruthy
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics and Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; NanoSafety Group, International Iberian Nanotechnology Laboratory, Braga 4715-330, Portugal.
| | - Ramón Rial
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics and Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Juan M Ruso
- Soft Matter and Molecular Biophysics Group, Department of Applied Physics and Institute of Materials (iMATUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
5
|
Boudin M, Diallo G, Drancé M, Mougin F. The OREGANO knowledge graph for computational drug repurposing. Sci Data 2023; 10:871. [PMID: 38057380 PMCID: PMC10700660 DOI: 10.1038/s41597-023-02757-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/16/2023] [Indexed: 12/08/2023] Open
Abstract
Drug repositioning is a faster and more affordable solution than traditional drug discovery approaches. From this perspective, computational drug repositioning using knowledge graphs is a very promising direction. Knowledge graphs constructed from drug data and information can be used to generate hypotheses (molecule/drug - target links) through link prediction using machine learning algorithms. However, it remains rare to have a holistically constructed knowledge graph using the broadest possible features and drug characteristics, which is freely available to the community. The OREGANO knowledge graph aims at filling this gap. The purpose of this paper is to present the OREGANO knowledge graph, which includes natural compounds related data. The graph was developed from scratch by retrieving data directly from the knowledge sources to be integrated. We therefore designed the expected graph model and proposed a method for merging nodes between the different knowledge sources, and finally, the data were cleaned. The knowledge graph, as well as the source codes for the ETL process, are openly available on the GitHub of the OREGANO project ( https://gitub.u-bordeaux.fr/erias/oregano ).
Collapse
Affiliation(s)
- Marina Boudin
- AHeaD team, Bordeaux Population Health Inserm U1219, Univ. Bordeaux, F-33000, Bordeaux, France.
| | - Gayo Diallo
- AHeaD team, Bordeaux Population Health Inserm U1219, Univ. Bordeaux, F-33000, Bordeaux, France
| | - Martin Drancé
- AHeaD team, Bordeaux Population Health Inserm U1219, Univ. Bordeaux, F-33000, Bordeaux, France
| | - Fleur Mougin
- AHeaD team, Bordeaux Population Health Inserm U1219, Univ. Bordeaux, F-33000, Bordeaux, France
| |
Collapse
|
6
|
Flanary VL, Fisher JL, Wilk EJ, Howton TC, Lasseigne BN. Computational Advancements in Cancer Combination Therapy Prediction. JCO Precis Oncol 2023; 7:e2300261. [PMID: 37824797 PMCID: PMC12012855 DOI: 10.1200/po.23.00261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/20/2023] [Accepted: 08/15/2023] [Indexed: 10/14/2023] Open
Abstract
Given the high attrition rate of de novo drug discovery and limited efficacy of single-agent therapies in cancer treatment, combination therapy prediction through in silico drug repurposing has risen as a time- and cost-effective alternative for identifying novel and potentially efficacious therapies for cancer. The purpose of this review is to provide an introduction to computational methods for cancer combination therapy prediction and to summarize recent studies that implement each of these methods. A systematic search of the PubMed database was performed, focusing on studies published within the past 10 years. Our search included reviews and articles of ongoing and retrospective studies. We prioritized articles with findings that suggest considerations for improving combination therapy prediction methods over providing a meta-analysis of all currently available cancer combination therapy prediction methods. Computational methods used for drug combination therapy prediction in cancer research include networks, regression-based machine learning, classifier machine learning models, and deep learning approaches. Each method class has its own advantages and disadvantages, so careful consideration is needed to determine the most suitable class when designing a combination therapy prediction method. Future directions to improve current combination therapy prediction technology include incorporation of disease pathobiology, drug characteristics, patient multiomics data, and drug-drug interactions to determine maximally efficacious and tolerable drug regimens for cancer. As computational methods improve in their capability to integrate patient, drug, and disease data, more comprehensive models can be developed to more accurately predict safe and efficacious combination drug therapies for cancer and other complex diseases.
Collapse
Affiliation(s)
- Victoria L. Flanary
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jennifer L. Fisher
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Elizabeth J. Wilk
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Timothy C. Howton
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Brittany N. Lasseigne
- Department of Cell, Developmental and Integrative Biology, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
7
|
Sun G, Dong D, Dong Z, Zhang Q, Fang H, Wang C, Zhang S, Wu S, Dong Y, Wan Y. Drug repositioning: A bibliometric analysis. Front Pharmacol 2022; 13:974849. [PMID: 36225586 PMCID: PMC9549161 DOI: 10.3389/fphar.2022.974849] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/12/2022] [Indexed: 11/14/2022] Open
Abstract
Drug repurposing has become an effective approach to drug discovery, as it offers a new way to explore drugs. Based on the Science Citation Index Expanded (SCI-E) and Social Sciences Citation Index (SSCI) databases of the Web of Science core collection, this study presents a bibliometric analysis of drug repurposing publications from 2010 to 2020. Data were cleaned, mined, and visualized using Derwent Data Analyzer (DDA) software. An overview of the history and development trend of the number of publications, major journals, major countries, major institutions, author keywords, major contributors, and major research fields is provided. There were 2,978 publications included in the study. The findings show that the United States leads in this area of research, followed by China, the United Kingdom, and India. The Chinese Academy of Science published the most research studies, and NIH ranked first on the h-index. The Icahn School of Medicine at Mt Sinai leads in the average number of citations per study. Sci Rep, Drug Discov. Today, and Brief. Bioinform. are the three most productive journals evaluated from three separate perspectives, and pharmacology and pharmacy are unquestionably the most commonly used subject categories. Cheng, FX; Mucke, HAM; and Butte, AJ are the top 20 most prolific and influential authors. Keyword analysis shows that in recent years, most research has focused on drug discovery/drug development, COVID-19/SARS-CoV-2/coronavirus, molecular docking, virtual screening, cancer, and other research areas. The hotspots have changed in recent years, with COVID-19/SARS-CoV-2/coronavirus being the most popular topic for current drug repurposing research.
Collapse
Affiliation(s)
- Guojun Sun
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Dashun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Zuojun Dong
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Qian Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Hui Fang
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| | - Chaojun Wang
- Hangzhou Aeronautical Sanatorium for Special Service of Chinese Air Force, Hangzhou, China
| | - Shaoya Zhang
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Shuaijun Wu
- Institute of Pharmaceutical Preparations, Department of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Yichen Dong
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuehua Wan
- Institute of Information Resource, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
8
|
A comprehensive review of Artificial Intelligence and Network based approaches to drug repurposing in Covid-19. Biomed Pharmacother 2022; 153:113350. [PMID: 35777222 PMCID: PMC9236981 DOI: 10.1016/j.biopha.2022.113350] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 11/26/2022] Open
Abstract
Conventional drug discovery and development is tedious and time-taking process; because of which it has failed to keep the required pace to mitigate threats and cater demands of viral and re-occurring diseases, such as Covid-19. The main reasons of this delay in traditional drug development are: high attrition rates, extensive time requirements, and huge financial investment with significant risk. The effective solution to de novo drug discovery is drug repurposing. Previous studies have shown that the network-based approaches and analysis are versatile platform for repurposing as the network biology is used to model the interactions between variety of biological concepts. Herein, we provide a comprehensive background of machine learning and deep learning in drug repurposing while specifically focusing on the applications of network-based approach to drug repurposing in Covid-19, data sources, and tools used. Furthermore, use of network proximity, network diffusion, and AI on network-based drug repurposing for Covid-19 is well-explained. Finally, limitations of network-based approaches in general and specific to network are stated along with future recommendations for better network-based models.
Collapse
|
9
|
System and network biology-based computational approaches for drug repositioning. COMPUTATIONAL APPROACHES FOR NOVEL THERAPEUTIC AND DIAGNOSTIC DESIGNING TO MITIGATE SARS-COV-2 INFECTION 2022. [PMCID: PMC9300680 DOI: 10.1016/b978-0-323-91172-6.00003-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Recent advances in computational biology have not only fastened the drug discovery process but have also proven to be a powerful tool for the search of existing molecules of therapeutic value for drug repurposing. The system biology-based drug repurposing approaches shorten the time and reduced the cost of the whole process when compared to de novo drug discovery. In the present pandemic situation, these computational approaches have emerged as a boon to tackle the COVID-19 associated morbidities and mortalities. In this chapter, we present the overview of system biology-based network system approaches which can be exploited for the drug repurposing of disease. Besides, we have included information on relevant repurposed drugs which are currently used for the treatment of COVID-19.
Collapse
|
10
|
Sonaye HV, Sheikh RY, Doifode CA. Drug repurposing: Iron in the fire for older drugs. Biomed Pharmacother 2021; 141:111638. [PMID: 34153846 DOI: 10.1016/j.biopha.2021.111638] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 12/22/2022] Open
Abstract
Repositioning or "repurposing" of existing therapies for indications of alternative disease is an attractive approach that can generate lower costs and require a shorter approval time than developing a de novo drug. The development of experimental drugs is time-consuming, expensive, and limited to a fairly small number of targets. The incorporation of separate and complementary data should be used, as each type of data set exposes a specific feature of organism knowledge Drug repurposing opportunities are often focused on sporadic findings or on time-consuming pre-clinical drug tests which are often not guided by hypothesis. In comparison, repurposing in-silico drugs is a new, hypothesis-driven method that takes advantage of big-data use. Nonetheless, the widespread use of omics technology, enhanced data storage, data sense, machine learning algorithms, and computational modeling all give unparalleled knowledge of the methods of action of biological processes and drugs, providing wide availability, for both disease-related data and drug-related data. This review has taken an in-depth look at the current state, possibilities, and limitations of further progress in the field of drug repositioning.
Collapse
Affiliation(s)
- H V Sonaye
- Shri Sachhidanand Shikshan Santh's Taywade College of Pharmacy, Nagpur 441111, India.
| | - R Y Sheikh
- K.E.M. Hospital Research Centre, Pune 411011, India.
| | - C A Doifode
- Shri Sachhidanand Shikshan Santh's Taywade College of Pharmacy, Nagpur 441111, India.
| |
Collapse
|
11
|
Ji X, Wang L, Hua L, Wang X, Zhang P, Shendre A, Feng W, Li J, Li L. Improved Adverse Drug Event Prediction Through Information Component Guided Pharmacological Network Model (IC-PNM). IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1113-1121. [PMID: 31443040 DOI: 10.1109/tcbb.2019.2928305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Improving adverse drug event (ADE) prediction is highly critical in pharmacovigilance research. We propose a novel information component guided pharmacological network model (IC-PNM) to predict drug-ADE signals. This new method combines the pharmacological network model and information component, a Bayes statistics method. We use 33,947 drug-ADE pairs from the FDA Adverse Event Reporting System (FAERS) 2010 data as the training data, and the new 21,065 drug-ADE pairs from FAERS 2011-2015 as the validations samples. The IC-PNM data analysis suggests that both large and small sample size drug-ADE pairs are needed in training the predictive model for its prediction performance to reach an area under the receiver operating characteristic curve [Formula: see text]. On the other hand, the IC-PNM prediction performance improved to [Formula: see text] if we removed the small sample size drug-ADE pairs from the prediction model during validation.
Collapse
|
12
|
Yang YF, Yu B, Zhang XX, Zhu YH. Identification of TNIK as a novel potential drug target in thyroid cancer based on protein druggability prediction. Medicine (Baltimore) 2021; 100:e25541. [PMID: 33879700 PMCID: PMC8078263 DOI: 10.1097/md.0000000000025541] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 03/25/2021] [Indexed: 01/04/2023] Open
Abstract
Thyroid cancer is a common endocrine malignancy; however, surgery remains its primary treatment option. A novel targeted drug for the development and application of targeted therapy in thyroid cancer treatment remain underexplored.We obtained RNA sequence data of thyroid cancer from The Cancer Genome Atlas database and identified differentially expressed genes (DEGs). Then, we constructed co-expression network with DEGs and combined it with differentially methylation analysis to screen the key genes in thyroid cancer. PockDrug-Server, an online tool, was applied to predict the druggability of the key genes. Finally, we constructed protein-protein interaction (PPI) network to observe potential targeted drugs for thyroid cancer.We identified 3 genes correlated with altered DNA methylation level and oncogenesis of thyroid cancer. According to the druggable analysis and PPI network, we predicted TRAF2 and NCK-interacting protein kinase (TNIK) sever as the drug targeted for thyroid cancer. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that genes in protein-protein interaction network of TNIK enriched in mitogen-activated protein kinase signaling pathway. For drug repositioning, we identified a targeted drug of genes in PPI network.Our study provides a bioinformatics method for screening drug targets and provides a theoretical basis for thyroid cancer targeted therapy.
Collapse
|
13
|
Turanli B, Altay O, Borén J, Turkez H, Nielsen J, Uhlen M, Arga KY, Mardinoglu A. Systems biology based drug repositioning for development of cancer therapy. Semin Cancer Biol 2019; 68:47-58. [PMID: 31568815 DOI: 10.1016/j.semcancer.2019.09.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/20/2023]
Abstract
Drug repositioning is a powerful method that can assists the conventional drug discovery process by using existing drugs for treatment of a disease rather than its original indication. The first examples of repurposed drugs were discovered serendipitously, however data accumulated by high-throughput screenings and advancements in computational biology methods have paved the way for rational drug repositioning methods. As chemotherapeutic agents have notorious side effects that significantly reduce quality of life, drug repositioning promises repurposed noncancer drugs with little or tolerable adverse effects for cancer patients. Here, we review current drug-related data types and databases including some examples of web-based drug repositioning tools. Next, we describe systems biology approaches to be used in drug repositioning for effective cancer therapy. Finally, we highlight examples of mostly repurposed drugs for cancer treatment and provide an overview of future expectations in the field for development of effective treatment strategies.
Collapse
Affiliation(s)
- Beste Turanli
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Department of Bioengineering, Marmara University, Istanbul, Turkey; Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital Gothenburg, Sweden
| | - Hasan Turkez
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25240, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | | | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom.
| |
Collapse
|
14
|
Gazerani P. Identification of novel analgesics through a drug repurposing strategy. Pain Manag 2019; 9:399-415. [DOI: 10.2217/pmt-2018-0091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The identification of new indications for approved or failed drugs is a process called drug repositioning or drug repurposing. The motivation includes overcoming the productivity gap that exists in drug development, which is a high-cost–high-risk process. Repositioning also includes rescuing drugs that have safely entered the market but have failed to demonstrate sufficient efficiency for the initial clinical indication. Considering the high prevalence of chronic pain, the lack of sufficient efficacy and the safety issues of current analgesics, repositioning seems to be an attractive approach. This review presents example of drugs that already have been repositioned and highlights new technologies that are available for the identification of additional compounds to stimulate the curiosity of readers for further exploration.
Collapse
Affiliation(s)
- Parisa Gazerani
- Biomedicine, Department of Health Science & Technology, Aalborg University, Frederik Bajers Vej 3 B, 9220 Aalborg East, Denmark
| |
Collapse
|
15
|
Park K. A review of computational drug repurposing. Transl Clin Pharmacol 2019; 27:59-63. [PMID: 32055582 PMCID: PMC6989243 DOI: 10.12793/tcp.2019.27.2.59] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 06/23/2019] [Accepted: 06/24/2019] [Indexed: 12/21/2022] Open
Abstract
Although sciences and technology have progressed rapidly, de novo drug development has been a costly and time-consuming process over the past decades. In view of these circumstances, ‘drug repurposing’ (or ‘drug repositioning’) has appeared as an alternative tool to accelerate drug development process by seeking new indications for already approved drugs rather than discovering de novo drug compounds, nowadays accounting for 30% of newly marked drugs in the U.S. In the meantime, the explosive and large-scale growth of molecular, genomic and phenotypic data of pharmacological compounds is enabling the development of new area of drug repurposing called computational drug repurposing. This review provides an overview of recent progress in the area of computational drug repurposing. First, it summarizes available repositioning strategies, followed by computational methods commonly used. Then, it describes validation techniques for repurposing studies. Finally, it concludes by discussing the remaining challenges in computational repurposing.
Collapse
Affiliation(s)
- Kyungsoo Park
- Department of Pharmacology, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
16
|
Lotfi Shahreza M, Ghadiri N, Mousavi SR, Varshosaz J, Green JR. A review of network-based approaches to drug repositioning. Brief Bioinform 2019; 19:878-892. [PMID: 28334136 DOI: 10.1093/bib/bbx017] [Citation(s) in RCA: 186] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Indexed: 01/17/2023] Open
Abstract
Experimental drug development is time-consuming, expensive and limited to a relatively small number of targets. However, recent studies show that repositioning of existing drugs can function more efficiently than de novo experimental drug development to minimize costs and risks. Previous studies have proven that network analysis is a versatile platform for this purpose, as the biological networks are used to model interactions between many different biological concepts. The present study is an attempt to review network-based methods in predicting drug targets for drug repositioning. For each method, the preferred type of data set is described, and their advantages and limitations are discussed. For each method, we seek to provide a brief description, as well as an evaluation based on its performance metrics.We conclude that integrating distinct and complementary data should be used because each type of data set reveals a unique aspect of information about an organism. We also suggest that applying a standard set of evaluation metrics and data sets would be essential in this fast-growing research domain.
Collapse
Affiliation(s)
- Maryam Lotfi Shahreza
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Nasser Ghadiri
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| | | | - Jaleh Varshosaz
- Drug Delivery Systems Research Center of Isfahan University of Medical Sciences
| | - James R Green
- Department of Electrical and Computer Engineering, Isfahan University of Technology, Isfahan, Iran
| |
Collapse
|
17
|
Yella JK, Yaddanapudi S, Wang Y, Jegga AG. Changing Trends in Computational Drug Repositioning. Pharmaceuticals (Basel) 2018; 11:E57. [PMID: 29874824 PMCID: PMC6027196 DOI: 10.3390/ph11020057] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/01/2018] [Accepted: 06/02/2018] [Indexed: 12/12/2022] Open
Abstract
Efforts to maximize the indications potential and revenue from drugs that are already marketed are largely motivated by what Sir James Black, a Nobel Prize-winning pharmacologist advocated-"The most fruitful basis for the discovery of a new drug is to start with an old drug". However, rational design of drug mixtures poses formidable challenges because of the lack of or limited information about in vivo cell regulation, mechanisms of genetic pathway activation, and in vivo pathway interactions. Hence, most of the successfully repositioned drugs are the result of "serendipity", discovered during late phase clinical studies of unexpected but beneficial findings. The connections between drug candidates and their potential adverse drug reactions or new applications are often difficult to foresee because the underlying mechanism associating them is largely unknown, complex, or dispersed and buried in silos of information. Discovery of such multi-domain pharmacomodules-pharmacologically relevant sub-networks of biomolecules and/or pathways-from collection of databases by independent/simultaneous mining of multiple datasets is an active area of research. Here, while presenting some of the promising bioinformatics approaches and pipelines, we summarize and discuss the current and evolving landscape of computational drug repositioning.
Collapse
Affiliation(s)
- Jaswanth K Yella
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
| | - Suryanarayana Yaddanapudi
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
| | - Yunguan Wang
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
| | - Anil G Jegga
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way MLC 7024, Cincinnati, OH 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
- Department of Computer Science, University of Cincinnati College of Engineering, Cincinnati, OH 45219, USA.
| |
Collapse
|
18
|
NMR-based metabonomic analysis of normal rat urine and faeces in response to (±)-venlafaxine treatment. J Pharm Biomed Anal 2016; 123:82-92. [DOI: 10.1016/j.jpba.2016.01.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 01/17/2016] [Accepted: 01/19/2016] [Indexed: 11/24/2022]
|
19
|
|
20
|
Regan K, Payne PRO. From Molecules to Patients: The Clinical Applications of Translational Bioinformatics. Yearb Med Inform 2015; 10:164-9. [PMID: 26293863 PMCID: PMC4587059 DOI: 10.15265/iy-2015-005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE In order to realize the promise of personalized medicine, Translational Bioinformatics (TBI) research will need to continue to address implementation issues across the clinical spectrum. In this review, we aim to evaluate the expanding field of TBI towards clinical applications, and define common themes and current gaps in order to motivate future research. METHODS Here we present the state-of-the-art of clinical implementation of TBI-based tools and resources. Our thematic analyses of a targeted literature search of recent TBI-related articles ranged across topics in genomics, data management, hypothesis generation, molecular epidemiology, diagnostics, therapeutics and personalized medicine. RESULTS Open areas of clinically-relevant TBI research identified in this review include developing data standards and best practices, publicly available resources, integrative systemslevel approaches, user-friendly tools for clinical support, cloud computing solutions, emerging technologies and means to address pressing legal, ethical and social issues. CONCLUSIONS There is a need for further research bridging the gap from foundational TBI-based theories and methodologies to clinical implementation. We have organized the topic themes presented in this review into four conceptual foci - domain analyses, knowledge engineering, computational architectures and computation methods alongside three stages of knowledge development in order to orient future TBI efforts to accelerate the goals of personalized medicine.
Collapse
Affiliation(s)
| | - P R O Payne
- Philip R.O. Payne, PhD, FACMI, The Ohio State University, Department of Biomedical Informatics, 250 Lincoln Tower, 1800 Cannon Drive, Columbus, OH 43210, USA, Tel: +1 614 292 4778, E-mail:
| |
Collapse
|
21
|
Ayvaz S, Horn J, Hassanzadeh O, Zhu Q, Stan J, Tatonetti NP, Vilar S, Brochhausen M, Samwald M, Rastegar-Mojarad M, Dumontier M, Boyce RD. Toward a complete dataset of drug-drug interaction information from publicly available sources. J Biomed Inform 2015; 55:206-17. [PMID: 25917055 PMCID: PMC4464899 DOI: 10.1016/j.jbi.2015.04.006] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Revised: 03/30/2015] [Accepted: 04/15/2015] [Indexed: 10/23/2022]
Abstract
Although potential drug-drug interactions (PDDIs) are a significant source of preventable drug-related harm, there is currently no single complete source of PDDI information. In the current study, all publically available sources of PDDI information that could be identified using a comprehensive and broad search were combined into a single dataset. The combined dataset merged fourteen different sources including 5 clinically-oriented information sources, 4 Natural Language Processing (NLP) Corpora, and 5 Bioinformatics/Pharmacovigilance information sources. As a comprehensive PDDI source, the merged dataset might benefit the pharmacovigilance text mining community by making it possible to compare the representativeness of NLP corpora for PDDI text extraction tasks, and specifying elements that can be useful for future PDDI extraction purposes. An analysis of the overlap between and across the data sources showed that there was little overlap. Even comprehensive PDDI lists such as DrugBank, KEGG, and the NDF-RT had less than 50% overlap with each other. Moreover, all of the comprehensive lists had incomplete coverage of two data sources that focus on PDDIs of interest in most clinical settings. Based on this information, we think that systems that provide access to the comprehensive lists, such as APIs into RxNorm, should be careful to inform users that the lists may be incomplete with respect to PDDIs that drug experts suggest clinicians be aware of. In spite of the low degree of overlap, several dozen cases were identified where PDDI information provided in drug product labeling might be augmented by the merged dataset. Moreover, the combined dataset was also shown to improve the performance of an existing PDDI NLP pipeline and a recently published PDDI pharmacovigilance protocol. Future work will focus on improvement of the methods for mapping between PDDI information sources, identifying methods to improve the use of the merged dataset in PDDI NLP algorithms, integrating high-quality PDDI information from the merged dataset into Wikidata, and making the combined dataset accessible as Semantic Web Linked Data.
Collapse
Affiliation(s)
- Serkan Ayvaz
- Department of Computer Science, Kent State University, 241 Math and Computer Science Building, Kent, OH 44242, USA.
| | - John Horn
- Department of Pharmacy, School of Pharmacy and University of Washington Medicine, Pharmacy Services, University of Washington, H375V Health Sciences Bldg, Box 357630, Seattle, WA 98195, USA.
| | - Oktie Hassanzadeh
- IBM T.J. Watson Research Center, 1101 Kitchawan Rd Route 134, P.O. Box 218, Yorktown Heights, NY 10598, USA.
| | - Qian Zhu
- Department of Information Systems, University of Maryland Baltimore County, Baltimore, MD 21250, USA.
| | - Johann Stan
- Lister Hill National Center for Biomedical Communications, National Library of Medicine, 8600 Rockville Pike, Bethesda, MD 20894, USA.
| | - Nicholas P Tatonetti
- Departments of Biomedical Informatics, Systems Biology, and Medicine, Columbia University, 622 West 168th St VC5, New York, NY 10032, USA.
| | - Santiago Vilar
- Departments of Biomedical Informatics, Systems Biology, and Medicine, Columbia University, 622 West 168th St VC5, New York, NY 10032, USA.
| | - Mathias Brochhausen
- Division of Biomedical Informatics, University of Arkansas for Medical Sciences, 4301 W. Markham St, #782, Little Rock, AR 72205-7199, USA.
| | - Matthias Samwald
- Section for Medical Expert and Knowledge-Based Systems, Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria.
| | - Majid Rastegar-Mojarad
- Biomedical Statistics & Informatics, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA.
| | - Michel Dumontier
- Stanford Center for Biomedical Informatics Research, Stanford, CA 94305, USA.
| | - Richard D Boyce
- Department of Biomedical Informatics, Suite 419, 5607 Baum Blvd, Pittsburgh, PA 15206-3701, USA.
| |
Collapse
|
22
|
Haas B, Chrusciel S, Fayad-Kobeissi S, Dubois-Randé JL, Azuaje F, Boczkowski J, Motterlini R, Foresti R. Permanent culture of macrophages at physiological oxygen attenuates the antioxidant and immunomodulatory properties of dimethyl fumarate. J Cell Physiol 2015; 230:1128-38. [PMID: 25303683 DOI: 10.1002/jcp.24844] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/01/2014] [Indexed: 12/20/2022]
Abstract
We hypothesized that O2 tension influences the redox state and the immunomodulatory responses of inflammatory cells to dimethyl fumarate (DMF), an activator of the nuclear factor Nrf2 that controls antioxidant genes expression. This concept was investigated in macrophages permanently cultured at either physiological (5% O2) or atmospheric (20% O2) oxygen levels and then treated with DMF or challenged with lipopolysaccharide (LPS) to induce inflammation. RAW 264.7 macrophages cultured at 20% O2 exhibited a pro-oxidant phenotype, reflected by a lower content of reduced glutathione, higher oxidized glutathione and increased production of reactive oxygen species when compared to macrophages continuously grown at 5% O2. At 20% O2, DMF induced a stronger antioxidant response compared to 5% O2 as evidenced by a higher expression of heme oxygenase-1, NAD(P)H:quinone oxydoreductase-1 and superoxide dismutase-2. After challenge of macrophages with LPS, several pro-inflammatory (iNOS, TNF-α, MMP-2, MMP-9), anti-inflammatory (arginase-1, IL-10) and pro-angiogenic (VEGF-A) mediators were evaluated in the presence or absence of DMF. All markers, with few interesting exceptions, were significantly reduced at 5% O2. This study brings new insights on the effects of O2 in the cellular adaptation to oxidative and inflammatory stimuli and highlights the importance of characterizing the effects of chemicals and drugs at physiologically relevant O2 tension. Our results demonstrate that the common practice of culturing cells at atmospheric O2 drives the endogenous cellular environment towards an oxidative stress phenotype, affecting inflammation and the expression of antioxidant pathways by exogenous modulators.
Collapse
Affiliation(s)
- Benjamin Haas
- Université Paris-Est, UMR_S955, UPEC, France; INSERM, U955, Equipe 03, France
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Malty RH, Jessulat M, Jin K, Musso G, Vlasblom J, Phanse S, Zhang Z, Babu M. Mitochondrial targets for pharmacological intervention in human disease. J Proteome Res 2014; 14:5-21. [PMID: 25367773 PMCID: PMC4286170 DOI: 10.1021/pr500813f] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
![]()
Over the past several years, mitochondrial
dysfunction has been
linked to an increasing number of human illnesses, making mitochondrial
proteins (MPs) an ever more appealing target for therapeutic intervention.
With 20% of the mitochondrial proteome (312 of an estimated 1500 MPs)
having known interactions with small molecules, MPs appear to be highly
targetable. Yet, despite these targeted proteins functioning in a
range of biological processes (including induction of apoptosis, calcium
homeostasis, and metabolism), very few of the compounds targeting
MPs find clinical use. Recent work has greatly expanded the number
of proteins known to localize to the mitochondria and has generated
a considerable increase in MP 3D structures available in public databases,
allowing experimental screening and in silico prediction of mitochondrial
drug targets on an unprecedented scale. Here, we summarize the current
literature on clinically active drugs that target MPs, with a focus
on how existing drug targets are distributed across biochemical pathways
and organelle substructures. Also, we examine current strategies for
mitochondrial drug discovery, focusing on genetic, proteomic, and
chemogenomic assays, and relevant model systems. As cell models and
screening techniques improve, MPs appear poised to emerge as relevant
targets for a wide range of complex human diseases, an eventuality
that can be expedited through systematic analysis of MP function.
Collapse
Affiliation(s)
- Ramy H Malty
- Department of Biochemistry, Research and Innovation Centre, University of Regina , Regina, Saskatchewan S4S 0A2, Canada
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Cami A, Reis BY. Concordance and predictive value of two adverse drug event data sets. BMC Med Inform Decis Mak 2014; 14:74. [PMID: 25149292 PMCID: PMC4150549 DOI: 10.1186/1472-6947-14-74] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 08/18/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Accurate prediction of adverse drug events (ADEs) is an important means of controlling and reducing drug-related morbidity and mortality. Since no single "gold standard" ADE data set exists, a range of different drug safety data sets are currently used for developing ADE prediction models. There is a critical need to assess the degree of concordance between these various ADE data sets and to validate ADE prediction models against multiple reference standards. METHODS We systematically evaluated the concordance of two widely used ADE data sets - Lexi-comp from 2010 and SIDER from 2012. The strength of the association between ADE (drug) counts in Lexi-comp and SIDER was assessed using Spearman rank correlation, while the differences between the two data sets were characterized in terms of drug categories, ADE categories and ADE frequencies. We also performed a comparative validation of the Predictive Pharmacosafety Networks (PPN) model using both ADE data sets. The predictive power of PPN using each of the two validation sets was assessed using the area under Receiver Operating Characteristic curve (AUROC). RESULTS The correlations between the counts of ADEs and drugs in the two data sets were 0.84 (95% CI: 0.82-0.86) and 0.92 (95% CI: 0.91-0.93), respectively. Relative to an earlier snapshot of Lexi-comp from 2005, Lexi-comp 2010 and SIDER 2012 introduced a mean of 1,973 and 4,810 new drug-ADE associations per year, respectively. The difference between these two data sets was most pronounced for Nervous System and Anti-infective drugs, Gastrointestinal and Nervous System ADEs, and postmarketing ADEs. A minor difference of 1.1% was found in the AUROC of PPN when SIDER 2012 was used for validation instead of Lexi-comp 2010. CONCLUSIONS In conclusion, the ADE and drug counts in Lexi-comp and SIDER data sets were highly correlated and the choice of validation set did not greatly affect the overall prediction performance of PPN. Our results also suggest that it is important to be aware of the differences that exist among ADE data sets, especially in modeling applications focused on specific drug and ADE categories.
Collapse
Affiliation(s)
- Aurel Cami
- Division of Emergency Medicine, Boston Children's Hospital, 1 Autumn Street, 5th Floor, Boston, MA 02215, USA.
| | | |
Collapse
|
25
|
Abstract
BACKGROUND More attention has been being paid to combinatorial effects of drugs to treat complex diseases or to avoid adverse combinations of drug cocktail. Although drug interaction information has been increasingly accumulated, a novel approach like network-based method is needed to analyse that information systematically and intuitively RESULTS Beyond focussing on drug-drug interactions, we examined interactions between functional drug groups. In this work, functional drug groups were defined based on the Anatomical Therapeutic Chemical (ATC) Classification System. We defined criteria whether two functional drug groups are related. Then we constructed the interaction network of drug groups. The resulting network provides intuitive interpretations. We further constructed another network based on interaction sharing ratio of the first network. Subsequent analysis of the networks showed that some features of drugs can be well described by this kind of interaction even for the case of structurally dissimilar drugs. CONCLUSION Our networks in this work provide intuitive insights into interactions among drug groups rather than those among single drugs. In addition, information on these interactions can be used as a useful source to describe mechanisms and features of drugs.
Collapse
|
26
|
Vandamme D, Fitzmaurice W, Kholodenko B, Kolch W. Systems medicine: helping us understand the complexity of disease. QJM 2013; 106:891-5. [PMID: 23904523 DOI: 10.1093/qjmed/hct163] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Advances in genomics and other -omic fields in the last decade have resulted in unprecedented volumes of complex data now being available. These data can enable physicians to provide their patients with care that is more personalized, predictive, preventive and participatory. The expertise required to manage and understand this data is to be found in fields outside of medical science, thus multidisciplinary collaboration coupled to a systems approach is key to unlocking its potential, with concomitant new ways of working. Systems medicine can build on the successes in the field of systems biology, recognizing the human body as the multidimensional network of networks that it is. While systems medicine can provide a conceptual and theoretical framework, its practical goal is to provide physicians the tools necessary for harnessing the rapid advances in basic biomedical science into their routine clinical arsenal.
Collapse
Affiliation(s)
- D Vandamme
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
| | | | | | | |
Collapse
|
27
|
|