1
|
Fonte M, Rôla C, Santana S, Prudêncio M, Almeida J, Ferraz R, Prudêncio C, Teixeira C, Gomes P. Repurposing antiplasmodial leads for cancer: Exploring the antiproliferative effects of N-cinnamoyl-aminoacridines. Bioorg Med Chem Lett 2024; 111:129894. [PMID: 39043264 DOI: 10.1016/j.bmcl.2024.129894] [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: 04/14/2024] [Revised: 06/26/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
Drug repurposing and rescuing have been widely explored as cost-effective approaches to expand the portfolio of chemotherapeutic agents. Based on the reported antitumor properties of both trans-cinnamic acids and quinacrine, an antimalarial aminoacridine, we explored the antiproliferative properties of two series of N-cinnamoyl-aminoacridines recently identified as multi-stage antiplasmodial leads. The compounds were evaluated in vitro against three cancer cell lines (MKN-28, Huh-7, and HepG2), and human primary dermal fibroblasts. One of the series displayed highly selective antiproliferative activity in the micromolar range against the three cancer cell lines tested, without any toxicity to non-carcinogenic cells.
Collapse
Affiliation(s)
- Mélanie Fonte
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal
| | - Catarina Rôla
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Sofia Santana
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Joana Almeida
- Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Ricardo Ferraz
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal; Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Cristina Prudêncio
- Centro de Investigação em Saúde Translacional e Biotecnologia Médica (TBIO)/Rede de Investigação em Saúde (RISE-Health), Escola Superior de Saúde, Instituto Politécnico do Porto, Portugal; Ciências Químicas e das Biomoléculas, Escola Superior de Saúde, Politécnico do Porto, Portugal
| | - Cátia Teixeira
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal; Gyros Protein Technologies Inc., Tucson, AZ, USA
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal.
| |
Collapse
|
2
|
Savva K, Zachariou M, Kynigopoulos D, Fella E, Vitali MI, Kosofidou X, Spyrou M, Sargiannidou I, Panayiotou E, Dietis N, Spyrou GM. Preliminary In Vitro and In Vivo Insights of In Silico Candidate Repurposed Drugs for Alzheimer's Disease. Life (Basel) 2023; 13:life13051095. [PMID: 37240740 DOI: 10.3390/life13051095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and is the most common type of dementia. Although a considerably large amount of money has been invested in drug development for AD, no disease modifying treatment has been detected so far. In our previous work, we developed a computational method to highlight stage-specific candidate repurposed drugs against AD. In this study, we tested the effect of the top 13 candidate repurposed drugs that we proposed in our previous work in a severity stage-specific manner using an in vitro BACE1 assay and the effect of a top-ranked drug from the list of our previous work, tetrabenazine (TBZ), in the 5XFAD as an AD mouse model. From our in vitro screening, we detected 2 compounds (clomiphene citrate and Pik-90) that showed statistically significant inhibition against the activity of the BACE1 enzyme. The administration of TBZ at the selected dose and therapeutic regimen in 5XFAD in male and female mice showed no significant effect in behavioral tests using the Y-maze and the ELISA immunoassay of Aβ40. To our knowledge, this is the first time the drug tetrabenazine has been tested in the 5XFAD mouse model of AD in a sex-stratified manner. Our results highlight 2 drugs (clomiphene citrate and Pik-90) from our previous computational work for further investigation.
Collapse
Affiliation(s)
- Kyriaki Savva
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Margarita Zachariou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Demos Kynigopoulos
- Department of Neuropathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Eleni Fella
- Department of Neuropathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Maria-Ioanna Vitali
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - Xeni Kosofidou
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - Michail Spyrou
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Elena Panayiotou
- Department of Neuropathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Nikolas Dietis
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - George M Spyrou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
3
|
Truong TTT, Panizzutti B, Kim JH, Walder K. Repurposing Drugs via Network Analysis: Opportunities for Psychiatric Disorders. Pharmaceutics 2022; 14:1464. [PMID: 35890359 PMCID: PMC9319329 DOI: 10.3390/pharmaceutics14071464] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023] Open
Abstract
Despite advances in pharmacology and neuroscience, the path to new medications for psychiatric disorders largely remains stagnated. Drug repurposing offers a more efficient pathway compared with de novo drug discovery with lower cost and less risk. Various computational approaches have been applied to mine the vast amount of biomedical data generated over recent decades. Among these methods, network-based drug repurposing stands out as a potent tool for the comprehension of multiple domains of knowledge considering the interactions or associations of various factors. Aligned well with the poly-pharmacology paradigm shift in drug discovery, network-based approaches offer great opportunities to discover repurposing candidates for complex psychiatric disorders. In this review, we present the potential of network-based drug repurposing in psychiatry focusing on the incentives for using network-centric repurposing, major network-based repurposing strategies and data resources, applications in psychiatry and challenges of network-based drug repurposing. This review aims to provide readers with an update on network-based drug repurposing in psychiatry. We expect the repurposing approach to become a pivotal tool in the coming years to battle debilitating psychiatric disorders.
Collapse
Affiliation(s)
- Trang T. T. Truong
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
| | - Bruna Panizzutti
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
| | - Jee Hyun Kim
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
- Mental Health Theme, The Florey Institute of Neuroscience and Mental Health, Parkville 3010, Australia
| | - Ken Walder
- IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong 3220, Australia; (T.T.T.T.); (B.P.); (J.H.K.)
| |
Collapse
|
4
|
Savva K, Zachariou M, Bourdakou MM, Dietis N, Spyrou GM. Network-based stage-specific drug repurposing for Alzheimer's disease. Comput Struct Biotechnol J 2022; 20:1427-1438. [PMID: 35386099 PMCID: PMC8957022 DOI: 10.1016/j.csbj.2022.03.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/14/2022] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common type of dementia. With no disease-curing drugs available and an ever-growing AD-related healthcare burden, novel approaches for identifying therapies are needed. In this work, we propose stage-specific candidate repurposed drugs against AD by using a novel network-based method for drug repurposing against different stages of AD severity. For each AD stage, this approach a) ranks the candidate repurposed drugs based on a novel network-based score emerging from the weighted sum of connections in a network resembling the structural similarity with failed, approved or currently ongoing drugs b) re-ranks the candidate drugs based on functional, structural and a priori information according to a recently developed method by our group and c) checks and re-ranks for permeability through the Blood Brain Barrier (BBB). Overall, we propose for further experimental validation 10 candidate repurposed drugs for each AD stage comprising a set of 26 elite candidate repurposed drugs due to overlaps between the three AD stages. We applied our methodology in a retrospective way on the known clinical trial drugs till 2016 and we show that we were able to highly rank a drug that did enter clinical trials in the following year. We expect that our proposed network-based drug-repurposing methodology will serve as a paradigm for application for ranking candidate repurposed drugs in other brain diseases beyond AD.
Collapse
Affiliation(s)
- Kyriaki Savva
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Margarita Zachariou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Marilena M. Bourdakou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Nikolas Dietis
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, Cyprus
| | - George M. Spyrou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
5
|
Lardos A, Aghaebrahimian A, Koroleva A, Sidorova J, Wolfram E, Anisimova M, Gil M. Computational Literature-based Discovery for Natural Products Research: Current State and Future Prospects. FRONTIERS IN BIOINFORMATICS 2022; 2:827207. [PMID: 36304281 PMCID: PMC9580913 DOI: 10.3389/fbinf.2022.827207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/28/2022] [Indexed: 11/21/2022] Open
Abstract
Literature-based discovery (LBD) mines existing literature in order to generate new hypotheses by finding links between previously disconnected pieces of knowledge. Although automated LBD systems are becoming widespread and indispensable in a wide variety of knowledge domains, little has been done to introduce LBD to the field of natural products research. Despite growing knowledge in the natural product domain, most of the accumulated information is found in detached data pools. LBD can facilitate better contextualization and exploitation of this wealth of data, for example by formulating new hypotheses for natural product research, especially in the context of drug discovery and development. Moreover, automated LBD systems promise to accelerate the currently tedious and expensive process of lead identification, optimization, and development. Focusing on natural product research, we briefly reflect the development of automated LBD and summarize its methods and principal data sources. In a thorough review of published use cases of LBD in the biomedical domain, we highlight the immense potential of this data mining approach for natural product research, especially in context with drug discovery or repurposing, mode of action, as well as drug or substance interactions. Most of the 91 natural product-related discoveries in our sample of reported use cases of LBD were addressed at a computer science audience. Therefore, it is the wider goal of this review to introduce automated LBD to researchers who work with natural products and to facilitate the dialogue between this community and the developers of automated LBD systems.
Collapse
Affiliation(s)
- Andreas Lardos
- Natural Product Chemistry and Phytopharmacy Research Group, Institute of Chemistry and Biotechnology, School of Life Sciences and Facility Management, Zurich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
| | - Ahmad Aghaebrahimian
- Institute of Applied Simulation, School of Life Sciences and Facility Management, Zürich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Anna Koroleva
- Institute of Applied Simulation, School of Life Sciences and Facility Management, Zürich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Julia Sidorova
- Instituto de Tecnología del Conocimiento, Universidad Complutense de Madrid, Madrid, Spain
| | - Evelyn Wolfram
- Natural Product Chemistry and Phytopharmacy Research Group, Institute of Chemistry and Biotechnology, School of Life Sciences and Facility Management, Zurich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
| | - Maria Anisimova
- Institute of Applied Simulation, School of Life Sciences and Facility Management, Zürich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Manuel Gil
- Institute of Applied Simulation, School of Life Sciences and Facility Management, Zürich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
- Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| |
Collapse
|
6
|
Gurbuz O, Alanis-Lobato G, Picart-Armada S, Sun M, Haslinger C, Lawless N, Fernandez-Albert F. Knowledge Graphs for Indication Expansion: An Explainable Target-Disease Prediction Method. Front Genet 2022; 13:814093. [PMID: 35360842 PMCID: PMC8963915 DOI: 10.3389/fgene.2022.814093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/28/2022] [Indexed: 11/19/2022] Open
Abstract
Indication expansion aims to find new indications for existing targets in order to accelerate the process of launching a new drug for a disease on the market. The rapid increase in data types and data sources for computational drug discovery has fostered the use of semantic knowledge graphs (KGs) for indication expansion through target centric approaches, or in other words, target repositioning. Previously, we developed a novel method to construct a KG for indication expansion studies, with the aim of finding and justifying alternative indications for a target gene of interest. In contrast to other KGs, ours combines human-curated full-text literature and gene expression data from biomedical databases to encode relationships between genes, diseases, and tissues. Here, we assessed the suitability of our KG for explainable target-disease link prediction using a glass-box approach. To evaluate the predictive power of our KG, we applied shortest path with tissue information- and embedding-based prediction methods to a graph constructed with information published before or during 2010. We also obtained random baselines by applying the shortest path predictive methods to KGs with randomly shuffled node labels. Then, we evaluated the accuracy of the top predictions using gene-disease links reported after 2010. In addition, we investigated the contribution of the KG’s tissue expression entity to the prediction performance. Our experiments showed that shortest path-based methods significantly outperform the random baselines and embedding-based methods outperform the shortest path predictions. Importantly, removing the tissue expression entity from the KG severely impacts the quality of the predictions, especially those produced by the embedding approaches. Finally, since the interpretability of the predictions is crucial in indication expansion, we highlight the advantages of our glass-box model through the examination of example candidate target-disease predictions.
Collapse
Affiliation(s)
- Ozge Gurbuz
- Discovery Research Coordination Germany, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- *Correspondence: Ozge Gurbuz, ; Francesc Fernandez-Albert,
| | - Gregorio Alanis-Lobato
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Sergio Picart-Armada
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Miao Sun
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Christian Haslinger
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Nathan Lawless
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Francesc Fernandez-Albert
- Global Computational Biology and Data Sciences, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
- *Correspondence: Ozge Gurbuz, ; Francesc Fernandez-Albert,
| |
Collapse
|
7
|
Relation extraction from DailyMed structured product labels by optimally combining crowd, experts and machines. J Biomed Inform 2021; 122:103902. [PMID: 34481057 DOI: 10.1016/j.jbi.2021.103902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
The effectiveness of machine learning models to provide accurate and consistent results in drug discovery and clinical decision support is strongly dependent on the quality of the data used. However, substantive amounts of open data that drive drug discovery suffer from a number of issues including inconsistent representation, inaccurate reporting, and incomplete context. For example, databases of FDA-approved drug indications used in computational drug repositioning studies do not distinguish between treatments that simply offer symptomatic relief from those that target the underlying pathology. Moreover, drug indication sources often lack proper provenance and have little overlap. Consequently, new predictions can be of poor quality as they offer little in the way of new insights. Hence, work remains to be done to establish higher quality databases of drug indications that are suitable for use in drug discovery and repositioning studies. Here, we report on the combination of weak supervision (i.e., programmatic labeling and crowdsourcing) and deep learning methods for relation extraction from DailyMed text to create a higher quality drug-disease relation dataset. The generated drug-disease relation data shows a high overlap with DrugCentral, a manually curated dataset. Using this dataset, we constructed a machine learning model to classify relations between drugs and diseases from text into four categories; treatment, symptomatic relief, contradiction, and effect, exhibiting an improvement of 15.5% with Bi-LSTM (F1 score of 71.8%) over the best performing discrete method. Access to high quality data is crucial to building accurate and reliable drug repurposing prediction models. Our work suggests how the combination of crowds, experts, and machine learning methods can go hand-in-hand to improve datasets and predictive models.
Collapse
|
8
|
Noor A, Assiri A. A novel computational drug repurposing approach for Systemic Lupus Erythematosus (SLE) treatment using Semantic Web technologies. Saudi J Biol Sci 2021; 28:3886-3892. [PMID: 34220244 PMCID: PMC8241633 DOI: 10.1016/j.sjbs.2021.03.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/20/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Affiliation(s)
- Adeeb Noor
- Department of Information Technology, Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah 80221, Saudi Arabia
| | - Abdullah Assiri
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| |
Collapse
|
9
|
Naveed H, Reglin C, Schubert T, Gao X, Arold ST, Maitland ML. Identifying Novel Drug Targets by iDTPnd: A Case Study of Kinase Inhibitors. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:986-997. [PMID: 33794377 PMCID: PMC9403029 DOI: 10.1016/j.gpb.2020.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/08/2020] [Accepted: 05/11/2020] [Indexed: 11/16/2022]
Abstract
Current FDA-approved kinase inhibitors cause diverse adverse effects, some of which are due to the mechanism-independent effects of these drugs. Identifying these mechanism-independent interactions could improve drug safety and support drug repurposing. Here, we develop iDTPnd (integrated Drug Target Predictor with negative dataset), a computational approach for large-scale discovery of novel targets for known drugs. For a given drug, we construct a positive structural signature as well as a negative structural signature that captures the weakly conserved structural features of drug-binding sites. To facilitate assessment of unintended targets, iDTPnd also provides a docking-based interaction score and its statistical significance. We confirm the interactions of sorafenib, imatinib, dasatinib, sunitinib, and pazopanib with their known targets at a sensitivity of 52% and a specificity of 55%. We also validate 10 predicted novel targets by using in vitro experiments. Our results suggest that proteins other than kinases, such as nuclear receptors, cytochrome P450, and MHC class I molecules, can also be physiologically relevant targets of kinase inhibitors. Our method is general and broadly applicable for the identification of protein–small molecule interactions, when sufficient drug–target 3D data are available. The code for constructing the structural signatures is available at https://sfb.kaust.edu.sa/Documents/iDTP.zip.
Collapse
Affiliation(s)
- Hammad Naveed
- Toyota Technological Institute at Chicago, Chicago, IL 60637, USA; Department of Computer Science, National University of Computer and Emerging Sciences, Islamabad 44000, Pakistan.
| | | | | | - Xin Gao
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, Thuwal 23955, Saudi Arabia
| | - Stefan T Arold
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Biological and Environmental Sciences and Engineering (BESE) Division, Thuwal 23955, Saudi Arabia
| | - Michael L Maitland
- Inova Center for Personalized Health and Schar Cancer Institute, Falls Church, VA 22042 USA,; University of Virginia Cancer Center, Annandale, Virginia 22003, USA
| |
Collapse
|
10
|
Ouellette TW, Wright GE, Drögemöller BI, Ross CJ, Carleton BC. Integrating disease and drug-related phenotypes for improved identification of pharmacogenomic variants. Pharmacogenomics 2021; 22:251-261. [PMID: 33769074 DOI: 10.2217/pgs-2020-0130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim: To improve the identification and interpretation of pharmacogenetic variants through the integration of disease and drug-related traits. Materials & methods: We hypothesized that integrating genome-wide disease and pharmacogenomic data may drive new insights into drug toxicity and response by identifying shared genetic architecture. Pleiotropic variants were identified using a methodological framework incorporating colocalization analysis. Results: Using genome-wide association studies summary statistics from the UK Biobank, European Bioinformatics Institute genome-wide association studies catalog and the Pharmacogenomics Research Network, we validated pleiotropy at the ABCG2 locus between allopurinol response and gout and identified novel pleiotropy between antihypertensive-induced new-onset diabetes, Crohn's disease and inflammatory bowel disease at the IL18RAP/SLC9A4 locus. Conclusion: New mechanistic insights and genetic loci can be uncovered by identifying pleiotropy between disease and drug-related traits.
Collapse
Affiliation(s)
- Tom W Ouellette
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada
| | - Galen Eb Wright
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Britt I Drögemöller
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Colin Jd Ross
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Bruce C Carleton
- BC Children's Hospital Research Institute, Vancouver, BC, V5Z 4H4, Canada.,Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| |
Collapse
|
11
|
Tamoxifen exposure induces cleft palate in mice. Br J Oral Maxillofac Surg 2020; 59:52-57. [PMID: 32723574 DOI: 10.1016/j.bjoms.2020.07.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 07/09/2020] [Indexed: 02/08/2023]
Abstract
Cleft palate is a common birth defect in mammals, which can be caused by genetic or environmental factors, or both. Decades have witnessed that many environmental exposures during gestation extremely increase the incidence of cleft palate. Tamoxifen (TAM), a widely-used drug in treating breast cancer, has been reported to be associated with craniofacial defects including micrognathia and cleft palate in humans. However, its exact effects on the developing palate remain unclear. Here we took advantage of a mouse model to explore how TAM affects palatal development at the molecular level. We showed that excess exposure of TAM in the early embryonic stages indeed leads to cleft palate in mice. RNA-sequencing results strongly suggest the involvement of mitogen-activated protein kinase (MAPK) signalling in TAM-induced cleft palate. Interestingly, in the anterior portion of the TAM-treated palatal shelf, phosphorylated (p)-AKT and p-ERK1/2 were activated but p-p38 was inhibited, while in the posterior palate, the p-AKT increased but the levels of p-p38 and p-JNK decreased. We conclude that excess TAM exposure causes cleft palate defects in mice by regulating MAPK pathways, which implicates the importance of tightly-regulated MAPK signalling in palatal development. This study provides a basis for further exploration of the molecular aetiology of cleft palate defects caused by environmental factors and, based on our results, we would give a serious warning regarding prescription of TAM and potential cleft palate defects in animal models involving the inducible Cre-LoxP system.
Collapse
|
12
|
Brown AS, Patel CJ. A review of validation strategies for computational drug repositioning. Brief Bioinform 2018; 19:174-177. [PMID: 27881429 PMCID: PMC5862266 DOI: 10.1093/bib/bbw110] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Indexed: 12/15/2022] Open
Abstract
Repositioning of previously approved drugs is a promising methodology because it reduces the cost and duration of the drug development pipeline and reduces the likelihood of unforeseen adverse events. Computational repositioning is especially appealing because of the ability to rapidly screen candidates in silico and to reduce the number of possible repositioning candidates. What is unclear, however, is how useful such methods are in producing clinically efficacious repositioning hypotheses. Furthermore, there is no agreement in the field over the proper way to perform validation of in silico predictions, and in fact no systematic review of repositioning validation methodologies. To address this unmet need, we review the computational repositioning literature and capture studies in which authors claimed to have validated their work. Our analysis reveals widespread variation in the types of strategies, predictions made and databases used as ‘gold standards’. We highlight a key weakness of the most commonly used strategy and propose a path forward for the consistent analytic validation of repositioning techniques.
Collapse
Affiliation(s)
- Adam S Brown
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, MA, USA
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, MA, USA
- Corresponding author: Chirag J. Patel, Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, MA 02115, USA. Tel.: (617) 432 1195; E-mail:
| |
Collapse
|
13
|
Singh S, Gupta SK, Seth PK. Biomarkers for detection, prognosis and therapeutic assessment of neurological disorders. Rev Neurosci 2018; 29:771-789. [PMID: 29466244 DOI: 10.1515/revneuro-2017-0097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 12/17/2017] [Indexed: 10/24/2023]
Abstract
Neurological disorders have aroused a significant concern among the health scientists globally, as diseases such as Parkinson's, Alzheimer's and dementia lead to disability and people have to live with them throughout the life. Recent evidence suggests that a number of environmental chemicals such as pesticides (paraquat) and metals (lead and aluminum) are also the cause of these diseases and other neurological disorders. Biomarkers can help in detecting the disorder at the preclinical stage, progression of the disease and key metabolomic alterations permitting identification of potential targets for intervention. A number of biomarkers have been proposed for some neurological disorders based on laboratory and clinical studies. In silico approaches have also been used by some investigators. Yet the ideal biomarker, which can help in early detection and follow-up on treatment and identifying the susceptible populations, is not available. An attempt has therefore been made to review the recent advancements of in silico approaches for discovery of biomarkers and their validation. In silico techniques implemented with multi-omics approaches have potential to provide a fast and accurate approach to identify novel biomarkers.
Collapse
Affiliation(s)
- Sarita Singh
- Distinguished Scientist Laboratory, Biotech Park, Sector-G Jankipram, Kursi Road, Lucknow 226021, Uttar Pradesh, India
| | - Sunil Kumar Gupta
- Distinguished Scientist Laboratory, Biotech Park, Lucknow 226021, Uttar Pradesh, India
| | - Prahlad Kishore Seth
- Distinguished Scientist Laboratory, Biotech Park, Lucknow 226021, Uttar Pradesh, India
| |
Collapse
|
14
|
Brown AS, Rasooly D, Patel CJ. Leveraging Population-Based Clinical Quantitative Phenotyping for Drug Repositioning. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2018; 7:124-129. [PMID: 28941007 PMCID: PMC5824113 DOI: 10.1002/psp4.12258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/07/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
Computational drug repositioning methods can scalably nominate approved drugs for new diseases, with reduced risk of unforeseen side effects. The majority of methods eschew individual‐level phenotypes despite the promise of biomarker‐driven repositioning. In this study, we propose a framework for discovering serendipitous interactions between drugs and routine clinical phenotypes in cross‐sectional observational studies. Key to our strategy is the use of a healthy and nondiabetic population derived from the National Health and Nutrition Examination Survey, mitigating risk for confounding by indication. We combine complementary diagnostic phenotypes (fasting glucose and glucose response) and associate them with prescription drug usage. We then sought confirmation of phenotype‐drug associations in unidentifiable member claims data from the Aetna Insurance company using a retrospective self‐controlled case analysis approach. We identify bupropion as a plausible glucose lowering agent, suggesting that surveying otherwise healthy individuals in cross‐sectional studies can discover new drug repositioning hypotheses that have applicability to longitudinal clinical practice.
Collapse
Affiliation(s)
- Adam S Brown
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Danielle Rasooly
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
15
|
Constructing faceted taxonomy for heterogeneous entities based on object properties in linked data. DATA KNOWL ENG 2017. [DOI: 10.1016/j.datak.2017.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Brown AS, Patel CJ. MeSHDD: Literature-based drug-drug similarity for drug repositioning. J Am Med Inform Assoc 2017; 24:614-618. [PMID: 27678460 PMCID: PMC5391732 DOI: 10.1093/jamia/ocw142] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 08/17/2016] [Accepted: 08/23/2016] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Drug repositioning is a promising methodology for reducing the cost and duration of the drug discovery pipeline. We sought to develop a computational repositioning method leveraging annotations in the literature, such as Medical Subject Heading (MeSH) terms. METHODS We developed software to determine significantly co-occurring drug-MeSH term pairs and a method to estimate pair-wise literature-derived distances between drugs. RESULTS We found that literature-based drug-drug similarities predicted the number of shared indications across drug-drug pairs. Clustering drugs based on their similarity revealed both known and novel drug indications. We demonstrate the utility of our approach by generating repositioning hypotheses for the commonly used diabetes drug metformin. CONCLUSION Our study demonstrates that literature-derived similarity is useful for identifying potential repositioning opportunities. We provided open-source code and deployed a free-to-use, interactive application to explore our database of similarity-based drug clusters (available at http://apps.chiragjpgroup.org/MeSHDD/ ).
Collapse
Affiliation(s)
- Adam S Brown
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
17
|
Brown AS, Patel CJ. A standard database for drug repositioning. Sci Data 2017; 4:170029. [PMID: 28291243 PMCID: PMC5349249 DOI: 10.1038/sdata.2017.29] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 01/20/2017] [Indexed: 02/03/2023] Open
Abstract
Drug repositioning, the process of discovering, validating, and marketing previously approved drugs for new indications, is of growing interest to academia and industry due to reduced time and costs associated with repositioned drugs. Computational methods for repositioning are appealing because they putatively nominate the most promising candidate drugs for a given indication. Comparing the wide array of computational repositioning methods, however, is a challenge due to inconsistencies in method validation in the field. Furthermore, a common simplifying assumption, that all novel predictions are false, is intellectually unsatisfying and hinders reproducibility. We address this assumption by providing a gold standard database, repoDB, that consists of both true positives (approved drugs), and true negatives (failed drugs). We have made the full database and all code used to prepare it publicly available, and have developed a web application that allows users to browse subsets of the data (http://apps.chiragjpgroup.org/repoDB/).
Collapse
Affiliation(s)
- Adam S Brown
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, Massachusetts 02115, USA
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck St, Boston, Massachusetts 02115, USA
| |
Collapse
|
18
|
Gomes A, Fernandes I, Teixeira C, Mateus N, Sottomayor MJ, Gomes P. A Quinacrine Analogue Selective Against Gastric Cancer Cells: Insight from Biochemical and Biophysical Studies. ChemMedChem 2016; 11:2703-2712. [PMID: 27863116 DOI: 10.1002/cmdc.201600477] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/11/2016] [Indexed: 12/29/2022]
Abstract
One of the earliest synthetic antimalarial drugs, quinacrine, was recently reported as interesting for the treatment of acute myeloid leukemia. Inspired by this and similar findings, we evaluated a set of quinacrine analogues against gastric (MKN-28), colon (Caco-2), and breast (MFC-7) cancer cell lines and one normal human fibroblast cell line (HFF-1). All the compounds, previously developed by us as dual-stage antimalarial leads, displayed antiproliferative activity, and one of the set stood out as selective toward the gastric cancer cell line, MKN-28. Interestingly, this compound was transported across an in vitro MKN-28 model cell line in low amounts, and approximately 80 % was trapped inside those cells. Nuclear targeting of the same compound and its interactions with calf thymus DNA were assessed through combined fluorescence microscopy, spectroscopy, and calorimetry studies, which provided evidence for the compound's ability to reach the nucleus and to interact with DNA.
Collapse
Affiliation(s)
- Ana Gomes
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal
| | - Iva Fernandes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal
| | - Cátia Teixeira
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal
| | - Nuno Mateus
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal
| | - M J Sottomayor
- CIQ-UP, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal
| | - Paula Gomes
- UCIBIO-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007, Porto, Portugal
| |
Collapse
|
19
|
Rezaei Kolahchi A, Khadem Mohtaram N, Pezeshgi Modarres H, Mohammadi MH, Geraili A, Jafari P, Akbari M, Sanati-Nezhad A. Microfluidic-Based Multi-Organ Platforms for Drug Discovery. MICROMACHINES 2016; 7:E162. [PMID: 30404334 PMCID: PMC6189912 DOI: 10.3390/mi7090162] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/23/2016] [Accepted: 08/24/2016] [Indexed: 12/18/2022]
Abstract
Development of predictive multi-organ models before implementing costly clinical trials is central for screening the toxicity, efficacy, and side effects of new therapeutic agents. Despite significant efforts that have been recently made to develop biomimetic in vitro tissue models, the clinical application of such platforms is still far from reality. Recent advances in physiologically-based pharmacokinetic and pharmacodynamic (PBPK-PD) modeling, micro- and nanotechnology, and in silico modeling have enabled single- and multi-organ platforms for investigation of new chemical agents and tissue-tissue interactions. This review provides an overview of the principles of designing microfluidic-based organ-on-chip models for drug testing and highlights current state-of-the-art in developing predictive multi-organ models for studying the cross-talk of interconnected organs. We further discuss the challenges associated with establishing a predictive body-on-chip (BOC) model such as the scaling, cell types, the common medium, and principles of the study design for characterizing the interaction of drugs with multiple targets.
Collapse
Affiliation(s)
- Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| | - Nima Khadem Mohtaram
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada.
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA.
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| | - Mohammad Hossein Mohammadi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Ave., Tehran 11155-9516, Iran.
| | - Armin Geraili
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Azadi Ave., Tehran 11155-9516, Iran.
| | - Parya Jafari
- Department of Electrical Engineering, Sharif University of Technology, Azadi Ave., Tehran 11155-9516, Iran.
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada.
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada.
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
- Center for Bioengineering Research and Education, Biomedical Engineering Program, University of Calgary, 2500 University Drive NW, Calgary, AB T2N 1N4, Canada.
| |
Collapse
|
20
|
Iyappan A, Kawalia SB, Raschka T, Hofmann-Apitius M, Senger P. NeuroRDF: semantic integration of highly curated data to prioritize biomarker candidates in Alzheimer's disease. J Biomed Semantics 2016; 7:45. [PMID: 27392431 PMCID: PMC4939021 DOI: 10.1186/s13326-016-0079-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/23/2016] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Neurodegenerative diseases are incurable and debilitating indications with huge social and economic impact, where much is still to be learnt about the underlying molecular events. Mechanistic disease models could offer a knowledge framework to help decipher the complex interactions that occur at molecular and cellular levels. This motivates the need for the development of an approach integrating highly curated and heterogeneous data into a disease model of different regulatory data layers. Although several disease models exist, they often do not consider the quality of underlying data. Moreover, even with the current advancements in semantic web technology, we still do not have cure for complex diseases like Alzheimer's disease. One of the key reasons accountable for this could be the increasing gap between generated data and the derived knowledge. RESULTS In this paper, we describe an approach, called as NeuroRDF, to develop an integrative framework for modeling curated knowledge in the area of complex neurodegenerative diseases. The core of this strategy lies in the usage of well curated and context specific data for integration into one single semantic web-based framework, RDF. This increases the probability of the derived knowledge to be novel and reliable in a specific disease context. This infrastructure integrates highly curated data from databases (Bind, IntAct, etc.), literature (PubMed), and gene expression resources (such as GEO and ArrayExpress). We illustrate the effectiveness of our approach by asking real-world biomedical questions that link these resources to prioritize the plausible biomarker candidates. Among the 13 prioritized candidate genes, we identified MIF to be a potential emerging candidate due to its role as a pro-inflammatory cytokine. We additionally report on the effort and challenges faced during generation of such an indication-specific knowledge base comprising of curated and quality-controlled data. CONCLUSION Although many alternative approaches have been proposed and practiced for modeling diseases, the semantic web technology is a flexible and well established solution for harmonized aggregation. The benefit of this work, to use high quality and context specific data, becomes apparent in speculating previously unattended biomarker candidates around a well-known mechanism, further leveraged for experimental investigations.
Collapse
Affiliation(s)
- Anandhi Iyappan
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for Information Technology, Rheinische Friedrich-Wilhelms-Universität Bonn, 53113, Bonn, Germany
| | - Shweta Bagewadi Kawalia
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754, Sankt Augustin, Germany.
- Bonn-Aachen International Center for Information Technology, Rheinische Friedrich-Wilhelms-Universität Bonn, 53113, Bonn, Germany.
| | - Tamara Raschka
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754, Sankt Augustin, Germany
- University of Applied Sciences Koblenz, RheinAhrCampus, Joseph-Rovan-Allee 2, 53424, Remagen, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754, Sankt Augustin, Germany
- Bonn-Aachen International Center for Information Technology, Rheinische Friedrich-Wilhelms-Universität Bonn, 53113, Bonn, Germany
| | - Philipp Senger
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53754, Sankt Augustin, Germany
| |
Collapse
|
21
|
Kaalia R, Ghosh I. Semantics based approach for analyzing disease-target associations. J Biomed Inform 2016; 62:125-35. [PMID: 27349858 DOI: 10.1016/j.jbi.2016.06.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 06/23/2016] [Accepted: 06/24/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND A complex disease is caused by heterogeneous biological interactions between genes and their products along with the influence of environmental factors. There have been many attempts for understanding the cause of these diseases using experimental, statistical and computational methods. In the present work the objective is to address the challenge of representation and integration of information from heterogeneous biomedical aspects of a complex disease using semantics based approach. METHODS Semantic web technology is used to design Disease Association Ontology (DAO-db) for representation and integration of disease associated information with diabetes as the case study. The functional associations of disease genes are integrated using RDF graphs of DAO-db. Three semantic web based scoring algorithms (PageRank, HITS (Hyperlink Induced Topic Search) and HITS with semantic weights) are used to score the gene nodes on the basis of their functional interactions in the graph. RESULTS Disease Association Ontology for Diabetes (DAO-db) provides a standard ontology-driven platform for describing genes, proteins, pathways involved in diabetes and for integrating functional associations from various interaction levels (gene-disease, gene-pathway, gene-function, gene-cellular component and protein-protein interactions). An automatic instance loader module is also developed in present work that helps in adding instances to DAO-db on a large scale. CONCLUSIONS Our ontology provides a framework for querying and analyzing the disease associated information in the form of RDF graphs. The above developed methodology is used to predict novel potential targets involved in diabetes disease from the long list of loose (statistically associated) gene-disease associations.
Collapse
Affiliation(s)
- Rama Kaalia
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Indira Ghosh
- School of Computational & Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
22
|
Moosavinasab S, Patterson J, Strouse R, Rastegar-Mojarad M, Regan K, Payne PRO, Huang Y, Lin SM. 'RE:fine drugs': an interactive dashboard to access drug repurposing opportunities. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2016; 2016:baw083. [PMID: 27189611 PMCID: PMC4869799 DOI: 10.1093/database/baw083] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/26/2016] [Indexed: 12/01/2022]
Abstract
The process of discovering new drugs has been extremely costly and slow in the last decades despite enormous investment in pharmaceutical research. Drug repurposing enables researchers to speed up the process of discovering other conditions that existing drugs can effectively treat, with low cost and fast FDA approval. Here, we introduce ‘RE:fine Drugs’, a freely available interactive website for integrated search and discovery of drug repurposing candidates from GWAS and PheWAS repurposing datasets constructed using previously reported methods in Nature Biotechnology. ‘RE:fine Drugs’ demonstrates the possibilities to identify and prioritize novelty of candidates for drug repurposing based on the theory of transitive Drug–Gene–Disease triads. This public website provides a starting point for research, industry, clinical and regulatory communities to accelerate the investigation and validation of new therapeutic use of old drugs. Database URL:http://drug-repurposing.nationwidechildrens.org
Collapse
Affiliation(s)
- Soheil Moosavinasab
- Research Information Solutions and Innovation, The Research Institute at Nationwide Children's Hospital Columbus, OH 43205, USA
| | - Jeremy Patterson
- Research Information Solutions and Innovation, The Research Institute at Nationwide Children's Hospital Columbus, OH 43205, USA
| | - Robert Strouse
- Research Information Solutions and Innovation, The Research Institute at Nationwide Children's Hospital Columbus, OH 43205, USA
| | | | - Kelly Regan
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Philip R O Payne
- Department of Biomedical Informatics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Yungui Huang
- Research Information Solutions and Innovation, The Research Institute at Nationwide Children's Hospital Columbus, OH 43205, USA
| | - Simon M Lin
- Research Information Solutions and Innovation, The Research Institute at Nationwide Children's Hospital Columbus, OH 43205, USA
| |
Collapse
|
23
|
Mei H, Feng G, Zhu J, Lin S, Qiu Y, Wang Y, Xia T. A Practical Guide for Exploring Opportunities of Repurposing Drugs for CNS Diseases in Systems Biology. Methods Mol Biol 2016; 1303:531-547. [PMID: 26235090 DOI: 10.1007/978-1-4939-2627-5_33] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Systems biology has shown its potential in facilitating pathway-focused therapy development for central nervous system (CNS) diseases. An integrated network can be utilized to explore the multiple disease mechanisms and to discover repositioning opportunities. This review covers current therapeutic gaps for CNS diseases and the role of systems biology in pharmaceutical industry. We conclude with a Multiple Level Network Modeling (MLNM) example to illustrate the great potential of systems biology for CNS diseases. The system focuses on the benefit and practical applications in pathway centric therapy and drug repositioning.
Collapse
Affiliation(s)
- Hongkang Mei
- Informatics and Structure Biology, R&D China, GlaxoSmithKline, 917 Halei Road, Shanghai, 201203, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Naveed H, Hameed US, Harrus D, Bourguet W, Arold ST, Gao X. An integrated structure- and system-based framework to identify new targets of metabolites and known drugs. Bioinformatics 2015; 31:3922-9. [PMID: 26286808 PMCID: PMC4673972 DOI: 10.1093/bioinformatics/btv477] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 08/08/2015] [Indexed: 02/07/2023] Open
Abstract
Motivation: The inherent promiscuity of small molecules towards protein targets impedes our understanding of healthy versus diseased metabolism. This promiscuity also poses a challenge for the pharmaceutical industry as identifying all protein targets is important to assess (side) effects and repositioning opportunities for a drug. Results: Here, we present a novel integrated structure- and system-based approach of drug-target prediction (iDTP) to enable the large-scale discovery of new targets for small molecules, such as pharmaceutical drugs, co-factors and metabolites (collectively called ‘drugs’). For a given drug, our method uses sequence order–independent structure alignment, hierarchical clustering and probabilistic sequence similarity to construct a probabilistic pocket ensemble (PPE) that captures promiscuous structural features of different binding sites on known targets. A drug’s PPE is combined with an approximation of its delivery profile to reduce false positives. In our cross-validation study, we use iDTP to predict the known targets of 11 drugs, with 63% sensitivity and 81% specificity. We then predicted novel targets for these drugs—two that are of high pharmacological interest, the peroxisome proliferator-activated receptor gamma and the oncogene B-cell lymphoma 2, were successfully validated through in vitro binding experiments. Our method is broadly applicable for the prediction of protein-small molecule interactions with several novel applications to biological research and drug development. Availability and implementation: The program, datasets and results are freely available to academic users at http://sfb.kaust.edu.sa/Pages/Software.aspx. Contact:xin.gao@kaust.edu.sa and stefan.arold@kaust.edu.sa Supplementary information:Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Hammad Naveed
- Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center
| | - Umar S Hameed
- Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Deborah Harrus
- Inserm U1054, Centre de Biochimie Structurale and CNRS UMR5048, Universités Montpellier 1 & 2, Montpellier, France
| | - William Bourguet
- Inserm U1054, Centre de Biochimie Structurale and CNRS UMR5048, Universités Montpellier 1 & 2, Montpellier, France
| | - Stefan T Arold
- Computational Bioscience Research Center, Biological and Environmental Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Xin Gao
- Computer, Electrical and Mathematical Sciences and Engineering Division, Computational Bioscience Research Center
| |
Collapse
|
25
|
|
26
|
Csermely P, Korcsmáros T, Kiss HJM, London G, Nussinov R. Structure and dynamics of molecular networks: a novel paradigm of drug discovery: a comprehensive review. Pharmacol Ther 2013; 138:333-408. [PMID: 23384594 PMCID: PMC3647006 DOI: 10.1016/j.pharmthera.2013.01.016] [Citation(s) in RCA: 512] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 01/22/2013] [Indexed: 02/02/2023]
Abstract
Despite considerable progress in genome- and proteome-based high-throughput screening methods and in rational drug design, the increase in approved drugs in the past decade did not match the increase of drug development costs. Network description and analysis not only give a systems-level understanding of drug action and disease complexity, but can also help to improve the efficiency of drug design. We give a comprehensive assessment of the analytical tools of network topology and dynamics. The state-of-the-art use of chemical similarity, protein structure, protein-protein interaction, signaling, genetic interaction and metabolic networks in the discovery of drug targets is summarized. We propose that network targeting follows two basic strategies. The "central hit strategy" selectively targets central nodes/edges of the flexible networks of infectious agents or cancer cells to kill them. The "network influence strategy" works against other diseases, where an efficient reconfiguration of rigid networks needs to be achieved by targeting the neighbors of central nodes/edges. It is shown how network techniques can help in the identification of single-target, edgetic, multi-target and allo-network drug target candidates. We review the recent boom in network methods helping hit identification, lead selection optimizing drug efficacy, as well as minimizing side-effects and drug toxicity. Successful network-based drug development strategies are shown through the examples of infections, cancer, metabolic diseases, neurodegenerative diseases and aging. Summarizing >1200 references we suggest an optimized protocol of network-aided drug development, and provide a list of systems-level hallmarks of drug quality. Finally, we highlight network-related drug development trends helping to achieve these hallmarks by a cohesive, global approach.
Collapse
Affiliation(s)
- Peter Csermely
- Department of Medical Chemistry, Semmelweis University, P.O. Box 260, H-1444 Budapest 8, Hungary.
| | | | | | | | | |
Collapse
|
27
|
Challenges and recommendations for obtaining chemical structures of industry-provided repurposing candidates. Drug Discov Today 2013; 18:58-70. [DOI: 10.1016/j.drudis.2012.11.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 10/17/2012] [Accepted: 11/08/2012] [Indexed: 02/07/2023]
|
28
|
Mei H, Xia T, Feng G, Zhu J, Lin SM, Qiu Y. Opportunities in systems biology to discover mechanisms and repurpose drugs for CNS diseases. Drug Discov Today 2012; 17:1208-16. [DOI: 10.1016/j.drudis.2012.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2012] [Revised: 06/04/2012] [Accepted: 06/25/2012] [Indexed: 01/07/2023]
|
29
|
Chen B, Ding Y, Wild DJ. Improving integrative searching of systems chemical biology data using semantic annotation. J Cheminform 2012; 4:6. [PMID: 22401035 PMCID: PMC3320537 DOI: 10.1186/1758-2946-4-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 03/08/2012] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Systems chemical biology and chemogenomics are considered critical, integrative disciplines in modern biomedical research, but require data mining of large, integrated, heterogeneous datasets from chemistry and biology. We previously developed an RDF-based resource called Chem2Bio2RDF that enabled querying of such data using the SPARQL query language. Whilst this work has proved useful in its own right as one of the first major resources in these disciplines, its utility could be greatly improved by the application of an ontology for annotation of the nodes and edges in the RDF graph, enabling a much richer range of semantic queries to be issued. RESULTS We developed a generalized chemogenomics and systems chemical biology OWL ontology called Chem2Bio2OWL that describes the semantics of chemical compounds, drugs, protein targets, pathways, genes, diseases and side-effects, and the relationships between them. The ontology also includes data provenance. We used it to annotate our Chem2Bio2RDF dataset, making it a rich semantic resource. Through a series of scientific case studies we demonstrate how this (i) simplifies the process of building SPARQL queries, (ii) enables useful new kinds of queries on the data and (iii) makes possible intelligent reasoning and semantic graph mining in chemogenomics and systems chemical biology. AVAILABILITY Chem2Bio2OWL is available at http://chem2bio2rdf.org/owl. The document is available at http://chem2bio2owl.wikispaces.com.
Collapse
Affiliation(s)
- Bin Chen
- School of Informatics and Computing, Indiana University, Bloomington, IN, USA
| | - Ying Ding
- School of Library and Information Science, Indiana University, Bloomington, IN, USA
| | - David J Wild
- School of Informatics and Computing, Indiana University, Bloomington, IN, USA
| |
Collapse
|
30
|
Sirota M, Dudley JT, Kim J, Chiang AP, Morgan AA, Sweet-Cordero A, Sage J, Butte AJ. Discovery and preclinical validation of drug indications using compendia of public gene expression data. Sci Transl Med 2012; 3:96ra77. [PMID: 21849665 DOI: 10.1126/scitranslmed.3001318] [Citation(s) in RCA: 549] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The application of established drug compounds to new therapeutic indications, known as drug repositioning, offers several advantages over traditional drug development, including reduced development costs and shorter paths to approval. Recent approaches to drug repositioning use high-throughput experimental approaches to assess a compound's potential therapeutic qualities. Here, we present a systematic computational approach to predict novel therapeutic indications on the basis of comprehensive testing of molecular signatures in drug-disease pairs. We integrated gene expression measurements from 100 diseases and gene expression measurements on 164 drug compounds, yielding predicted therapeutic potentials for these drugs. We recovered many known drug and disease relationships using computationally derived therapeutic potentials and also predict many new indications for these 164 drugs. We experimentally validated a prediction for the antiulcer drug cimetidine as a candidate therapeutic in the treatment of lung adenocarcinoma, and demonstrate its efficacy both in vitro and in vivo using mouse xenograft models. This computational method provides a systematic approach for repositioning established drugs to treat a wide range of human diseases.
Collapse
Affiliation(s)
- Marina Sirota
- Division of Systems Medicine, Department of Pediatrics, Stanford University School of Medicine, 251 Campus Drive, Stanford, CA 94305-5415, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Cheminformatic/bioinformatic analysis of large corporate databases: Application to drug repurposing. ACTA ACUST UNITED AC 2011. [DOI: 10.1016/j.ddstr.2011.06.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
32
|
Mathur S, Dinakarpandian D. Drug repositioning using disease associated biological processes and network analysis of drug targets. AMIA ... ANNUAL SYMPOSIUM PROCEEDINGS. AMIA SYMPOSIUM 2011; 2011:305-311. [PMID: 22195082 PMCID: PMC3243256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
The analysis of disease using protein-protein interaction networks and network pharmacology has enabled better understanding of disease etiology and drug action. New insights into disease etiology and a better understanding of biological subsystems have opened up the possibility of finding new uses for existing drugs besides their original medical indication. We present an approach which makes use of the biological processes associated with diseases along with their known drugs and drug targets to predict Biological Process-Drug relationships. Network analysis is used to further refine these associations to eventually predict new Disease-Drug relationships. The approach is validated by the observation that, out of 2078 predicted disease-drug relationships, 401 (18.1%) have been used in a clinical trial.
Collapse
Affiliation(s)
- Sachin Mathur
- University of Missouri-Kansas City, Kansas City, MO, USA
| | | |
Collapse
|
33
|
Xie L, Xie L, Kinnings SL, Bourne PE. Novel computational approaches to polypharmacology as a means to define responses to individual drugs. Annu Rev Pharmacol Toxicol 2011; 52:361-79. [PMID: 22017683 DOI: 10.1146/annurev-pharmtox-010611-134630] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Polypharmacology, which focuses on designing therapeutics to target multiple receptors, has emerged as a new paradigm in drug discovery. Polypharmacological effects are an attribute of most, if not all, drug molecules. The efficacy and toxicity of drugs, whether designed as single- or multitarget therapeutics, result from complex interactions between pharmacodynamic, pharmacokinetic, genetic, epigenetic, and environmental factors. Ultimately, to predict a drug response phenotype, it is necessary to understand the change in information flow through cellular networks resulting from dynamic drug-target interactions and the impact that this has on the complete biological system. Although such is a future objective, we review recent progress and challenges in computational techniques that enable the prediction and analysis of in vitro and in vivo drug-response phenotypes.
Collapse
Affiliation(s)
- Lei Xie
- Department of Computer Science, Hunter College, The City University of New York, New York, New York 10065, USA.
| | | | | | | |
Collapse
|
34
|
Dudley JT, Deshpande T, Butte AJ. Exploiting drug-disease relationships for computational drug repositioning. Brief Bioinform 2011; 12:303-11. [PMID: 21690101 DOI: 10.1093/bib/bbr013] [Citation(s) in RCA: 315] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Finding new uses for existing drugs, or drug repositioning, has been used as a strategy for decades to get drugs to more patients. As the ability to measure molecules in high-throughput ways has improved over the past decade, it is logical that such data might be useful for enabling drug repositioning through computational methods. Many computational predictions for new indications have been borne out in cellular model systems, though extensive animal model and clinical trial-based validation are still pending. In this review, we show that computational methods for drug repositioning can be classified in two axes: drug based, where discovery initiates from the chemical perspective, or disease based, where discovery initiates from the clinical perspective of disease or its pathology. Newer algorithms for computational drug repositioning will likely span these two axes, will take advantage of newer types of molecular measurements, and will certainly play a role in reducing the global burden of disease.
Collapse
|
35
|
Prussia A, Thepchatri P, Snyder JP, Plemper RK. Systematic approaches towards the development of host-directed antiviral therapeutics. Int J Mol Sci 2011; 12:4027-52. [PMID: 21747723 PMCID: PMC3131607 DOI: 10.3390/ijms12064027] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2011] [Revised: 06/03/2011] [Accepted: 06/03/2011] [Indexed: 12/11/2022] Open
Abstract
Since the onset of antiviral therapy, viral resistance has compromised the clinical value of small-molecule drugs targeting pathogen components. As intracellular parasites, viruses complete their life cycle by hijacking a multitude of host-factors. Aiming at the latter rather than the pathogen directly, host-directed antiviral therapy has emerged as a concept to counteract evolution of viral resistance and develop broad-spectrum drug classes. This approach is propelled by bioinformatics analysis of genome-wide screens that greatly enhance insights into the complex network of host-pathogen interactions and generate a shortlist of potential gene targets from a multitude of candidates, thus setting the stage for a new era of rational identification of drug targets for host-directed antiviral therapies. With particular emphasis on human immunodeficiency virus and influenza virus, two major human pathogens, we review screens employed to elucidate host-pathogen interactions and discuss the state of database ontology approaches applicable to defining a therapeutic endpoint. The value of this strategy for drug discovery is evaluated, and perspectives for bioinformatics-driven hit identification are outlined.
Collapse
Affiliation(s)
- Andrew Prussia
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA; E-Mails: (A.P.); (J.P.S.)
- Emory Institute for Drug Discovery (EIDD), Emory University, Atlanta, GA 30322, USA
| | - Pahk Thepchatri
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA; E-Mails: (A.P.); (J.P.S.)
- Emory Institute for Drug Discovery (EIDD), Emory University, Atlanta, GA 30322, USA
- Authors to whom correspondence should be addressed; E-Mails: (P.T.); (R.K.P.); Tel.: +1-404-593-0547 (P.T.); +1-404-727-1605 (R.K.P.); Fax: +1-404-727-9223 (P.T.); +1-404-727-9223 (R.K.P.)
| | - James P. Snyder
- Department of Chemistry, Emory University, Atlanta, GA 30322, USA; E-Mails: (A.P.); (J.P.S.)
- Emory Institute for Drug Discovery (EIDD), Emory University, Atlanta, GA 30322, USA
| | - Richard K. Plemper
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
- Children’s Healthcare of Atlanta, Atlanta, GA 30322, USA
- Authors to whom correspondence should be addressed; E-Mails: (P.T.); (R.K.P.); Tel.: +1-404-593-0547 (P.T.); +1-404-727-1605 (R.K.P.); Fax: +1-404-727-9223 (P.T.); +1-404-727-9223 (R.K.P.)
| |
Collapse
|
36
|
Chen H, Xie G. The use of web ontology languages and other semantic web tools in drug discovery. Expert Opin Drug Discov 2010; 5:413-23. [DOI: 10.1517/17460441003762709] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
37
|
Kann MG. Advances in translational bioinformatics: computational approaches for the hunting of disease genes. Brief Bioinform 2010; 11:96-110. [PMID: 20007728 PMCID: PMC2810112 DOI: 10.1093/bib/bbp048] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 09/15/2009] [Indexed: 12/29/2022] Open
Abstract
Over a 100 years ago, William Bateson provided, through his observations of the transmission of alkaptonuria in first cousin offspring, evidence of the application of Mendelian genetics to certain human traits and diseases. His work was corroborated by Archibald Garrod (Archibald AE. The incidence of alkaptonuria: a study in chemical individuality. Lancert 1902;ii:1616-20) and William Farabee (Farabee WC. Inheritance of digital malformations in man. In: Papers of the Peabody Museum of American Archaeology and Ethnology. Cambridge, Mass: Harvard University, 1905; 65-78), who recorded the familial tendencies of inheritance of malformations of human hands and feet. These were the pioneers of the hunt for disease genes that would continue through the century and result in the discovery of hundreds of genes that can be associated with different diseases. Despite many ground-breaking discoveries during the last century, we are far from having a complete understanding of the intricate network of molecular processes involved in diseases, and we are still searching for the cures for most complex diseases. In the last few years, new genome sequencing and other high-throughput experimental techniques have generated vast amounts of molecular and clinical data that contain crucial information with the potential of leading to the next major biomedical discoveries. The need to mine, visualize and integrate these data has motivated the development of several informatics approaches that can broadly be grouped in the research area of 'translational bioinformatics'. This review highlights the latest advances in the field of translational bioinformatics, focusing on the advances of computational techniques to search for and classify disease genes.
Collapse
Affiliation(s)
- Maricel G Kann
- University of Maryland, Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
38
|
Mousses S, Kiefer J, Von Hoff D, Trent J. Using biointelligence to search the cancer genome: an epistemological perspective on knowledge recovery strategies to enable precision medical genomics. Oncogene 2009; 27 Suppl 2:S58-66. [DOI: 10.1038/onc.2009.354] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
39
|
Clermont G, Auffray C, Moreau Y, Rocke DM, Dalevi D, Dubhashi D, Marshall DR, Raasch P, Dehne F, Provero P, Tegner J, Aronow BJ, Langston MA, Benson M. Bridging the gap between systems biology and medicine. Genome Med 2009; 1:88. [PMID: 19754960 PMCID: PMC2768995 DOI: 10.1186/gm88] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 06/11/2009] [Accepted: 09/15/2009] [Indexed: 11/10/2022] Open
Abstract
Systems biology has matured considerably as a discipline over the last decade, yet some of the key challenges separating current research efforts in systems biology and clinically useful results are only now becoming apparent. As these gaps are better defined, the new discipline of systems medicine is emerging as a translational extension of systems biology. How is systems medicine defined? What are relevant ontologies for systems medicine? What are the key theoretic and methodologic challenges facing computational disease modeling? How are inaccurate and incomplete data, and uncertain biologic knowledge best synthesized in useful computational models? Does network analysis provide clinically useful insight? We discuss the outstanding difficulties in translating a rapidly growing body of data into knowledge usable at the bedside. Although core-specific challenges are best met by specialized groups, it appears fundamental that such efforts should be guided by a roadmap for systems medicine drafted by a coalition of scientists from the clinical, experimental, computational, and theoretic domains.
Collapse
Affiliation(s)
- Gilles Clermont
- Department of Critical Care Medicine and CRISMA laboratory, University of Pittsburgh School of Medicine, Scaife 602, 3550 Terrace, Pittsburgh, PA 15261, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Systems pharmacology is an emerging area of pharmacology which utilizes network analysis of drug action as one of its approaches. By considering drug actions and side effects in the context of the regulatory networks within which the drug targets and disease gene products function, network analysis promises to greatly increase our knowledge of the mechanisms underlying the multiple actions of drugs. Systems pharmacology can provide new approaches for drug discovery for complex diseases. The integrated approach used in systems pharmacology can allow for drug action to be considered in the context of the whole genome. Network-based studies are becoming an increasingly important tool in understanding the relationships between drug action and disease susceptibility genes. This review discusses how analysis of biological networks has contributed to the genesis of systems pharmacology and how these studies have improved global understanding of drug targets, suggested new targets and approaches for therapeutics, and provided a deeper understanding of the effects of drugs. Taken together, these types of analyses can lead to new therapeutic options while improving the safety and efficacy of existing medications.
Collapse
Affiliation(s)
- Seth I Berger
- Department of Pharmacology and Systems Therapeutics, Mount Sinai School of Medicine, One Gustave Levy Place, New York, NY 10029, USA
| | | |
Collapse
|