151
|
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
|
152
|
Zhao C, Dai X, Li Y, Guo Q, Zhang J, Zhang X, Wang L. EK-DRD: A Comprehensive Database for Drug Repositioning Inspired by Experimental Knowledge. J Chem Inf Model 2019; 59:3619-3624. [PMID: 31433187 DOI: 10.1021/acs.jcim.9b00365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Drug repositioning, or the identification of new indications for approved therapeutic drugs, has gained substantial traction with both academics and pharmaceutical companies because it reduces the cost and duration of the drug development pipeline and the likelihood of unforeseen adverse events. To date there has not been a systematic effort to identify such opportunities, in part because of the lack of a comprehensive resource for an enormous amount of unsystematic drug repositioning information to support scientists who could benefit from this endeavor. To address this challenge, we developed a new database, the Experimental Knowledge-Based Drug Repositioning Database (EK-DRD), by using text and data mining as well as manual curation. EK-DRD contains experimentally validated drug repositioning annotation for 1861 FDA-approved and 102 withdrawn small-molecule drugs. Annotation was done at four levels using 30 944 target assay records, 3999 cell assay records, 585 organism assay records, and 8889 clinical trial records. Additionally, approximately 1799 repositioning protein or target sequences coupled with 856 related diseases and 1332 pathways are linked to the drug entries. Our web-based software displays a network of integrative relationships between drugs, their repositioning targets, and related diseases. The database is fully searchable and supports extensive text, sequence, chemical structure, and relational query searches. EK-DRD is freely accessible at http://www.idruglab.com/drd/index.php .
Collapse
Affiliation(s)
- Chongze Zhao
- Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , China
| | - Xi Dai
- Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , China
| | - Yecheng Li
- Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , China
| | - Qingqing Guo
- Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , China
| | - Jianhua Zhang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , China
| | - Xiaotong Zhang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , China
| | - Ling Wang
- Joint International Research Laboratory of Synthetic Biology and Medicine, Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, School of Biology and Biological Engineering , South China University of Technology , Guangzhou 510006 , China
| |
Collapse
|
153
|
Delou JMA, Souza ASO, Souza LCM, Borges HL. Highlights in Resistance Mechanism Pathways for Combination Therapy. Cells 2019; 8:E1013. [PMID: 31480389 PMCID: PMC6770082 DOI: 10.3390/cells8091013] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022] Open
Abstract
Combination chemotherapy has been a mainstay in cancer treatment for the last 60 years. Although the mechanisms of action and signaling pathways affected by most treatments with single antineoplastic agents might be relatively well understood, most combinations remain poorly understood. This review presents the most common alterations of signaling pathways in response to cytotoxic and targeted anticancer drug treatments, with a discussion of how the knowledge of signaling pathways might support and orient the development of innovative strategies for anticancer combination therapy. The ultimate goal is to highlight possible strategies of chemotherapy combinations based on the signaling pathways associated with the resistance mechanisms against anticancer drugs to maximize the selective induction of cancer cell death. We consider this review an extensive compilation of updated known information on chemotherapy resistance mechanisms to promote new combination therapies to be to discussed and tested.
Collapse
Affiliation(s)
- João M A Delou
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Alana S O Souza
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Leonel C M Souza
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Helena L Borges
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
154
|
HyperFoods: Machine intelligent mapping of cancer-beating molecules in foods. Sci Rep 2019; 9:9237. [PMID: 31270435 PMCID: PMC6610092 DOI: 10.1038/s41598-019-45349-y] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/03/2019] [Indexed: 01/02/2023] Open
Abstract
Recent data indicate that up-to 30–40% of cancers can be prevented by dietary and lifestyle measures alone. Herein, we introduce a unique network-based machine learning platform to identify putative food-based cancer-beating molecules. These have been identified through their molecular biological network commonality with clinically approved anti-cancer therapies. A machine-learning algorithm of random walks on graphs (operating within the supercomputing DreamLab platform) was used to simulate drug actions on human interactome networks to obtain genome-wide activity profiles of 1962 approved drugs (199 of which were classified as “anti-cancer” with their primary indications). A supervised approach was employed to predict cancer-beating molecules using these ‘learned’ interactome activity profiles. The validated model performance predicted anti-cancer therapeutics with classification accuracy of 84–90%. A comprehensive database of 7962 bioactive molecules within foods was fed into the model, which predicted 110 cancer-beating molecules (defined by anti-cancer drug likeness threshold of >70%) with expected capacity comparable to clinically approved anti-cancer drugs from a variety of chemical classes including flavonoids, terpenoids, and polyphenols. This in turn was used to construct a ‘food map’ with anti-cancer potential of each ingredient defined by the number of cancer-beating molecules found therein. Our analysis underpins the design of next-generation cancer preventative and therapeutic nutrition strategies.
Collapse
|
155
|
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
|
156
|
Karatzas E, Minadakis G, Kolios G, Delis A, Spyrou GM. A Web Tool for Ranking Candidate Drugs Against a Selected Disease Based on a Combination of Functional and Structural Criteria. Comput Struct Biotechnol J 2019; 17:939-945. [PMID: 31360332 PMCID: PMC6637175 DOI: 10.1016/j.csbj.2019.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/21/2019] [Accepted: 05/26/2019] [Indexed: 11/29/2022] Open
Abstract
Drug repurposing techniques allow existing drugs to be tested against diseases outside their initial spectrum, resulting in reduced cost and eliminating the long time-frames of new drug development. In silico drug repurposing further speeds up the process either by proposing drugs suitable to invert the transcriptomic profile of a disease or by indicating drugs based on their common targets or structural similarity with other drugs with similar mode of action. Such methods usually return a number of potential repurposed drugs that need to be tested against the disease in in vitro, pre-clinical and clinical studies. Thus, it is crucial to have a more sophisticated candidate drug ranking in order to start testing from the most promising chemical substances. As a means to enhance the above decision process, we present CoDReS (Composite Drug Reranking Scoring), a drug (re-)ranking web-based tool, which combines an initial drug ranking (i.e. repurposing score or hypothesis/potentiality score) with a functional score of each drug considered in conjunction with the disease under study as well as with a structural score derived from potential drugability violations. Furthermore, a structural similarity clustering is applied on the considered drugs and a handful of structural exemplars are suggested for further in vitro and in vivo validation. The user is able to filter the results further, through structural similarity examination of the candidate drugs with drugs that have failed against the queried disease where related clinical trials have been carried out. CoDReS is publicly available online at http://bioinformatics.cing.ac.cy/codres.
Collapse
Affiliation(s)
- Evangelos Karatzas
- Department of Informatics and Telecommunications, University of Athens, 15703 Athens, Greece.,The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Nicosia, 2370, Cyprus
| | - George Minadakis
- The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Nicosia, 2370, Cyprus.,The Cyprus School of Molecular Medicine, 6 International Airport Avenue, Nicosia 2370, Cyprus
| | - George Kolios
- Laboratory of Pharmacology, Department of Medicine, Democritus University of Thrace, Greece
| | - Alex Delis
- Department of Informatics and Telecommunications, University of Athens, 15703 Athens, Greece
| | - George M Spyrou
- The Cyprus Institute of Neurology and Genetics, 6 International Airport Avenue, Nicosia, 2370, Cyprus.,The Cyprus School of Molecular Medicine, 6 International Airport Avenue, Nicosia 2370, Cyprus
| |
Collapse
|
157
|
Song Y, Park IS, Kim J, Seo HR. Actinomycin D inhibits the expression of the cystine/glutamate transporter xCT via attenuation of CD133 synthesis in CD133 + HCC. Chem Biol Interact 2019; 309:108713. [PMID: 31226288 DOI: 10.1016/j.cbi.2019.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 06/07/2019] [Accepted: 06/13/2019] [Indexed: 01/03/2023]
Abstract
Liver cancer is one of the most frequently occurring types of cancer with high mortality rate. Hepatocellular carcinoma (HCC) frequently metastasizes to lung, portal vein, and portal lymph nodes and most HCCs show strong resistance to conventional anticancer drugs. Cancer stem cells (CSCs) are considered to be responsible for resistance to therapies. Hence, recent advancements in the use of liver cancer stem cells (LCSCs) are rapidly gaining recognition as an efficient and organized means for developing antitumor agents. We aimed to use a non-target-based high-throughput screening (HTS) approach to specifically target α-fetoprotein (AFP)+/cluster of differentiation (CD)133+ HCC present in mixed populations of HCC cells and hepatocytes. Herein, we identified actinomycin D (ActD) as a potential antitumor agent that significantly inhibits activity of LCSCs without affecting the co-cultured hepatocytes. To determine the mechanism of ActD-induced tumor-specificity in LCSC, we applied various cell-based assay models in vitro. In fact, ActD significantly increased reactive oxygen species (ROS) accumulation and DNA damage in Huh7 HCC cells, but not in Fa2N-4 cells, immortalized hepatocytes. Treatment of spheroid-forming LCSCs with ActD effectively decreased spheroid formation and the CD133+ HCC cell population. Importantly, these ActD-mediated effects are a result of inhibition of cystine/glutamate transporter xCT expression, via attenuation of CD133 synthesis. These results indicate that ActD suppresses stemness and malignant properties in HCC cells through destabilization of xCT, by inhibition of CD133 expression in LCSCs. The effects of ActD on LCSCs provide novel therapeutic strategies for targeting cancer stem-like cells in liver cancer.
Collapse
Affiliation(s)
- Yeonhwa Song
- Cancer Biology Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| | - I-Seul Park
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea. iseul.park.@ip-korea.org
| | - Jiho Kim
- Screening Discovery Platform, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| | - Haeng Ran Seo
- Cancer Biology Laboratory, Institut Pasteur Korea, 16, Daewangpangyo-ro 712 beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13488, Republic of Korea.
| |
Collapse
|
158
|
Agarwal G, Gupta S, Gabrani R, Gupta A, Chaudhary VK, Gupta V. Virtual screening of inhibitors against Envelope glycoprotein of Chikungunya Virus: a drug repositioning approach. Bioinformation 2019; 15:439-447. [PMID: 31312082 PMCID: PMC6614119 DOI: 10.6026/97320630015439] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 04/16/2019] [Indexed: 01/01/2023] Open
Abstract
Chikungunya virus (CHIKV) a re-emerging mosquito-borne alpha virus causes significant distress which is further accentuated in the lack of specific therapeutics or a preventive vaccine, mandating accelerated research for anti-CHIKV therapeutics. In recent years, drug repositioning has gained recognition for the curative interventions for its cost and time efficacy. CHIKV envelope proteins are considered to be the promising targets for drug discovery because of their essential role in viral attachment and entry in the host cells. In the current study, we propose structure-based virtual screening of drug molecule on the crystal structure of mature Chikungunya envelope protein (PDB 3N41) using a library of FDA approved drug molecules. Several cephalosporin drugs docked successfully within two binding sites prepared at E1-E2 interface of CHIKV envelop protein complex with significantly low binding energies. Cefmenoxime, ceforanide, cefotetan, cefonicid sodium and cefpiramide were identified as top leads with a cumulative score of -67.67, -64.90, -63.78, -61.99, and - 61.77, forming electrostatic, hydrogen and hydrophobic bonds within both the binding sites. These shortlisted leads could be potential inhibitors of E1-E2 hetero dimer in CHIKV, hence might disrupt the integrity of envelope glycoprotein leading to loss of its ability to form mature viral particles and gain entry into the host.
Collapse
Affiliation(s)
- Garima Agarwal
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP 201309, India
| | - Sanjay Gupta
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP 201309, India
| | - Reema Gabrani
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP 201309, India
| | - Amita Gupta
- Centre for Innovation in Infectious Disease Research, Education and Training, University of Delhi South Campus, Benito Juarez Marg, New Delhi 110021, India
| | - Vijay Kumar Chaudhary
- Centre for Innovation in Infectious Disease Research, Education and Training, University of Delhi South Campus, Benito Juarez Marg, New Delhi 110021, India
| | - Vandana Gupta
- Department of Microbiology, Ram Lal Anand College, University of Delhi South Campus (UDSC), Benito Juarez Marg, New Delhi 110021, India
| |
Collapse
|
159
|
Pawar G, Madden JC, Ebbrell D, Firman JW, Cronin MTD. In Silico Toxicology Data Resources to Support Read-Across and (Q)SAR. Front Pharmacol 2019; 10:561. [PMID: 31244651 PMCID: PMC6580867 DOI: 10.3389/fphar.2019.00561] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022] Open
Abstract
A plethora of databases exist online that can assist in in silico chemical or drug safety assessment. However, a systematic review and grouping of databases, based on purpose and information content, consolidated in a single source, has been lacking. To resolve this issue, this review provides a comprehensive listing of the key in silico data resources relevant to: chemical identity and properties, drug action, toxicology (including nano-material toxicity), exposure, omics, pathways, Absorption, Distribution, Metabolism and Elimination (ADME) properties, clinical trials, pharmacovigilance, patents-related databases, biological (genes, enzymes, proteins, other macromolecules etc.) databases, protein-protein interactions (PPIs), environmental exposure related, and finally databases relating to animal alternatives in support of 3Rs policies. More than nine hundred databases were identified and reviewed against criteria relating to accessibility, data coverage, interoperability or application programming interface (API), appropriate identifiers, types of in vitro, in vivo,-clinical or other data recorded and suitability for modelling, read-across, or similarity searching. This review also specifically addresses the need for solutions for mapping and integration of databases into a common platform for better translatability of preclinical data to clinical data.
Collapse
Affiliation(s)
| | | | | | | | - Mark T. D. Cronin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| |
Collapse
|
160
|
Translational role of natural coumarins and their derivatives as anticancer agents. Future Med Chem 2019; 11:1057-1082. [PMID: 31140865 DOI: 10.4155/fmc-2018-0375] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Natural coumarins and their derivatives isolated from various plants or microorganisms have inherent antioxidant, antibacterial, antifungal, antiviral and anticancer properties among many biological activities. Some of these coumarins and their derivatives lead to self-programmed cancer cell death (apoptosis) via different mechanisms, which will be discussed. The link between bacterial and viral infections to cancer compels us to highlight fascinating reports from coumarin isolation from microorganisms; comment on the recent bioavailability studies of natural or derived coumarins; and discuss our perspectives with respect to bioisosterism in coumarins, p-glycoprotein inhibition and covalent modification, and bioprobes. Overall, this review hopes to stimulate and offer in particular medicinal chemists and the reader in general an outlook on natural coumarins and their derivatives with potential for cancer therapy.
Collapse
|
161
|
Masoudi-Sobhanzadeh Y, Omidi Y, Amanlou M, Masoudi-Nejad A. DrugR+: A comprehensive relational database for drug repurposing, combination therapy, and replacement therapy. Comput Biol Med 2019; 109:254-262. [PMID: 31096089 DOI: 10.1016/j.compbiomed.2019.05.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 04/26/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
Abstract
Drug repurposing or repositioning, which introduces new applications of the existing drugs, is an emerging field in drug discovery scope. To enhance the success rate of the research and development (R&D) process in a cost- and time-effective manner, a number of pharmaceutical companies worldwide have made tremendous investments. Besides, many researchers have proposed various methods and databases for the repurposing of various drugs. However, there is not a proper and well-organized database available. To this end, for the first time, we developed a new database based on DrugBank and KEGG data, which is named "DrugR+". Our developed database provides some advantages relative to the DrugBank, and its interface supplies new capabilities for both single and synthetic repositioning of drugs. Moreover, it includes four new datasets which can be used for predicting drug-target interactions using supervised machine learning methods. As a case study, we introduced novel applications of some drugs and discussed the obtained results. A comparison of several machine learning methods on the generated datasets has also been reported in the Supplementary File. Having included several normalized tables, DrugR + has been organized to provide key information on data structures for the repurposing and combining applications of drugs. It provides the SQL query capability for professional users and an appropriate method with different options for unprofessional users. Additionally, DrugR + consists of repurposing service that accepts a drug and proposes a list of potential drugs for some usages. Taken all, DrugR+ is a free web-based database and accessible using (http://www.drugr.ir), which can be updated through a map-reduce parallel processing method to provide the most relevant information.
Collapse
Affiliation(s)
- Yosef Masoudi-Sobhanzadeh
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology and Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Massoud Amanlou
- Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, 14176-53955, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran. http://LBB.ut.ac.ir
| |
Collapse
|
162
|
Das SS, Sinha R, Chakravorty N. Integrative microRNA and gene expression analysis identifies new drug repurposing candidates for fetal hemoglobin induction in β-hemoglobinopathies. Gene 2019; 706:77-83. [PMID: 31048070 DOI: 10.1016/j.gene.2019.04.077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 04/18/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic induction of fetal hemoglobin (HbF) is one of the most promising approaches to ameliorate the severity of hemoglobinopathies like β-thalassemia and sickle cell anemia. Although several pharmacological agents have been investigated for HbF induction in adults, the majority of these are associated with significant side-effects. While drug repurposing is known to open new doors for the use of approved drugs in unexplored clinical conditions, the primary challenge lies in identifying such candidates. In this study, we aimed to identify repurposing candidates for HbF induction using a novel in silico approach utilizing microRNA-pathway-drug relationships. A computational drug repurposing strategy identified several unique candidates for HbF induction; among which Curcumin, Ginsenoside, Valproate, and Vorinostat were found to be most suitable for future trials. This study identified new drug repurposing candidates for HbF induction and demonstrates an easily adaptable methodology that can be used for other pathophysiological conditions.
Collapse
Affiliation(s)
- Sankha Subhra Das
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| | - Rashmi Sinha
- B. C. Roy Technology Hospital, Indian Institute of Technology Kharagpur, West Bengal 721302, India; Plant Hospital, Bharatiya Reserve Bank Note Mudran Private Limited (BRBNMPL), Salboni, West Bengal 721132, India
| | - Nishant Chakravorty
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, West Bengal 721302, India.
| |
Collapse
|
163
|
Nowak-Sliwinska P, Scapozza L, Ruiz i Altaba A. Drug repurposing in oncology: Compounds, pathways, phenotypes and computational approaches for colorectal cancer. Biochim Biophys Acta Rev Cancer 2019; 1871:434-454. [PMID: 31034926 PMCID: PMC6528778 DOI: 10.1016/j.bbcan.2019.04.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/09/2019] [Accepted: 04/15/2019] [Indexed: 02/08/2023]
Abstract
The strategy of using existing drugs originally developed for one disease to treat other indications has found success across medical fields. Such drug repurposing promises faster access of drugs to patients while reducing costs in the long and difficult process of drug development. However, the number of existing drugs and diseases, together with the heterogeneity of patients and diseases, notably including cancers, can make repurposing time consuming and inefficient. The key question we address is how to efficiently repurpose an existing drug to treat a given indication. As drug efficacy remains the main bottleneck for overall success, we discuss the need for machine-learning computational methods in combination with specific phenotypic studies along with mechanistic studies, chemical genetics and omics assays to successfully predict disease-drug pairs. Such a pipeline could be particularly important to cancer patients who face heterogeneous, recurrent and metastatic disease and need fast and personalized treatments. Here we focus on drug repurposing for colorectal cancer and describe selected therapeutics already repositioned for its prevention and/or treatment as well as potential candidates. We consider this review as a selective compilation of approaches and methodologies, and argue how, taken together, they could bring drug repurposing to the next level.
Collapse
Affiliation(s)
- Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland; Translational Research Center in Oncohaematology, University of Geneva, Rue Michel Servet 1, 1211 Geneva 4, Switzerland.
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, Geneva, Switzerland
| | - Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva, Rue Michel Servet 1, 1211 Geneva 4, Switzerland
| |
Collapse
|
164
|
Drug prioritization using the semantic properties of a knowledge graph. Sci Rep 2019; 9:6281. [PMID: 31000794 PMCID: PMC6472420 DOI: 10.1038/s41598-019-42806-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 03/28/2019] [Indexed: 02/01/2023] Open
Abstract
Compounds that are candidates for drug repurposing can be ranked by leveraging knowledge available in the biomedical literature and databases. This knowledge, spread across a variety of sources, can be integrated within a knowledge graph, which thereby comprehensively describes known relationships between biomedical concepts, such as drugs, diseases, genes, etc. Our work uses the semantic information between drug and disease concepts as features, which are extracted from an existing knowledge graph that integrates 200 different biological knowledge sources. RepoDB, a standard drug repurposing database which describes drug-disease combinations that were approved or that failed in clinical trials, is used to train a random forest classifier. The 10-times repeated 10-fold cross-validation performance of the classifier achieves a mean area under the receiver operating characteristic curve (AUC) of 92.2%. We apply the classifier to prioritize 21 preclinical drug repurposing candidates that have been suggested for Autosomal Dominant Polycystic Kidney Disease (ADPKD). Mozavaptan, a vasopressin V2 receptor antagonist is predicted to be the drug most likely to be approved after a clinical trial, and belongs to the same drug class as tolvaptan, the only treatment for ADPKD that is currently approved. We conclude that semantic properties of concepts in a knowledge graph can be exploited to prioritize drug repurposing candidates for testing in clinical trials.
Collapse
|
165
|
García-Serradilla M, Risco C, Pacheco B. Drug repurposing for new, efficient, broad spectrum antivirals. Virus Res 2019; 264:22-31. [PMID: 30794895 PMCID: PMC7114681 DOI: 10.1016/j.virusres.2019.02.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 12/26/2022]
Abstract
Emerging viruses are a major threat to human health. Recent outbreaks have emphasized the urgent need for new antiviral treatments. For several pathogenic viruses, considerable efforts have focused on vaccine development. However, during epidemics infected individuals need to be treated urgently. High-throughput screening of clinically tested compounds provides a rapid means to identify undiscovered, antiviral functions for well-characterized therapeutics. Repurposed drugs can bypass part of the early cost and time needed for validation and authorization. In this review we describe recent efforts to find broad spectrum antivirals through drug repurposing. We have chosen several candidates and propose strategies to understand their mechanism of action and to determine how resistance to antivirals develops in infected cells.
Collapse
Affiliation(s)
- Moisés García-Serradilla
- Cell Structure Laboratory, National Center for Biotechnology, National Research Council, CNB-CSIC, Darwin 3, UAM, campus de Cantoblanco, 28049 Madrid, Spain
| | - Cristina Risco
- Cell Structure Laboratory, National Center for Biotechnology, National Research Council, CNB-CSIC, Darwin 3, UAM, campus de Cantoblanco, 28049 Madrid, Spain.
| | - Beatriz Pacheco
- Cell Structure Laboratory, National Center for Biotechnology, National Research Council, CNB-CSIC, Darwin 3, UAM, campus de Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
166
|
Almenar-Pérez E, Sánchez-Fito T, Ovejero T, Nathanson L, Oltra E. Impact of Polypharmacy on Candidate Biomarker miRNomes for the Diagnosis of Fibromyalgia and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: Striking Back on Treatments. Pharmaceutics 2019; 11:126. [PMID: 30889846 PMCID: PMC6471415 DOI: 10.3390/pharmaceutics11030126] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 12/14/2022] Open
Abstract
Fibromyalgia (FM) and myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) are diseases of unknown etiology presenting complex and often overlapping symptomatology. Despite promising advances on the study of miRNomes of these diseases, no validated molecular diagnostic biomarker yet exists. Since FM and ME/CFS patient treatments commonly include polypharmacy, it is of concern that biomarker miRNAs are masked by drug interactions. Aiming at discriminating between drug-effects and true disease-associated differential miRNA expression, we evaluated the potential impact of commonly prescribed drugs on disease miRNomes, as reported by the literature. By using the web search tools SM2miR, Pharmaco-miR, and repoDB, we found a list of commonly prescribed drugs that impact FM and ME/CFS miRNomes and therefore could be interfering in the process of biomarker discovery. On another end, disease-associated miRNomes may incline a patient's response to treatment and toxicity. Here, we explored treatments for diseases in general that could be affected by FM and ME/CFS miRNomes, finding a long list of them, including treatments for lymphoma, a type of cancer affecting ME/CFS patients at a higher rate than healthy population. We conclude that FM and ME/CFS miRNomes could help refine pharmacogenomic/pharmacoepigenomic analysis to elevate future personalized medicine and precision medicine programs in the clinic.
Collapse
Affiliation(s)
- Eloy Almenar-Pérez
- Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain.
| | - Teresa Sánchez-Fito
- Escuela de Doctorado, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain.
| | - Tamara Ovejero
- School of Medicine, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain.
| | - Lubov Nathanson
- Kiran C Patel College of Osteopathic Medicine, Nova Southeastern University, Ft Lauderdale, FL 33314, USA.
- Institute for Neuro Immune Medicine, Nova Southeastern University, Ft Lauderdale, FL 33314, USA.
| | - Elisa Oltra
- School of Medicine, Universidad Católica de Valencia San Vicente Mártir, 46001 Valencia, Spain.
- Unidad Mixta CIPF-UCV, Centro de Investigación Príncipe Felipe, 46012 Valencia, Spain.
| |
Collapse
|
167
|
Xuan P, Cao Y, Zhang T, Wang X, Pan S, Shen T. Drug repositioning through integration of prior knowledge and projections of drugs and diseases. Bioinformatics 2019; 35:4108-4119. [DOI: 10.1093/bioinformatics/btz182] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 02/24/2019] [Accepted: 03/12/2019] [Indexed: 12/20/2022] Open
Abstract
Abstract
Motivation
Identifying and developing novel therapeutic effects for existing drugs contributes to reduction of drug development costs. Most of the previous methods focus on integration of the heterogeneous data of drugs and diseases from multiple sources for predicting the candidate drug–disease associations. However, they fail to take the prior knowledge of drugs and diseases and their sparse characteristic into account. It is essential to develop a method that exploits the more useful information to predict the reliable candidate associations.
Results
We present a method based on non-negative matrix factorization, DisDrugPred, to predict the drug-related candidate disease indications. A new type of drug similarity is firstly calculated based on their associated diseases. DisDrugPred completely integrates two types of disease similarities, the associations between drugs and diseases, and the various similarities between drugs from different levels including the chemical structures of drugs, the target proteins of drugs, the diseases associated with drugs and the side effects of drugs. The prior knowledge of drugs and diseases and the sparse characteristic of drug–disease associations provide a deep biological perspective for capturing the relationships between drugs and diseases. Simultaneously, the possibility that a drug is associated with a disease is also dependant on their projections in the low-dimension feature space. Therefore, DisDrugPred deeply integrates the diverse prior knowledge, the sparse characteristic of associations and the projections of drugs and diseases. DisDrugPred achieves superior prediction performance than several state-of-the-art methods for drug–disease association prediction. During the validation process, DisDrugPred also can retrieve more actual drug–disease associations in the top part of prediction result which often attracts more attention from the biologists. Moreover, case studies on five drugs further confirm DisDrugPred’s ability to discover potential candidate disease indications for drugs.
Availability and implementation
The fourth type of drug similarity and the predicted candidates for all the drugs are available at https://github.com/pingxuan-hlju/DisDrugPred.
Supplementary information
Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Ping Xuan
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| | - Yangkun Cao
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| | - Tiangang Zhang
- School of Mathematical Science, Heilongjiang University, Harbin, China
| | - Xiao Wang
- School of Computer Science, Beijing University of Posts and Telecommunications, Beijing, China
| | - Shuxiang Pan
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| | - Tonghui Shen
- School of Computer Science and Technology, Heilongjiang University, Harbin, China
| |
Collapse
|
168
|
Farha MA, Brown ED. Drug repurposing for antimicrobial discovery. Nat Microbiol 2019; 4:565-577. [PMID: 30833727 DOI: 10.1038/s41564-019-0357-1] [Citation(s) in RCA: 222] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/03/2019] [Indexed: 12/17/2022]
Abstract
Antimicrobial resistance continues to be a public threat on a global scale. The ongoing need to develop new antimicrobial drugs that are effective against multi-drug-resistant pathogens has spurred the research community to invest in various drug discovery strategies, one of which is drug repurposing-the process of finding new uses for existing drugs. While still nascent in the antimicrobial field, the approach is gaining traction in both the public and private sector. While the approach has particular promise in fast-tracking compounds into clinical studies, it nevertheless has substantial obstacles to success. This Review covers the art of repurposing existing drugs for antimicrobial purposes. We discuss enabling screening platforms for antimicrobial discovery and present encouraging findings of novel antimicrobial therapeutic strategies. Also covered are general advantages of repurposing over de novo drug development and challenges of the strategy, including scientific, intellectual property and regulatory issues.
Collapse
Affiliation(s)
- Maya A Farha
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Eric D Brown
- Michael G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
169
|
Regan-Fendt KE, Xu J, DiVincenzo M, Duggan MC, Shakya R, Na R, Carson WE, Payne PRO, Li F. Synergy from gene expression and network mining (SynGeNet) method predicts synergistic drug combinations for diverse melanoma genomic subtypes. NPJ Syst Biol Appl 2019; 5:6. [PMID: 30820351 PMCID: PMC6391384 DOI: 10.1038/s41540-019-0085-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 01/23/2019] [Indexed: 12/31/2022] Open
Abstract
Systems biology perspectives are crucial for understanding the pathophysiology of complex diseases, and therefore hold great promise for the discovery of novel treatment strategies. Drug combinations have been shown to improve durability and reduce resistance to available first-line therapies in a variety of cancers; however, traditional drug discovery approaches are prohibitively cost and labor-intensive to evaluate large-scale matrices of potential drug combinations. Computational methods are needed to efficiently model complex interactions of drug target pathways and identify mechanisms underlying drug combination synergy. In this study, we employ a computational approach, SynGeNet (Synergy from Gene expression and Network mining), which integrates transcriptomics-based connectivity mapping and network centrality analysis to analyze disease networks and predict drug combinations. As an exemplar of a disease in which combination therapies demonstrate efficacy in genomic-specific contexts, we investigate malignant melanoma. We employed SynGeNet to generate drug combination predictions for each of the four major genomic subtypes of melanoma (BRAF, NRAS, NF1, and triple wild type) using publicly available gene expression and mutation data. We validated synergistic drug combinations predicted by our method across all genomic subtypes using results from a high-throughput drug screening study across. Finally, we prospectively validated the drug combination for BRAF-mutant melanoma that was top ranked by our approach, vemurafenib (BRAF inhibitor) + tretinoin (retinoic acid receptor agonist), using both in vitro and in vivo models of BRAF-mutant melanoma and RNA-sequencing analysis of drug-treated melanoma cells to validate the predicted mechanisms. Our approach is applicable to a wide range of disease domains, and, importantly, can model disease-relevant protein subnetworks in precision medicine contexts.
Collapse
Affiliation(s)
- Kelly E Regan-Fendt
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Jielin Xu
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Mallory DiVincenzo
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Megan C Duggan
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Reena Shakya
- Target Validation Shared Resource, The Ohio State University, Columbus, OH, USA
| | - Ryejung Na
- Target Validation Shared Resource, The Ohio State University, Columbus, OH, USA
| | - William E Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Philip R O Payne
- Institute for Informatics, Washington University in St. Louis, St. Louis, MO, USA
| | - Fuhai Li
- Institute for Informatics, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Pediatrics, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
170
|
Goody D, Gupta SK, Engelmann D, Spitschak A, Marquardt S, Mikkat S, Meier C, Hauser C, Gundlach JP, Egberts JH, Martin H, Schumacher T, Trauzold A, Wolkenhauer O, Logotheti S, Pützer BM. Drug Repositioning Inferred from E2F1-Coregulator Interactions Studies for the Prevention and Treatment of Metastatic Cancers. Theranostics 2019; 9:1490-1509. [PMID: 30867845 PMCID: PMC6401510 DOI: 10.7150/thno.29546] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
Metastasis management remains a long-standing challenge. High abundance of E2F1 triggers tumor progression by developing protein-protein interactions (PPI) with coregulators that enhance its potential to activate a network of prometastatic transcriptional targets. Methods: To identify E2F1-coregulators, we integrated high-throughput Co-immunoprecipitation (IP)/mass spectometry, GST-pull-down assays, and structure modeling. Potential inhibitors of PPI discovered were found by bioinformatics-based pharmacophore modeling, and transcriptome profiling was conducted to screen for coregulated downstream targets. Expression and target gene regulation was validated using qRT-PCR, immunoblotting, chromatin IP, and luciferase assays. Finally, the impact of the E2F1-coregulator complex and its inhibiting drug on metastasis was investigated in vitro in different cancer entities and two mouse metastasis models. Results: We unveiled that E2F1 forms coactivator complexes with metastasis-associated protein 1 (MTA1) which, in turn, is directly upregulated by E2F1. The E2F1:MTA1 complex potentiates hyaluronan synthase 2 (HAS2) expression, increases hyaluronan production and promotes cell motility. Disruption of this prometastatic E2F1:MTA1 interaction reduces hyaluronan synthesis and infiltration of tumor-associated macrophages in the tumor microenvironment, thereby suppressing metastasis. We further demonstrate that E2F1:MTA1 assembly is abrogated by small-molecule, FDA-approved drugs. Treatment of E2F1/MTA1-positive, highly aggressive, circulating melanoma cells and orthotopic pancreatic tumors with argatroban prevents metastasis and cancer relapses in vivo through perturbation of the E2F1:MTA1/HAS2 axis. Conclusion: Our results propose argatroban as an innovative, E2F-coregulator-based, antimetastatic drug. Cancer patients with the infaust E2F1/MTA1/HAS2 signature will likely benefit from drug repositioning.
Collapse
|
171
|
Potential for Drug Repositioning of Midazolam for Dentin Regeneration. Int J Mol Sci 2019; 20:ijms20030670. [PMID: 30720745 PMCID: PMC6387224 DOI: 10.3390/ijms20030670] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/24/2019] [Accepted: 01/31/2019] [Indexed: 01/03/2023] Open
Abstract
Drug repositioning promises the advantages of reducing costs and expediting approval schedules. An induction of the anesthetic and sedative drug; midazolam (MDZ), regulates inhibitory neurotransmitters in the vertebrate nervous system. In this study we show the potential for drug repositioning of MDZ for dentin regeneration. A porcine dental pulp-derived cell line (PPU-7) that we established was cultured in MDZ-only, the combination of MDZ with bone morphogenetic protein 2, and the combination of MDZ with transforming growth factor-beta 1. The differentiation of PPU-7 into odontoblasts was investigated at the cell biological and genetic level. Mineralized nodules formed in PPU-7 were characterized at the protein and crystal engineering levels. The MDZ-only treatment enhanced the alkaline phosphatase activity and mRNA levels of odontoblast differentiation marker genes, and precipitated nodule formation containing a dentin-specific protein (dentin phosphoprotein). The nodules consisted of randomly oriented hydroxyapatite nanorods and nanoparticles. The morphology, orientation, and chemical composition of the hydroxyapatite crystals were similar to those of hydroxyapatite that had transformed from amorphous calcium phosphate nanoparticles, as well as the hydroxyapatite in human molar dentin. Our investigation showed that a combination of MDZ and PPU-7 cells possesses high potential of drug repositioning for dentin regeneration.
Collapse
|
172
|
Griesenauer RH, Schillebeeckx C, Kinch MS. Assessing the public landscape of clinical-stage pharmaceuticals through freely available online databases. Drug Discov Today 2019; 24:1010-1016. [PMID: 30690196 DOI: 10.1016/j.drudis.2019.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 12/19/2018] [Accepted: 01/22/2019] [Indexed: 12/23/2022]
Abstract
Several public databases have emerged over the past decade to enable chemo- and bio-informatics research in the field of drug development. To a naive observer, as well as many seasoned professionals, the differences among many drug databases are unclear. We assessed the availability of all pharmaceuticals with evidence of clinical testing (i.e., been in at least a Phase I clinical trial) and highlight the major differences and similarities between public databases containing clinically tested pharmaceuticals. We review a selection of the most recent and prominent databases including: ChEMBL, CRIB NME, DrugBank, DrugCentral, PubChem, repoDB, SuperDrug2 and WITHDRAWN, and found that ∼11700 unique active pharmaceutical ingredients are available in the public domain, with evidence of clinical testing.
Collapse
Affiliation(s)
- Rebekah H Griesenauer
- Center for Research Innovation in Biotechnology, Washington University in St Louis, MO 63130, USA
| | | | - Michael S Kinch
- Center for Research Innovation in Biotechnology, Washington University in St Louis, MO 63130, USA.
| |
Collapse
|
173
|
Ursu O, Holmes J, Bologa CG, Yang JJ, Mathias SL, Stathias V, Nguyen DT, Schürer S, Oprea T. DrugCentral 2018: an update. Nucleic Acids Res 2019; 47:D963-D970. [PMID: 30371892 PMCID: PMC6323925 DOI: 10.1093/nar/gky963] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/02/2018] [Accepted: 10/26/2018] [Indexed: 01/21/2023] Open
Abstract
DrugCentral is a drug information resource (http://drugcentral.org) open to the public since 2016 and previously described in the 2017 Nucleic Acids Research Database issue. Since the 2016 release, 103 new approved drugs were updated. The following new data sources have been included: Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS), FDA Orange Book information, L1000 gene perturbation profile distance/similarity matrices and estimated protonation constants. New and existing entries have been updated with the latest information from scientific literature, drug labels and external databases. The web interface has been updated to display and query new data. The full database dump and data files are available for download from the DrugCentral website.
Collapse
Affiliation(s)
- Oleg Ursu
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Jayme Holmes
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Cristian G Bologa
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Jeremy J Yang
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Stephen L Mathias
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| | - Vasileios Stathias
- Center for Computational Science, Miller School of Medicine, University of Miami, Coral Gables, FL 33146, USA
| | - Dac-Trung Nguyen
- National Center for Advancing Translational Science, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Stephan Schürer
- Center for Computational Science, Miller School of Medicine, University of Miami, Coral Gables, FL 33146, USA
| | - Tudor Oprea
- Translational Informatics Division, Department of Internal Medicine, The University of New Mexico Health Science Center, Albuquerque, NM 87131, USA
| |
Collapse
|
174
|
Grenier L, Hu P. Computational drug repurposing for inflammatory bowel disease using genetic information. Comput Struct Biotechnol J 2019; 17:127-135. [PMID: 30728920 PMCID: PMC6352300 DOI: 10.1016/j.csbj.2019.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 12/22/2022] Open
Abstract
As knowledge of the genetics behind inflammatory bowel disease (IBD) has continually improved, there has been a demand for methods that can use this data in a clinically significant way. Genome-wide association analyses for IBD have identified 232 risk genetic loci for the disorder. While identification of these risk loci enriches our understanding of the underlying biology of the disorder, their identification does not serve a clinical purpose. A potential use of this genetic information is to look for potential IBD drugs that target these loci in a procedure known as drug repurposing. The demand for new drug treatments for IBD is high due to the side effects and high costs of current treatments. We hypothesize that IBD genetic variants obtained from GWAS and the candidate genes prioritized from the variants have a causal relationship with IBD drug targets. A computational drug repositioning study was done due to its efficiency and inexpensiveness compared to traditional in vitro or biochemical approaches. Our approach for drug repurposing was multi-layered; it not only focused on the interactions between drugs and risk IBD genes, but also the interactions between drugs and all of the biological pathways the risk genes are involved in. We prioritized IBD candidate genes using identified genetic variants and identified potential drug targets and drugs that can be potentially repositioned or developed for IBD using the identified candidate genes. Our analysis strategy can be applied to repurpose drugs for other complex diseases using their risk genes identified from genetic analysis.
Collapse
Affiliation(s)
- Liam Grenier
- Department of Biochemistry and Medical Genetics and The George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Winnipeg, MB, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics and The George and Fay Yee Centre for Healthcare Innovation, University of Manitoba, Winnipeg, MB, Canada
- Department of Electrical and Computer Engineering, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
175
|
Griesenauer RH, Schillebeeckx C, Kinch MS. CDEK: Clinical Drug Experience Knowledgebase. Database (Oxford) 2019; 2019:baz087. [PMID: 31411687 PMCID: PMC6693031 DOI: 10.1093/database/baz087] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/29/2019] [Accepted: 06/07/2019] [Indexed: 12/16/2022]
Abstract
The Clinical Drug Experience Knowledgebase (CDEK) is a database and web platform of active pharmaceutical ingredients with evidence of clinical testing as well as the organizations involved in their research and development. CDEK was curated by disambiguating intervention and organization names from ClinicalTrials.gov and cross-referencing these entries with other prominent drug databases. Approximately 43% of active pharmaceutical ingredients in the CDEK database were sourced from ClinicalTrials.gov and cannot be found in any other prominent compound-oriented database. The contents of CDEK are structured around three pillars: active pharmaceutical ingredients (n = 22 292), clinical trials (n = 127 223) and organizations (n = 24 728). The envisioned use of the CDEK is to support the investigation of many aspects of drug development, including discovery, repurposing opportunities, chemo- and bio-informatics, clinical and translational research and regulatory sciences.
Collapse
Affiliation(s)
- Rebekah H Griesenauer
- Center for Research Innovation in Biotechnology, Washington University in St. Louis, MO 63110, USA
| | | | - Michael S Kinch
- Center for Research Innovation in Biotechnology, Washington University in St. Louis, MO 63110, USA
| |
Collapse
|
176
|
Song Y, Lee SY, Kim AR, Kim S, Heo J, Shum D, Kim SH, Choi I, Lee YJ, Seo HR. Identification of radiation-induced EndMT inhibitors through cell-based phenomic screening. FEBS Open Bio 2018; 9:82-91. [PMID: 30652076 PMCID: PMC6325571 DOI: 10.1002/2211-5463.12552] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/09/2018] [Accepted: 10/19/2018] [Indexed: 12/24/2022] Open
Abstract
Radiation‐induced pulmonary fibrosis (RIPF) triggers physiological abnormalities. Endothelial‐to‐mesenchymal transition (EndMT) is the phenotypic conversion of endothelial cells to fibroblast‐like cells and is involved in RIPF. In this study, we established a phenomic screening platform to measure radiation‐induced stress fibers and optimized the conditions for high‐throughput screening using human umbilical vein endothelial cells (HUVECs) to develop compounds targeting RIPF. The results of screening indicated that CHIR‐99021 reduced radiation‐induced fibrosis, as evidenced by an enlargement of cell size and increases in actin stress fibers and α‐smooth muscle actin expression. These effects were elicited without inducing serious toxicity in HUVECs, and the cytotoxic effect of ionizing radiation (IR) in nonsmall cell lung cancer was also enhanced. These results demonstrate that CHIR‐99021 enhanced the effects of IR therapy by suppressing radiation‐induced EndMT in lung cancer.
Collapse
Affiliation(s)
- Yeonhwa Song
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Su-Yeon Lee
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - A-Ram Kim
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Sanghwa Kim
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Jinyeong Heo
- Assay Development and Screening Institut Pasteur Korea Seongnam-si Korea
| | - David Shum
- Assay Development and Screening Institut Pasteur Korea Seongnam-si Korea
| | - Se-Hyuk Kim
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| | - Inhee Choi
- Medicinal Chemistry Institut Pasteur Korea Seongnam-si Korea
| | - Yoon-Jin Lee
- Division of Radiation Effects Korea Institute of Radiological and Medical Sciences Seoul Korea
| | - Haeng Ran Seo
- Cancer Biology Laboratory Institut Pasteur Korea Seongnam-si Korea
| |
Collapse
|
177
|
Wilson JL, Racz R, Liu T, Adeniyi O, Sun J, Ramamoorthy A, Pacanowski M, Altman R. PathFX provides mechanistic insights into drug efficacy and safety for regulatory review and therapeutic development. PLoS Comput Biol 2018; 14:e1006614. [PMID: 30532240 PMCID: PMC6285459 DOI: 10.1371/journal.pcbi.1006614] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/31/2018] [Indexed: 12/14/2022] Open
Abstract
Failure to demonstrate efficacy and safety issues are important reasons that drugs do not reach the market. An incomplete understanding of how drugs exert their effects hinders regulatory and pharmaceutical industry projections of a drug's benefits and risks. Signaling pathways mediate drug response and while many signaling molecules have been characterized for their contribution to disease or their role in drug side effects, our knowledge of these pathways is incomplete. To better understand all signaling molecules involved in drug response and the phenotype associations of these molecules, we created a novel method, PathFX, a non-commercial entity, to identify these pathways and drug-related phenotypes. We benchmarked PathFX by identifying drugs' marketed disease indications and reported a sensitivity of 41%, a 2.7-fold improvement over similar approaches. We then used PathFX to strengthen signals for drug-adverse event pairs occurring in the FDA Adverse Event Reporting System (FAERS) and also identified opportunities for drug repurposing for new diseases based on interaction paths that associated a marketed drug to that disease. By discovering molecular interaction pathways, PathFX improved our understanding of drug associations to safety and efficacy phenotypes. The algorithm may provide a new means to improve regulatory and therapeutic development decisions.
Collapse
Affiliation(s)
- Jennifer L. Wilson
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
| | - Rebecca Racz
- Division of Applied Regulatory Science, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Tianyun Liu
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
| | - Oluseyi Adeniyi
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Jielin Sun
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Anuradha Ramamoorthy
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Michael Pacanowski
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring Maryland, United States of America
| | - Russ Altman
- Department of Bioengineering, Stanford University, Palo Alto California, United States of America
- Department of Genetics, Stanford University, Palo Alto California, United States of America
| |
Collapse
|
178
|
Chávez-Fumagalli MA, Lage DP, Tavares GSV, Mendonça DVC, Dias DS, Ribeiro PAF, Ludolf F, Costa LE, Coelho VTS, Coelho EAF. In silico Leishmania proteome mining applied to identify drug target potential to be used to treat against visceral and tegumentary leishmaniasis. J Mol Graph Model 2018; 87:89-97. [PMID: 30522092 DOI: 10.1016/j.jmgm.2018.11.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 11/12/2018] [Accepted: 11/28/2018] [Indexed: 12/11/2022]
Abstract
New therapeutic strategies against leishmaniasis are desirable, since the treatment against disease presents problems, such as the toxicity, high cost and/or parasite resistance. As consequence, new antileishmanial compounds are necessary to be identified, as presenting high activity against Leishmania, but low toxicity in mammalian hosts. In the present study, a Leishmania proteome mining strategy was developed, in order to select new drug targets with low homology to human proteins, but that are considered relevant for the parasite' survival. Results showed a hypothetical protein, which was functionally annotated as a glucosidase-like protein, as presenting such characteristics. This protein was associated with the metabolic network of the N-Glycan biosynthesis pathway in Leishmania, and two specific inhibitors - acarbose and miglitol - were predicted to be potential targets against it. In this context, miglitol [1-(2-Hydroxyethyl)-2-(hydroxymethyl)piperidine-3,4,5-triol] was tested against stationary promastigotes and axenic amastigotes of the Leishmania amazonensis and L. infantum species, and results showed high values of antileishmanial inhibition against both parasite species. Miglitol showed also efficacy in the treatment of Leishmania-infected macrophages; thus denoting its potential use as an antileishmanial candidate. In conclusion, this work presents a new drug target identified by a proteome mining strategy associated with bioinformatics tools, and suggested its use as a possible candidate to be applied in the treatment against disease.
Collapse
Affiliation(s)
- Miguel A Chávez-Fumagalli
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniela P Lage
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Grasiele S V Tavares
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Débora V C Mendonça
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Daniel S Dias
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Patrícia A F Ribeiro
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fernanda Ludolf
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lourena E Costa
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vinicio T S Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eduardo A F Coelho
- Programa de Pós-Graduação em Ciências da Saúde: Infectologia e Medicina Tropical, Faculdade de Medicina, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Departamento de Patologia Clínica, COLTEC, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
179
|
Capoci IRG, Faria DR, Sakita KM, Rodrigues-Vendramini FAV, Bonfim-Mendonça PDS, Becker TCA, Kioshima ÉS, Svidzinski TIE, Maigret B. Repurposing approach identifies new treatment options for invasive fungal disease. Bioorg Chem 2018; 84:87-97. [PMID: 30496872 DOI: 10.1016/j.bioorg.2018.11.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/17/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022]
Abstract
Drug repositioning is the process of discovery, validation and marketing of previously approved drugs for new indications. Our aim was drug repositioning, using ligand-based and structure-based computational methods, of compounds that are similar to two hit compounds previously selected by our group that show promising antifungal activity. Through the ligand-based method, 100 compounds from each of three databases (MDDR, DrugBank and TargetMol) were selected by the Tanimoto coefficient, as similar to LMM5 or LMM11. These compounds were analyzed by the scaffold trees, and up to 10 compounds from each database were selected. The structure-based method (molecular docking) using thioredoxin reductase as the target drug was performed as a complementary approach, resulting in six compounds that were tested in an in vitro assay. All compounds, particularly raltegravir, showed antifungal activity against the genus Paracoccidioides. Raltegravir, an antiviral drug, showed promising antifungal activity against the experimental murine paracoccidioidomycosis, with significant reduction of the fungal burden and decreased alterations in the lung structure of mice treated with 1 mg/kg of raltegravir. In conclusion, the combination of two in silico methods for drug repositioning was able to select an antiviral drug with promising antifungal activity for treatment of paracoccidioidomycosis.
Collapse
Affiliation(s)
| | - Daniella Renata Faria
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, Brazil
| | - Karina Mayumi Sakita
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, Brazil
| | | | | | | | - Érika Seki Kioshima
- Department of Clinical Analysis and Biomedicine, State University of Maringá, Maringá, Paraná, Brazil
| | | | | |
Collapse
|
180
|
Wu Z, Li W, Liu G, Tang Y. Network-Based Methods for Prediction of Drug-Target Interactions. Front Pharmacol 2018; 9:1134. [PMID: 30356768 PMCID: PMC6189482 DOI: 10.3389/fphar.2018.01134] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/18/2018] [Indexed: 01/10/2023] Open
Abstract
Drug-target interaction (DTI) is the basis of drug discovery. However, it is time-consuming and costly to determine DTIs experimentally. Over the past decade, various computational methods were proposed to predict potential DTIs with high efficiency and low costs. These methods can be roughly divided into several categories, such as molecular docking-based, pharmacophore-based, similarity-based, machine learning-based, and network-based methods. Among them, network-based methods, which do not rely on three-dimensional structures of targets and negative samples, have shown great advantages over the others. In this article, we focused on network-based methods for DTI prediction, in particular our network-based inference (NBI) methods that were derived from recommendation algorithms. We first introduced the methodologies and evaluation of network-based methods, and then the emphasis was put on their applications in a wide range of fields, including target prediction and elucidation of molecular mechanisms of therapeutic effects or safety problems. Finally, limitations and perspectives of network-based methods were discussed. In a word, network-based methods provide alternative tools for studies in drug repurposing, new drug discovery, systems pharmacology and systems toxicology.
Collapse
Affiliation(s)
| | | | | | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
181
|
Piñero J, Gonzalez-Perez A, Guney E, Aguirre-Plans J, Sanz F, Oliva B, Furlong LI. Network, Transcriptomic and Genomic Features Differentiate Genes Relevant for Drug Response. Front Genet 2018; 9:412. [PMID: 30319692 PMCID: PMC6168038 DOI: 10.3389/fgene.2018.00412] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/05/2018] [Indexed: 11/13/2022] Open
Abstract
Understanding the mechanisms underlying drug therapeutic action and toxicity is crucial for the prevention and management of drug adverse reactions, and paves the way for a more efficient and rational drug design. The characterization of drug targets, drug metabolism proteins, and proteins associated to side effects according to their expression patterns, their tolerance to genomic variation and their role in cellular networks, is a necessary step in this direction. In this contribution, we hypothesize that different classes of proteins involved in the therapeutic effect of drugs and in their adverse effects have distinctive transcriptomics, genomics and network features. We explored the properties of these proteins within global and organ-specific interactomes, using multi-scale network features, evaluated their gene expression profiles in different organs and tissues, and assessed their tolerance to loss-of-function variants leveraging data from 60K subjects. We found that drug targets that mediate side effects are more central in cellular networks, more intolerant to loss-of-function variation, and show a wider breadth of tissue expression than targets not mediating side effects. In contrast, drug metabolizing enzymes and transporters are less central in the interactome, more tolerant to deleterious variants, and are more constrained in their tissue expression pattern. Our findings highlight distinctive features of proteins related to drug action, which could be applied to prioritize drugs with fewer probabilities of causing side effects.
Collapse
Affiliation(s)
- Janet Piñero
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Abel Gonzalez-Perez
- Institute for Research in Biomedicine, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Emre Guney
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Joaquim Aguirre-Plans
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Ferran Sanz
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Baldo Oliva
- Structural Bioinformatics Group, Research Programme on Biomedical Informatics, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| | - Laura I Furlong
- Integrative Biomedical Informatics Group, Research Programme on Biomedical Informatics, Hospital del Mar Medical Research Institute, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
| |
Collapse
|
182
|
Nishimura Y, Hara H. Editorial: Drug Repositioning: Current Advances and Future Perspectives. Front Pharmacol 2018; 9:1068. [PMID: 30294274 PMCID: PMC6158627 DOI: 10.3389/fphar.2018.01068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/03/2018] [Indexed: 01/08/2023] Open
Affiliation(s)
- Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
183
|
Bakal G, Talari P, Kakani EV, Kavuluru R. Exploiting semantic patterns over biomedical knowledge graphs for predicting treatment and causative relations. J Biomed Inform 2018; 82:189-199. [PMID: 29763706 PMCID: PMC6070294 DOI: 10.1016/j.jbi.2018.05.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 01/31/2018] [Accepted: 05/09/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND Identifying new potential treatment options for medical conditions that cause human disease burden is a central task of biomedical research. Since all candidate drugs cannot be tested with animal and clinical trials, in vitro approaches are first attempted to identify promising candidates. Likewise, identifying different causal relations between biomedical entities is also critical to understand biomedical processes. Generally, natural language processing (NLP) and machine learning are used to predict specific relations between any given pair of entities using the distant supervision approach. OBJECTIVE To build high accuracy supervised predictive models to predict previously unknown treatment and causative relations between biomedical entities based only on semantic graph pattern features extracted from biomedical knowledge graphs. METHODS We used 7000 treats and 2918 causes hand-curated relations from the UMLS Metathesaurus to train and test our models. Our graph pattern features are extracted from simple paths connecting biomedical entities in the SemMedDB graph (based on the well-known SemMedDB database made available by the U.S. National Library of Medicine). Using these graph patterns connecting biomedical entities as features of logistic regression and decision tree models, we computed mean performance measures (precision, recall, F-score) over 100 distinct 80-20% train-test splits of the datasets. For all experiments, we used a positive:negative class imbalance of 1:10 in the test set to model relatively more realistic scenarios. RESULTS Our models predict treats and causes relations with high F-scores of 99% and 90% respectively. Logistic regression model coefficients also help us identify highly discriminative patterns that have an intuitive interpretation. We are also able to predict some new plausible relations based on false positives that our models scored highly based on our collaborations with two physician co-authors. Finally, our decision tree models are able to retrieve over 50% of treatment relations from a recently created external dataset. CONCLUSIONS We employed semantic graph patterns connecting pairs of candidate biomedical entities in a knowledge graph as features to predict treatment/causative relations between them. We provide what we believe is the first evidence in direct prediction of biomedical relations based on graph features. Our work complements lexical pattern based approaches in that the graph patterns can be used as additional features for weakly supervised relation prediction.
Collapse
Affiliation(s)
- Gokhan Bakal
- Department of Computer Science, University of Kentucky, United States.
| | - Preetham Talari
- Division of Hospital Medicine, Department of Internal Medicine, University of Kentucky, United States.
| | - Elijah V Kakani
- Division of Hospital Medicine, Department of Internal Medicine, University of Kentucky, United States.
| | - Ramakanth Kavuluru
- Division of Biomedical Informatics, Department of Internal Medicine, University of Kentucky, United States; Department of Computer Science, University of Kentucky, United States.
| |
Collapse
|
184
|
Kang S, Lee JM, Jeon B, Elkamhawy A, Paik S, Hong J, Oh SJ, Paek SH, Lee CJ, Hassan AHE, Kang SS, Roh EJ. Repositioning of the antipsychotic trifluoperazine: Synthesis, biological evaluation and in silico study of trifluoperazine analogs as anti-glioblastoma agents. Eur J Med Chem 2018; 151:186-198. [PMID: 29614416 DOI: 10.1016/j.ejmech.2018.03.055] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/18/2018] [Accepted: 03/19/2018] [Indexed: 11/25/2022]
Abstract
Repositioning of the antipsychotic drug trifluoperazine for treatment of glioblastoma, an aggressive brain tumor, has been previously suggested. However, trifluoperazine did not increase the survival time in mice models of glioblastoma. In attempt to identify an effective trifluoperazine analog, fourteen compounds have been synthesized and biologically in vitro and in vivo assessed. Using MTT assay, compounds 3dc and 3dd elicited 4-5 times more potent inhibitory activity than trifluoperazine with IC50 = 2.3 and 2.2 μM against U87MG glioblastoma cells, as well as, IC50 = 2.2 and 2.1 μM against GBL28 human glioblastoma patient derived primary cells, respectively. Furthermore, they have shown a reasonable selectivity for glioblastoma cells over NSC normal neural cell. In vivo evaluation of analog 3dc confirmed its advantageous effect on reduction of tumor size and increasing the survival time in brain xenograft mouse model of glioblastoma. Molecular modeling simulation provided a reasonable explanation for the observed variation in the capability of the synthesized analogs to increase the intracellular Ca2+ levels. In summary, this study presents compound 3dc as a proposed new tool for the adjuvant chemotherapy of glioblastoma.
Collapse
Affiliation(s)
- Seokmin Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Jung Moo Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Borami Jeon
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ahmed Elkamhawy
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Sora Paik
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jinpyo Hong
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Soo-Jin Oh
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sun Ha Paek
- Department of Neurosurgery, College of Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - C Justin Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea; Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ahmed H E Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Sang Soo Kang
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju, 52727, Republic of Korea.
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
185
|
Demner-Fushman D, Shooshan SE, Rodriguez L, Aronson AR, Lang F, Rogers W, Roberts K, Tonning J. A dataset of 200 structured product labels annotated for adverse drug reactions. Sci Data 2018; 5:180001. [PMID: 29381145 PMCID: PMC5789866 DOI: 10.1038/sdata.2018.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 12/13/2017] [Indexed: 11/09/2022] Open
Abstract
Adverse drug reactions (ADRs), unintended and sometimes dangerous effects that a drug may have, are one of the leading causes of morbidity and mortality during medical care. To date, there is no structured machine-readable authoritative source of known ADRs. The United States Food and Drug Administration (FDA) partnered with the National Library of Medicine to create a pilot dataset containing standardised information about known adverse reactions for 200 FDA-approved drugs. The Structured Product Labels (SPLs), the documents FDA uses to exchange information about drugs and other products, were manually annotated for adverse reactions at the mention level to facilitate development and evaluation of text mining tools for extraction of ADRs from all SPLs. The ADRs were then normalised to the Unified Medical Language System (UMLS) and to the Medical Dictionary for Regulatory Activities (MedDRA). We present the curation process and the structure of the publicly available database SPL-ADR-200db containing 5,098 distinct ADRs. The database is available at https://bionlp.nlm.nih.gov/tac2017adversereactions/; the code for preparing and validating the data is available at https://github.com/lhncbc/fda-ars.
Collapse
Affiliation(s)
- Dina Demner-Fushman
- U.S. National Library of Medicine, NIH, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Sonya E Shooshan
- U.S. National Library of Medicine, NIH, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Laritza Rodriguez
- U.S. National Library of Medicine, NIH, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Alan R Aronson
- U.S. National Library of Medicine, NIH, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Francois Lang
- U.S. National Library of Medicine, NIH, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Willie Rogers
- U.S. National Library of Medicine, NIH, 8600 Rockville Pike, Bethesda, MD 20894, USA
| | - Kirk Roberts
- UT Health School of Biomedical Informatics, 7000 Fannin St., Houston, TX 77030, USA
| | - Joseph Tonning
- Office of New Drugs, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, 10001 New Hampshire Ave, Silver Spring, MD 20903, USA
| |
Collapse
|
186
|
Ravikumar B, Aittokallio T. Improving the efficacy-safety balance of polypharmacology in multi-target drug discovery. Expert Opin Drug Discov 2017; 13:179-192. [DOI: 10.1080/17460441.2018.1413089] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Balaguru Ravikumar
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Turku, Turku, Finland
| |
Collapse
|
187
|
Sam E, Athri P. Web-based drug repurposing tools: a survey. Brief Bioinform 2017; 20:299-316. [DOI: 10.1093/bib/bbx125] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Elizabeth Sam
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| | - Prashanth Athri
- Department of Computer Science & Engineering Amrita, University Bengaluru, India
| |
Collapse
|
188
|
Turanli B, Gulfidan G, Arga KY. Transcriptomic-Guided Drug Repositioning Supported by a New Bioinformatics Search Tool: geneXpharma. ACTA ACUST UNITED AC 2017; 21:584-591. [DOI: 10.1089/omi.2017.0127] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Beste Turanli
- Department of Bioengineering, Marmara University, Istanbul, Turkey
- Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Gizem Gulfidan
- Department of Bioengineering, Marmara University, Istanbul, Turkey
| | | |
Collapse
|
189
|
Himmelstein DS, Lizee A, Hessler C, Brueggeman L, Chen SL, Hadley D, Green A, Khankhanian P, Baranzini SE. Systematic integration of biomedical knowledge prioritizes drugs for repurposing. eLife 2017; 6:26726. [PMID: 28936969 PMCID: PMC5640425 DOI: 10.7554/elife.26726] [Citation(s) in RCA: 276] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 09/11/2017] [Indexed: 12/16/2022] Open
Abstract
The ability to computationally predict whether a compound treats a disease would improve the economy and success rate of drug approval. This study describes Project Rephetio to systematically model drug efficacy based on 755 existing treatments. First, we constructed Hetionet (neo4j.het.io), an integrative network encoding knowledge from millions of biomedical studies. Hetionet v1.0 consists of 47,031 nodes of 11 types and 2,250,197 relationships of 24 types. Data were integrated from 29 public resources to connect compounds, diseases, genes, anatomies, pathways, biological processes, molecular functions, cellular components, pharmacologic classes, side effects, and symptoms. Next, we identified network patterns that distinguish treatments from non-treatments. Then, we predicted the probability of treatment for 209,168 compound-disease pairs (het.io/repurpose). Our predictions validated on two external sets of treatment and provided pharmacological insights on epilepsy, suggesting they will help prioritize drug repurposing candidates. This study was entirely open and received realtime feedback from 40 community members.
Collapse
Affiliation(s)
- Daniel Scott Himmelstein
- Biological and Medical Informatics Program, University of California, San Francisco, San Francisco, United States.,Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, United States
| | - Antoine Lizee
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,ITUN-CRTI-UMR 1064 Inserm, University of Nantes, Nantes, France
| | - Christine Hessler
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Leo Brueggeman
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,University of Iowa, Iowa City, United States
| | - Sabrina L Chen
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Johns Hopkins University, Baltimore, United States
| | - Dexter Hadley
- Department of Pediatrics, University of California, San Fransisco, San Fransisco, United States.,Institute for Computational Health Sciences, University of California, San Francisco, San Francisco, United States
| | - Ari Green
- Department of Neurology, University of California, San Francisco, San Francisco, United States
| | - Pouya Khankhanian
- Department of Neurology, University of California, San Francisco, San Francisco, United States.,Center for Neuroengineering and Therapeutics, University of Pennsylvania, Philadelphia, United States
| | - Sergio E Baranzini
- Biological and Medical Informatics Program, University of California, San Francisco, San Francisco, United States.,Department of Neurology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
190
|
March-Vila E, Pinzi L, Sturm N, Tinivella A, Engkvist O, Chen H, Rastelli G. On the Integration of In Silico Drug Design Methods for Drug Repurposing. Front Pharmacol 2017; 8:298. [PMID: 28588497 PMCID: PMC5440551 DOI: 10.3389/fphar.2017.00298] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/10/2017] [Indexed: 11/13/2022] Open
Abstract
Drug repurposing has become an important branch of drug discovery. Several computational approaches that help to uncover new repurposing opportunities and aid the discovery process have been put forward, or adapted from previous applications. A number of successful examples are now available. Overall, future developments will greatly benefit from integration of different methods, approaches and disciplines. Steps forward in this direction are expected to help to clarify, and therefore to rationally predict, new drug–target, target–disease, and ultimately drug–disease associations.
Collapse
Affiliation(s)
- Eric March-Vila
- Molecular Modelling & Drug Design Lab, Department of Life Sciences, University of Modena and Reggio EmiliaModena, Italy
| | - Luca Pinzi
- Molecular Modelling & Drug Design Lab, Department of Life Sciences, University of Modena and Reggio EmiliaModena, Italy
| | - Noé Sturm
- Molecular Modelling & Drug Design Lab, Department of Life Sciences, University of Modena and Reggio EmiliaModena, Italy
| | - Annachiara Tinivella
- Molecular Modelling & Drug Design Lab, Department of Life Sciences, University of Modena and Reggio EmiliaModena, Italy
| | - Ola Engkvist
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D GothenburgMölndal, Sweden
| | - Hongming Chen
- Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D GothenburgMölndal, Sweden
| | - Giulio Rastelli
- Molecular Modelling & Drug Design Lab, Department of Life Sciences, University of Modena and Reggio EmiliaModena, Italy
| |
Collapse
|
191
|
González-Medina M, Naveja JJ, Sánchez-Cruz N, Medina-Franco JL. Open chemoinformatic resources to explore the structure, properties and chemical space of molecules. RSC Adv 2017. [DOI: 10.1039/c7ra11831g] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Open chemoinformatic servers facilitate analysis of chemical space and structure–activity relationships.
Collapse
Affiliation(s)
- Mariana González-Medina
- Department of Pharmacy
- School of Chemistry
- Universidad Nacional Autónoma de México
- Mexico City 04510
- Mexico
| | - J. Jesús Naveja
- Department of Pharmacy
- School of Chemistry
- Universidad Nacional Autónoma de México
- Mexico City 04510
- Mexico
| | - Norberto Sánchez-Cruz
- Department of Pharmacy
- School of Chemistry
- Universidad Nacional Autónoma de México
- Mexico City 04510
- Mexico
| | - José L. Medina-Franco
- Department of Pharmacy
- School of Chemistry
- Universidad Nacional Autónoma de México
- Mexico City 04510
- Mexico
| |
Collapse
|