Please use this identifier to cite or link to this item: http://cmuir.cmu.ac.th/jspui/handle/6653943832/57757
Full metadata record
DC FieldValueLanguage
dc.contributor.authorParunya Chaiyawaten_US
dc.contributor.authorJongkolnee Settakornen_US
dc.contributor.authorApiruk Sangsinen_US
dc.contributor.authorPimpisa Teeyakasemen_US
dc.contributor.authorJeerawan Klangjorhoren_US
dc.contributor.authorAungsumalee Soongkhawen_US
dc.contributor.authorDumnoensun Pruksakornen_US
dc.date.accessioned2018-09-05T03:49:17Z-
dc.date.available2018-09-05T03:49:17Z-
dc.date.issued2017-02-01en_US
dc.identifier.issn11786930en_US
dc.identifier.other2-s2.0-85011976859en_US
dc.identifier.other10.2147/OTT.S119993en_US
dc.identifier.urihttps://www.scopus.com/inward/record.uri?partnerID=HzOxMe3b&scp=85011976859&origin=inwarden_US
dc.identifier.urihttp://cmuir.cmu.ac.th/jspui/handle/6653943832/57757-
dc.description.abstract© 2017 Chaiyawat et al. Despite multimodal therapeutic treatments of osteosarcoma (OS), some patients develop resistance to currently available regimens and eventually end up with recurrent or metastatic outcomes. Many attempts have been made to discover effective drugs for improving outcome; however, due to the heterogeneity of the disease, new therapeutic options have not yet been identified. This study aims to explore potential targeted therapy related to protein profiles of OS. In this review of proteomics studies, we extracted data on differentially expressed proteins (DEPs) from archived literature in PubMed and our in-house repository. The data were divided into three experimental groups, DEPs in 1) OS/OB: OS vs osteoblastic (OB) cells, 2) metastasis: metastatic vs non-metastatic sublines plus fresh tissues from primary OS with and without pulmonary metastasis, and 3) chemoresistance: spheroid (higher chemoresistance) vs monolayer cells plus fresh tissues from biopsies from good and poor responders. All up-regulated protein entities in the list of DEPs were sorted and cross-referenced with identifiers of targets of US Food and Drug Administration (FDA)-approved agents and chemical inhibitors. We found that many targets of FDA-approved antineoplastic agents, mainly a group of epigenetic regulators, kinases, and proteasomes, were highly expressed in OS cells. Additionally, some overexpressed proteins were targets of FDA-approved non-cancer drugs, including immunosuppressive and antiarrhythmic drugs. The resulting list of chemical agents showed that some transferase enzyme inhibitors might have anticancer activity. We also explored common targets of OS/OB and metastasis groups, including amidophosphoribosyltransferase (PPAT), l-lactate dehydrogenase B chain (LDHB), and pyruvate kinase M2 (PKM2) as well as the common target of all categories, cathepsin D (CTSD). This study demonstrates the benefits of a text mining approach to exploring therapeutic targets related to protein expression patterns. These results suggest possible repurposing of some FDA-approved medicines for the treatment of OS and using chemical inhibitors in drug screening tests.en_US
dc.subjectMedicineen_US
dc.titleExploring targeted therapy of osteosarcoma using proteomics dataen_US
dc.typeJournalen_US
article.title.sourcetitleOncoTargets and Therapyen_US
article.volume10en_US
article.stream.affiliationsChiang Mai Universityen_US
Appears in Collections:CMUL: Journal Articles

Files in This Item:
There are no files associated with this item.


Items in CMUIR are protected by copyright, with all rights reserved, unless otherwise indicated.