Cargando…

Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release

[Image: see text] Histone deacetylase inhibitors (HDACi) target abnormal epigenetic states associated with a variety of pathologies, including cancer. Here, the development of a prodrug of the canonical broad-spectrum HDACi suberoylanilide hydroxamic acid (SAHA) is described. Although hydroxamic aci...

Descripción completa

Detalles Bibliográficos
Autores principales: Daniel, Kevin B., Sullivan, Eric D., Chen, Yao, Chan, Joshua C., Jennings, Patricia A., Fierke, Carol A., Cohen, Seth M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Chemical Society 2015
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4467547/
https://www.ncbi.nlm.nih.gov/pubmed/25974739
http://dx.doi.org/10.1021/acs.jmedchem.5b00539
_version_ 1782376378596851712
author Daniel, Kevin B.
Sullivan, Eric D.
Chen, Yao
Chan, Joshua C.
Jennings, Patricia A.
Fierke, Carol A.
Cohen, Seth M.
author_facet Daniel, Kevin B.
Sullivan, Eric D.
Chen, Yao
Chan, Joshua C.
Jennings, Patricia A.
Fierke, Carol A.
Cohen, Seth M.
author_sort Daniel, Kevin B.
collection PubMed
description [Image: see text] Histone deacetylase inhibitors (HDACi) target abnormal epigenetic states associated with a variety of pathologies, including cancer. Here, the development of a prodrug of the canonical broad-spectrum HDACi suberoylanilide hydroxamic acid (SAHA) is described. Although hydroxamic acids are utilized universally in the development of metalloenzyme inhibitors, they are considered to be poor pharmacophores with reduced activity in vivo. We developed a prodrug of SAHA by appending a promoiety, sensitive to thiols, to the hydroxamic acid warhead (termed SAHA-TAP). After incubation of SAHA-TAP with an HDAC, the thiol of a conserved HDAC cysteine residue becomes covalently tagged with the promoiety, initiating a cascade reaction that leads to the release of SAHA. Mass spectrometry and enzyme kinetics experiments validate that the cysteine residue is covalently appended with the TAP promoiety. SAHA-TAP demonstrates cytotoxicity activity against various cancer cell lines. This strategy represents an original prodrug design with a dual mode of action for HDAC inhibition.
format Online
Article
Text
id pubmed-4467547
institution National Center for Biotechnology Information
language English
publishDate 2015
publisher American Chemical Society
record_format MEDLINE/PubMed
spelling pubmed-44675472016-05-14 Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release Daniel, Kevin B. Sullivan, Eric D. Chen, Yao Chan, Joshua C. Jennings, Patricia A. Fierke, Carol A. Cohen, Seth M. J Med Chem [Image: see text] Histone deacetylase inhibitors (HDACi) target abnormal epigenetic states associated with a variety of pathologies, including cancer. Here, the development of a prodrug of the canonical broad-spectrum HDACi suberoylanilide hydroxamic acid (SAHA) is described. Although hydroxamic acids are utilized universally in the development of metalloenzyme inhibitors, they are considered to be poor pharmacophores with reduced activity in vivo. We developed a prodrug of SAHA by appending a promoiety, sensitive to thiols, to the hydroxamic acid warhead (termed SAHA-TAP). After incubation of SAHA-TAP with an HDAC, the thiol of a conserved HDAC cysteine residue becomes covalently tagged with the promoiety, initiating a cascade reaction that leads to the release of SAHA. Mass spectrometry and enzyme kinetics experiments validate that the cysteine residue is covalently appended with the TAP promoiety. SAHA-TAP demonstrates cytotoxicity activity against various cancer cell lines. This strategy represents an original prodrug design with a dual mode of action for HDAC inhibition. American Chemical Society 2015-05-14 2015-06-11 /pmc/articles/PMC4467547/ /pubmed/25974739 http://dx.doi.org/10.1021/acs.jmedchem.5b00539 Text en Copyright © 2015 American Chemical Society This is an open access article published under an ACS AuthorChoice License (http://pubs.acs.org/page/policy/authorchoice_termsofuse.html) , which permits copying and redistribution of the article or any adaptations for non-commercial purposes.
spellingShingle Daniel, Kevin B.
Sullivan, Eric D.
Chen, Yao
Chan, Joshua C.
Jennings, Patricia A.
Fierke, Carol A.
Cohen, Seth M.
Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release
title Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release
title_full Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release
title_fullStr Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release
title_full_unstemmed Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release
title_short Dual-Mode HDAC Prodrug for Covalent Modification and Subsequent Inhibitor Release
title_sort dual-mode hdac prodrug for covalent modification and subsequent inhibitor release
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC4467547/
https://www.ncbi.nlm.nih.gov/pubmed/25974739
http://dx.doi.org/10.1021/acs.jmedchem.5b00539
work_keys_str_mv AT danielkevinb dualmodehdacprodrugforcovalentmodificationandsubsequentinhibitorrelease
AT sullivanericd dualmodehdacprodrugforcovalentmodificationandsubsequentinhibitorrelease
AT chenyao dualmodehdacprodrugforcovalentmodificationandsubsequentinhibitorrelease
AT chanjoshuac dualmodehdacprodrugforcovalentmodificationandsubsequentinhibitorrelease
AT jenningspatriciaa dualmodehdacprodrugforcovalentmodificationandsubsequentinhibitorrelease
AT fierkecarola dualmodehdacprodrugforcovalentmodificationandsubsequentinhibitorrelease
AT cohensethm dualmodehdacprodrugforcovalentmodificationandsubsequentinhibitorrelease