Cargando…

Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity

[Image: see text] Diclofenac (DCF) is widely used as a nonsteroidal anti-inflammatory drug; however, it is associated with severe liver injury. This adverse reaction is thought to be related to the reactive quinone imine (QI) and acyl glucuronide (AG) metabolites of DCF, but it remains controversial...

Descripción completa

Detalles Bibliográficos
Autores principales: Tateishi, Yasuhiro, Ohe, Tomoyuki, Ogawa, Mai, Takahashi, Kyoko, Nakamura, Shigeo, Mashino, Tadahiko
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Chemical Society 2020
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7758955/
https://www.ncbi.nlm.nih.gov/pubmed/33376898
http://dx.doi.org/10.1021/acsomega.0c04942
_version_ 1783627032704319488
author Tateishi, Yasuhiro
Ohe, Tomoyuki
Ogawa, Mai
Takahashi, Kyoko
Nakamura, Shigeo
Mashino, Tadahiko
author_facet Tateishi, Yasuhiro
Ohe, Tomoyuki
Ogawa, Mai
Takahashi, Kyoko
Nakamura, Shigeo
Mashino, Tadahiko
author_sort Tateishi, Yasuhiro
collection PubMed
description [Image: see text] Diclofenac (DCF) is widely used as a nonsteroidal anti-inflammatory drug; however, it is associated with severe liver injury. This adverse reaction is thought to be related to the reactive quinone imine (QI) and acyl glucuronide (AG) metabolites of DCF, but it remains controversial which reactive metabolites mainly contribute to DCF-induced toxicity. In this study, we synthesized five types of DCF analogs that were designed to mitigate the formation of reactive QI and/or AG metabolites and evaluated their metabolic stability, cyclooxygenase (COX) inhibitory activity, and toxicity to cryopreserved human hepatocytes. Compounds with fluorine at the 5- and 4′-positions of aromatic rings exhibited modest and high metabolic stability to oxidation by cytochrome P450, respectively, but induced cytotoxicity comparable to DCF. Replacing the carboxylic group of DCF with its bioisosteres was effective in terms of stability to oxidative metabolism and glucuronidation; however, sulfonic acid and sulfonamide groups were not preferable for COX inhibition, and tetrazole-containing analogs induced strong cytotoxicity. On the other hand, compounds that have fluorine at the benzylic position were resistant to glucuronidation and showed little toxicity to hepatocytes. In addition, among these compounds, those with hydrogen at the 4′-position (2a and 2c) selectively inhibited the COX-2 enzyme. Throughout these data, it was suggested that compounds 2a and 2c might be novel safer and more efficacious drug candidates instead of DCF.
format Online
Article
Text
id pubmed-7758955
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher American Chemical Society
record_format MEDLINE/PubMed
spelling pubmed-77589552020-12-28 Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity Tateishi, Yasuhiro Ohe, Tomoyuki Ogawa, Mai Takahashi, Kyoko Nakamura, Shigeo Mashino, Tadahiko ACS Omega [Image: see text] Diclofenac (DCF) is widely used as a nonsteroidal anti-inflammatory drug; however, it is associated with severe liver injury. This adverse reaction is thought to be related to the reactive quinone imine (QI) and acyl glucuronide (AG) metabolites of DCF, but it remains controversial which reactive metabolites mainly contribute to DCF-induced toxicity. In this study, we synthesized five types of DCF analogs that were designed to mitigate the formation of reactive QI and/or AG metabolites and evaluated their metabolic stability, cyclooxygenase (COX) inhibitory activity, and toxicity to cryopreserved human hepatocytes. Compounds with fluorine at the 5- and 4′-positions of aromatic rings exhibited modest and high metabolic stability to oxidation by cytochrome P450, respectively, but induced cytotoxicity comparable to DCF. Replacing the carboxylic group of DCF with its bioisosteres was effective in terms of stability to oxidative metabolism and glucuronidation; however, sulfonic acid and sulfonamide groups were not preferable for COX inhibition, and tetrazole-containing analogs induced strong cytotoxicity. On the other hand, compounds that have fluorine at the benzylic position were resistant to glucuronidation and showed little toxicity to hepatocytes. In addition, among these compounds, those with hydrogen at the 4′-position (2a and 2c) selectively inhibited the COX-2 enzyme. Throughout these data, it was suggested that compounds 2a and 2c might be novel safer and more efficacious drug candidates instead of DCF. American Chemical Society 2020-12-08 /pmc/articles/PMC7758955/ /pubmed/33376898 http://dx.doi.org/10.1021/acsomega.0c04942 Text en © 2020 American Chemical Society This is an open access article published under a Creative Commons Non-Commercial No Derivative Works (CC-BY-NC-ND) Attribution License (http://pubs.acs.org/page/policy/authorchoice_ccbyncnd_termsofuse.html) , which permits copying and redistribution of the article, and creation of adaptations, all for non-commercial purposes.
spellingShingle Tateishi, Yasuhiro
Ohe, Tomoyuki
Ogawa, Mai
Takahashi, Kyoko
Nakamura, Shigeo
Mashino, Tadahiko
Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity
title Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity
title_full Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity
title_fullStr Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity
title_full_unstemmed Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity
title_short Development of Novel Diclofenac Analogs Designed to Avoid Metabolic Activation and Hepatocyte Toxicity
title_sort development of novel diclofenac analogs designed to avoid metabolic activation and hepatocyte toxicity
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7758955/
https://www.ncbi.nlm.nih.gov/pubmed/33376898
http://dx.doi.org/10.1021/acsomega.0c04942
work_keys_str_mv AT tateishiyasuhiro developmentofnoveldiclofenacanalogsdesignedtoavoidmetabolicactivationandhepatocytetoxicity
AT ohetomoyuki developmentofnoveldiclofenacanalogsdesignedtoavoidmetabolicactivationandhepatocytetoxicity
AT ogawamai developmentofnoveldiclofenacanalogsdesignedtoavoidmetabolicactivationandhepatocytetoxicity
AT takahashikyoko developmentofnoveldiclofenacanalogsdesignedtoavoidmetabolicactivationandhepatocytetoxicity
AT nakamurashigeo developmentofnoveldiclofenacanalogsdesignedtoavoidmetabolicactivationandhepatocytetoxicity
AT mashinotadahiko developmentofnoveldiclofenacanalogsdesignedtoavoidmetabolicactivationandhepatocytetoxicity