Please use this identifier to cite or link to this item: https://repository.monashhealth.org/monashhealthjspui/handle/1/29264
Title: Pirfenidone: Molecular mechanisms and potential clinical applications in lung disease.
Authors: Ruwanpura S.M.;Bardin P.G.;Thomas B.J.
Monash Health Department(s): Respiratory and Sleep Medicine
Institution: (Ruwanpura, Thomas, Bardin) Monash Lung and Sleep, Monash Health, Monash Medical Centre, 246 Clayton Road, Clayton, VIC 3168, Australia (Thomas, Bardin) Hudson Institute of Medical Research, Clayton, VIC, Australia
Issue Date: 30-Jun-2020
Copyright year: 2020
Publisher: American Thoracic Society (E-mail: malexander@thoracic.org)
Place of publication: United States
Publication information: American Journal of Respiratory Cell and Molecular Biology. 62 (4) (pp 413-422), 2020. Date of Publication: 2020.
Journal: American Journal of Respiratory Cell and Molecular Biology
Abstract: Pirfenidone (PFD) is a pharmacological compound with therapeutic efficacy in idiopathic pulmonary fibrosis. It has been chiefly characterized as an antifibrotic agent, although it was initially developed as an antiinflammatory compound because of its ability to diminish the accumulation of inflammatory cells and cytokines. Despite recent studies that have elucidated key mechanisms, the precise molecular activities of PFD remain incompletely understood. PFD modulates fibrogenic growth factors, thereby attenuating fibroblast proliferation, myofibroblast differentiation, collagen and fibronectin synthesis, and deposition of extracellular matrix. This effect is mediated by suppression of TGF-b1 (transforming growth factor-b1) and other growth factors. Here, we appraise the impact of PFD on TGF-b1 production and its downstream pathways. Accumulating evidence indicates that PFD also downregulates inflammatory pathways and therefore has considerable potential as a viable and innovative antiinflammatory compound. We examine the effects of PFD on inflammatory cells and the production of pro- and antiinflammatory cytokines in the lung. In this context, recent evidence that PFD can target inflammasome pathways and ensuing lung inflammation is highlighted. Finally, the antioxidant properties of PFD, such as its ability to inhibit redox reactions and regulate oxidative stress-related genes and enzymes, are detailed. In summary, this narrative review examines molecular mechanisms underpinning PFD and its recognized benefits in lung fibrosis. We highlight preclinical data that demonstrate the potential of PFD as a nonsteroidal antiinflammatory agent and outline areas for future research.Copyright © 2020 by the American Thoracic Society.
DOI: http://monash.idm.oclc.org/login?url=http://dx.doi.org/10.1165/rcmb.2019-0328TR
PubMed URL: 31967851 [http://www.ncbi.nlm.nih.gov/pubmed/?term=31967851]
ISSN: 1044-1549
URI: https://repository.monashhealth.org/monashhealthjspui/handle/1/29264
Type: Review
Subjects: antiinflammatory activity
drug effect
human
inflammatory cell
lung disease/dt [Drug Therapy]
*lung fibrosis/dt [Drug Therapy]
antifibrotic activity
*pirfenidone/pd [Pharmacology]
transforming growth factor beta1/ec [Endogenous Compound]
nonhuman
pharmacology
review
cytokine/ec [Endogenous Compound]
inflammasome/ec [Endogenous Compound]
*pirfenidone/dt [Drug Therapy]
antiinflammatory activity
drug effect
inflammatory cell
lung disease
lung fibrosis
antifibrotic activity
pirfenidone
transforming growth factor beta1
pharmacology
cytokine
inflammasome
pirfenidone
Review
*lung fibrosis / *drug therapy
lung disease / drug therapy
inflammatory cell
nonhuman
drug effect
human
pharmacology
antiinflammatory activity
antifibrotic activity
Type of Clinical Study or Trial: Review article (e.g. literature review, narrative review)
Appears in Collections:Articles

Show full item record

Page view(s)

28
checked on Aug 17, 2024

Google ScholarTM

Check


Items in Monash Health Research Repository are protected by copyright, with all rights reserved, unless otherwise indicated.