Saracatinib, a Selective Src Kinase Inhibitor, Blocks Fibrotic Responses in Preclinical Models of Pulmonary Fibrosis

Farida Ahangari, Christine Becker, Daniel G. Foster, Maurizio Chioccioli, Meghan Nelson, Keriann Beke, Xing Wang, Aurelien Justet, Taylor Adams, Benjamin Readhead, Carly Meador, Kelly Correll, Loukia N. Lili, Helen M. Roybal, Kadi Ann Rose, Shuizi Ding, Thomas Barnthaler, Natalie Briones, Giuseppe DeIuliis, Jonas C. SchuppQin Li, Norihito Omote, Yael Aschner, Lokesh Sharma, Katrina W. Kopf, Björn Magnusson, Ryan Hicks, Anna Backmark, Charles S. Dela Cruz, Ivan Rosas, Leslie P. Cousens, Joel T. Dudley, Naftali Kaminski, Gregory P. Downey

Research output: Contribution to journalArticlepeer-review

23 Scopus citations

Abstract

Rationale: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and often fatal disorder. Two U.S. Food and Drug Administration-approved antifibrotic drugs, nintedanib and pirfenidone, slow the rate of decline in lung function, but responses are variable and side effects are common. Objectives: Using an in silico data-driven approach, we identified a robust connection between the transcriptomic perturbations in IPF disease and those induced by saracatinib, a selective Src kinase inhibitor originally developed for oncological indications. Based on these observations, we hypothesized that saracatinib would be effective at attenuating pulmonary fibrosis. Methods: We investigated the antifibrotic efficacy of saracatinib relative to nintedanib and pirfenidone in three preclinical models: 1) in vitro in normal human lung fibroblasts; 2) in vivo in bleomycin and recombinant Ad-TGF-β (adenovirus transforming growth factor-β) murine models of pulmonary fibrosis; and 3) ex vivo in mice and human precision-cut lung slices from these two murine models as well as patients with IPF and healthy donors. Measurements and Main Results: In each model, the effectiveness of saracatinib in blocking fibrogenic responses was equal or superior to nintedanib and pirfenidone. Transcriptomic analyses of TGF-β-stimulated normal human lung fibroblasts identified specific gene sets associated with fibrosis, including epithelial-mesenchymal transition, TGF-β, and WNT signaling that was uniquely altered by saracatinib. Transcriptomic analysis of whole-lung extracts from the two animal models of pulmonary fibrosis revealed that saracatinib reverted many fibrogenic pathways, including epithelial-mesenchymal transition, immune responses, and extracellular matrix organization. Amelioration of fibrosis and inflammatory cascades in human precision-cut lung slices confirmed the potential therapeutic efficacy of saracatinib in human lung fibrosis. Conclusions: These studies identify novel Src-dependent fibrogenic pathways and support the study of the therapeutic effectiveness of saracatinib in IPF treatment.

Original languageEnglish (US)
Pages (from-to)1463-1479
Number of pages17
JournalAmerican Journal of Respiratory and Critical Care Medicine
Volume206
Issue number12
DOIs
StatePublished - Dec 15 2022
Externally publishedYes

Keywords

  • Src family kinase
  • idiopathic pulmonary fibrosis
  • lung fibrosis
  • preclinical models
  • tyrosine kinase

ASJC Scopus subject areas

  • Pulmonary and Respiratory Medicine
  • Critical Care and Intensive Care Medicine

Fingerprint

Dive into the research topics of 'Saracatinib, a Selective Src Kinase Inhibitor, Blocks Fibrotic Responses in Preclinical Models of Pulmonary Fibrosis'. Together they form a unique fingerprint.

Cite this