Pirfenidone ameliorates murine chronic GVHD through inhibition of macrophage infiltration and TGF-β production

J Du, K Paz, R Flynn, A Vulic… - Blood, The Journal …, 2017 - ashpublications.org
J Du, K Paz, R Flynn, A Vulic, TM Robinson, KE Lineburg, KA Alexander, J Meng, S Roy
Blood, The Journal of the American Society of Hematology, 2017ashpublications.org
Allogeneic hematopoietic stem cell transplantation is hampered by chronic graft-versus-host
disease (cGVHD), resulting in multiorgan fibrosis and diminished function. Fibrosis in lung
and skin leads to progressive bronchiolitis obliterans (BO) and scleroderma, respectively, for
which new treatments are needed. We evaluated pirfenidone, a Food and Drug
Administration (FDA)-approved drug for idiopathic pulmonary fibrosis, for its therapeutic
effect in cGVHD mouse models with distinct pathophysiology. In a full major …
Abstract
Allogeneic hematopoietic stem cell transplantation is hampered by chronic graft-versus-host disease (cGVHD), resulting in multiorgan fibrosis and diminished function. Fibrosis in lung and skin leads to progressive bronchiolitis obliterans (BO) and scleroderma, respectively, for which new treatments are needed. We evaluated pirfenidone, a Food and Drug Administration (FDA)-approved drug for idiopathic pulmonary fibrosis, for its therapeutic effect in cGVHD mouse models with distinct pathophysiology. In a full major histocompatibility complex (MHC)-mismatched, multiorgan system model with BO, donor T-cell responses that support pathogenic antibody production are required for cGVHD development. Pirfenidone treatment beginning one month post-transplant restored pulmonary function and reversed lung fibrosis, which was associated with reduced macrophage infiltration and transforming growth factor-β production. Pirfenidone dampened splenic germinal center B-cell and T-follicular helper cell frequencies that collaborate to produce antibody. In both a minor histocompatibility antigen–mismatched as well as a MHC-haploidentical model of sclerodermatous cGVHD, pirfenidone significantly reduced macrophages in the skin, although clinical improvement of scleroderma was only seen in one model. In vitro chemotaxis assays demonstrated that pirfenidone impaired macrophage migration to monocyte chemoattractant protein-1 (MCP-1) as well as IL-17A, which has been linked to cGVHD generation. Taken together, our data suggest that pirfenidone is a potential therapeutic agent to ameliorate fibrosis in cGVHD.
ashpublications.org