Role and New Insights of Pirfenidone in Fibrotic Diseases

David Alejandro Lopez-de la Mora, Cibeles Sanchez-Roque, Margarita Montoya-Buelna, Sergio Sanchez-Enriquez, Silvia Lucano-Landeros, Jose Macias-Barragan, Juan Armendariz-Borunda, David Alejandro Lopez-de la Mora, Cibeles Sanchez-Roque, Margarita Montoya-Buelna, Sergio Sanchez-Enriquez, Silvia Lucano-Landeros, Jose Macias-Barragan, Juan Armendariz-Borunda

Abstract

Pirfenidone (PFD) is a non-peptide synthetic molecule issued as a broad-spectrum anti-fibrotic drug with the ability to decrease TGF-β1, TNF-α, PDGF and COL1A1 expression, which is highly related to prevent or remove excessive deposition of scar tissue in several organs. Basic and clinical evidence suggests that PFD may safely slow or inhibit the progressive fibrosis swelling after tissue injuries. Furthermore, a number of evidence suggests that this molecule will have positive effects in the treatment of other inflammatory diseases. This review contains current research in which PFD has been used as the treatment of several diseases, and focus mainly in the outcomes related to improve inflammation and fibrogenesis. Therefore, the main goal of this review is to focus on the novel findings of PFD efficacy rather than deepen in the chemical aspects of the molecule.

Keywords: Pirfenidone; fibrosis; idiopathic pulmonary fibrosis; inflammation.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interest exists.

Figures

Figure 1
Figure 1
Pirfenidone targets. Different targets in vivo and in vitro for PFD have been described, the most prominent being inhibition of TGF-β1 and TNF-α. However, it has also been shown that PFD has either direct or indirect action on other molecules such as collagen I, PDGF, IL-6, IL-1β, IL-13, IL-12p40, fibronectin, HSP47 and ICAM-1.

References

    1. Huang NY, Ding L, Wang J, Zhang QY, Liu X, Lin HD, Hua WY. Pharmacokinetics, safety and tolerability of pirfenidone and its major metabolite after single and multiple oral doses in healthy Chinese subjects under fed conditions. Drug Res. 2013;63:388–95.
    1. Macias-Barragan J, Sandoval-Rodríguez AS, Navarro-Partida J, Armendariz-Borunda J. The multifaceted role of pirfenidone and its novel targets. Fibrogenesis Tissue Repair. 2010;3:16.
    1. Paz Z, Shoenfeld Y; Antifibrosis. To reverse the irreversible. Clinic Rev Allerg Immunol. 2010;38:276–86.
    1. Zamara E, Novo E, Parola M. Oxidative stress and liver fibrosis: from liver injury to the modulation of cell signaling and response; Ali S, Mann DA, Friedman SL, eds; Liver Diseases: Biochemical Mechanisms and New Therapeutic Insights; Enfield, NH, USA: Science Publishers 2004:93-114.
    1. Salazar-Montes A, Ruiz-Corro L, López-Reyes A, Castrejón-Gómez E, Armendáriz-Borunda J. Potent antioxidant role of pirfenidone in experimental cirrhosis. Eur J Pharmacol. 2008;24:69–77.
    1. Seki E, Brenner DA. Recent advancement of molecular mechanisms of liver fibrosis. J Hepatobiliary Pancreat Sci; 2015.
    1. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028–1040.
    1. Visner GA, Liu F, Bizargity P, Liu H, Liu K, Yang J, Wang L, Hancock WW. Pirfenidone inhibits T cell activation, proliferation, cytokine and chemokine production, and host alloresponses. Transplantation. 2009;88:330–338.
    1. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby TV, Cordier JF, Flaherty KR, Lasky JA, Lynch DA, Ryu JH, Swigris JJ, Wells AU, Ancochea J, Bouros D, Carvalho C, Costabel U, Ebina M, Hansell DM, Johkoh T, Kim DS, King TE Jr, Kondoh Y, Myers J, Müller NL, Nicholson AG, Richeldi L, Selman M, Dudden RF, Griss BS, Protzko SL, Schünemann HJ; ATS/ERS/JRS/ALAT Committee on Idiopathic Pulmonary Fibrosis. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med. 2011;183(6):788–824.
    1. Azuma A, Taguchi Y, Ogura T, Ebina M, Taniguchi H, Kondoh Y, Suga M, Takahashi H, Nakata K, Sato A, Kudoh S, Nukiwa T; Pirfenidone clinical study group in Japan. Exploratory analysis of a phase III trial of pirfenidone identifies a subpopulation of patients with idiopathic pulmonary fibrosis as benefiting from treatment. Respiratory Research. 2011;12:143.
    1. Cottin V. The role of pirfenidone in the treatment of idiopathic pulmonary fibrosis. Respiratory Research. 2013;14(Suppl 1):S5.
    1. Albera C, Ferrero C, Rindone E, Zanotto S, Rizza E. Where do we stand with IPF treatment? Respiratory Research. 2013;14(Suppl 1):S7.
    1. Hisatomi K, Mukae H, Sakamoto N, Ishimatsu Y, Kakugawa T, Hara S, Fujita H, Nakamichi S, Oku H, Urata Y, Kubota H, Nagata K, Kohno S. Pirfenidone inhibits TGF-β1-induced over-expression of collagen type I and heat shock protein 47 in A549 cells. BMC Pulmonary Medicine. 2012;12:24.
    1. Friedman SL, Sheppard D, Duffield JS, Violette S. Therapy for fibrotic diseases: nearing the starting line. Sci Transl Med. 2013;5:167.
    1. Rafii R, Juarez MM, Albertson TE, Chan AL. A review of current and novel therapies for idiopathic pulmonary fibrosis. J Thorac Dis. 2013;5:48–73.
    1. Richeldi L, Yasothan U, Kirkpatrick P. Pirfenidone. Nature Reviews Drug Discovery. 2011;10:489–90.
    1. Castelino FV, Varga J. Interstitial lung disease in connective tissue diseases: evolving concepts of pathogenesis and management. Arthritis Res Ther. 2010;12:213.
    1. King TE Jr, Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, Gorina E, Hopkins PM, Kardatzke D, Lancaster L, Lederer DJ, Nathan SD, Pereira CA, Sahn SA, Sussman R, Swigris JJ, Noble PW; ASCEND Study Group. A phase 3 trial of pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2083–92.
    1. Taniguchi H, Kondoh Y, Ebina M, Azuma A, Ogura T, Taguchi Y, Suga M, Takahashi H, Nakata K, Sato A, Sugiyama Y, Kudoh S, Nukiwa T; Pirfenidone Clinical Study Group in Japan. The clinical significance of 5% change in vital capacity in patients with idiopathic pulmonary fibrosis: extended analysis of the pirfenidone trial. Respir Res. 2011;12:93.
    1. Loveman E, Copley VR, Colquitt J, Scott DA, Clegg A, Jones J, O'Reilly KM, Singh S, Bausewein C, Wells A. The clinical effectiveness and cost-effectiveness of treatments for idiopathic pulmonary fibrosis: a systematic review and economic evaluation. Health Technol Assess. 2015;19(20):1–336.
    1. 21. Kometani T, Okamoto T, Yoshida S, Yoshino I. Acute respiratory distress syndrome after pulmonary resection. Gen Thorac Cardiovasc Surg. 2013;61:504–12.
    1. Jiang C, Huang H, Liu J, Wang Y, Lu Z, Xu Z. Adverse events of pirfenidone for the treatment of pulmonary fibrosis: a meta-analysis of randomized controlled trials. PLoS One. 2012;7:47024.
    1. Onoue S, Seto Y, Kato M, Aoki Y, Kojo Y, Yamada S. Inhalable powder formulation of pirfenidone with reduced phototoxic risk for treatment of pulmonary fibrosis. Pharm Res. 2013;30:1586–1596.
    1. Couluris M. Treatment of idiopathic pulmonary fibrosis with losartan: a pilot project. Lung. 2012;190:523–527.
    1. O'Brien K, Troendle J, Gochuico BR, Markello TC, Salas J, Cardona H, Yao J, Bernardini I, Hess R, Gahl WA. Pirfenidone for the treatment of Hermansky-Pudlak Syndrome pulmonary fibrosis. Mol Genet Metab. 2011;103:128–134.
    1. Thielen N, Huizing M, Krabbe JG, White JG, Jansen TJ, Merle PA, Gahl WA, Zweegman S. Hermansky-Pudlak syndrome: the importance of molecular subtyping. J Thromb Haemost. 2010;8:1643–1645.
    1. Zamani N. Pirfenidone; can it be a new horizon for the treatment of pulmonary fibrosis in mustard gas-intoxicated patients? DARU Journal of Pharmaceutical Sciences. 2013;21:13.
    1. Gan Y, Herzog EL, Gomer RH. Pirfenidone treatment of idiopathic pulmonary fibrosis. Therapeutics and Clinical Risk Management 2011. 2014;7:39–47.
    1. Richeldi L, du Bois RM, Raghu G, Azuma A, Brown KK, Costabel U, Cottin V, Flaherty KR, Hansell DM, Inoue Y, Kim DS, Kolb M, Nicholson AG, Noble PW, Selman M, Taniguchi H, Brun M, Le Maulf F, Girard M, Stowasser S, Schlenker-Herceg R, Disse B, Collard HR; INPULSIS Trial Investigators. Efficacy and safety of nintedanib in idiopathic pulmonary fibrosis. N Engl J Med. 2014;370(22):2071–82.
    1. : Silver Spring (MD). FDA News Release: FDA approves Esbriet to treat idiopathic pulmonary fibrosis. Revised 24 Mar 2015. .
    1. Cho ME, Kopp JB. Pirfenidone: an anti-fibrotic and cytoprotective agent as therapy for progressive kidney disease. Expert Opin Investig Drugs. 2010;19:275–283.
    1. Tanios BY, Ziyadeh FN. Emerging therapies for diabetic nephropathy patients: beyond blockade of the renin-angiotensin system. Nephron Extra. 2012;2:278–282.
    1. Sharma K, Ix JH, Mathew AV, Cho M, Pflueger A, Dunn SR. Pirfenidone for diabetic nephropathy. Am Soc Nephrol. 2011;22:1144–1151.
    1. Anil Karihaloo. Anti-fibrosis therapy and diabetic nephropathy. Curr Diab Rep. 2012;12:414–422.
    1. Shi Q, Liu X, Bai Y, Cui C, Li J, Li Y, Hu S, Wei Y. In vitro effects of pirfenidone on cardiac fibroblasts: proliferation, myofibroblast differentiation, migration and cytokine secretion. PLoS One. 2011;6:28134.
    1. Tamargo J, López-Sendón J. Novel therapeutic targets for the treatment of heart failure. Nature Reviews Drug Discovery. 2011;10:536–555.
    1. Wang Y, Hou X, Li Y. Association between transforming growth factor β1 polymorphisms and atrial fibrillation in essential hypertensive subjects. Journal of Biomedical Science. 2010;17:23.
    1. Yamazaki T, Yamashita N, Izumi Y, Nakamura Y, Shiota M, Hanatani A, Shimada K, Muro T, Iwao H, Yoshiyama M. The antifibrotic agent pirfenidone inhibits angiotensin II-induced cardiac hypertrophy in mice. Hypertens Res. 2012;35:34–40.
    1. . Pirfenidone to Treat Hypertrophic Cardiomyopathy. .
    1. . Heart Function and Exercise Capacity in Patients With Hypertrophic Cardiomyopathy. .
    1. Zhong H, Sun G, Lin X, Wu K, Yu M. Evaluation of pirfenidone as a new postoperative antiscarring agent in experimental glaucoma surgery. Invest Ophthalmol Vis Sci. 2011;52:3136–42.
    1. Sun G, Lin X, Zhong H, Yang Y, Qiu X, Ye C, Wu K, Yu M. Pharmacokinetics of pirfenidone after topical administration in rabbit eye. Mol Vis. 2011;17:2191–6.
    1. Choi K, Lee K, Seung-Wook R, Im M, Kook KH, Choi C. Pirfenidone inhibits transforming growth factor-β1-induced fibrogenesis by blocking nuclear translocation of Smads in human retinal pigment epithelial cell line ARPE-19. Mol Vis. 2012;18:1010–1020.
    1. Yang Y, Ye Y, Lin X, Wu K, Yu M. Inhibition of pirfenidone on TGF-beta2 induced proliferation, migration and epithlial-mesenchymal transition of human lens epithelial cells line SRA01/04. PLoS One. 2013;8:56837.
    1. Kim H, Choi YH, Park SJ, Lee SY, Kim SJ, Jou I, Kook KH. Antifibrotic effect of pirfenidone on orbital fibroblasts of patients with thyroid-associated ophthalmopathy by decreasing TIMP-1 and collagen levels. Invest Ophthalmol Vis Sci. 2010;51:3061–6.
    1. Armendáriz-Borunda J, Islas-Carbajal MC, Meza-García E, Rincón AR, Lucano S, Sandoval AS, Salazar A, Berumen J, Alvarez A, Covarrubias A, Aréchiga G, García L. A pilot study in patients with established advanced liver fibrosis using pirfenidone. Gut. 2006;55:1663–1665.
    1. Flores-Contreras L, Sandoval-Rodríguez AS, Mena-Enriquez MG, Lucano-Landeros S, Arellano-Olivera I, Alvarez-Álvarez A, Sanchez-Parada MG, Armendáriz-Borunda J. Treatment with pirfenidone for two years decreases fibrosis, cytokine levels and enhances CB2 gene expression in patients with chronic hepatitis C. BMC Gastroenterol. 2014;14:131.
    1. Tsuchiya H, Kaibori M, Yanagida H, Yokoigawa N, Kwon AH, Okumura T, Kamiyama Y. Pirfenidone prevents endotoxin-induced liver injury after partial hepatectomy in rats. J Hepatol. 2004;40:94–101.
    1. Wang F, Wen T, Chen XY, Wu H. Protective effects of pirfenidone on D-galactosamine and lipopolysaccharide-induced acute hepatotoxicity in rats. Inflamm Res. 2008;57:183–188.
    1. García L, Hernández I, Sandoval A, Salazar A, Garcia J, Vera J, Grijalva G, Muriel P, Margolin S, Armendariz-Borunda J. Pirfenidone effectively reverses experimental liver fibrosis. J Hepatol. 2002;37:797–805.
    1. Di Sario A, Bendia E, Macarri G, Candelaresi C, Taffetani S, Marzioni M, Omenetti A, De Minicis S, Trozzi L, Benedetti A. The anti-fibrotic effects of pirfenidone in rat liver fibrosis is mediated by downregulation of procollagen alpha 1(l), TIMP-1 and MMP2. DigLiverDis. 2004;36:744–751.
    1. Chen JF, Ni HF, Pan MM, Liu H, Xu M, Zhang MH, Liu BC. Pirfenidone inhibits macrophage infiltration in 5/6 nephrectomized rats. Am J Physiol Renal Physiol. 2013;15:304.
    1. Chen JF, Liu H, Ni HF, Lv LL, Zhang MH, Zhang AH, Tang RN, Chen PS, Liu BC. Improved mitochondrial function underlies the protective effect of pirfenidone against tubulointerstitial fibrosis in 5/6 nephrectomized rats. PLoS One. 2013;8:83593.
    1. Racanelli V, Rehermann B. The liver as an immunological organ. Hepatology. 2006;43(2 Suppl 1):S54–62.
    1. : Bethesda (MD). Safety and tolerability of pirfenidone in patients with systemic sclerosis-related interstitial lung disease (SSc-ILD) (LOTUSS). Identifier: NCT01933334. Revised 2 Oct 2014. .

Source: PubMed

3
Suscribir