An update on the molecular actions of fenofibrate and its clinical effects on diabetic retinopathy and other microvascular end points in patients with diabetes

Jonathan E Noonan, Alicia J Jenkins, Jian-Xing Ma, Anthony C Keech, Jie Jin Wang, Ecosse L Lamoureux, Jonathan E Noonan, Alicia J Jenkins, Jian-Xing Ma, Anthony C Keech, Jie Jin Wang, Ecosse L Lamoureux

Abstract

The drug fenofibrate has received major attention as a novel medical treatment for diabetic retinopathy (DR) and other diabetes-induced microvascular complications. This interest stems from two recent large, well-designed clinical trials that demonstrated large reductions in the progression of DR and the need for laser intervention, in addition to a reduction in renal and neurological outcomes, in patients with type 2 diabetes. In both trials, the greatest benefit on DR progression was observed in those patients with DR at baseline. Originally considered a lipid-modifying drug, it now appears that multiple mechanisms may underpin the benefit of fenofibrate on diabetic microvascular end points. Fenofibrate regulates the expression of many different genes, with a range of beneficial effects on lipid control, inflammation, angiogenesis, and cell apoptosis. These factors are believed to be important in the development of DR regardless of the underlying diabetes etiology. Cell experiments have demonstrated improved survival of retinal endothelial and pigment epithelial cells in conjunction with reduced stress signaling under diabetic conditions. Further, fenofibrate improves retinal outcomes in rodent models of diabetes and retinal neovascularization. Given the results of these preclinical studies, further clinical trials are needed to establish the benefits of fenofibrate in other forms of diabetes, including type 1 diabetes. In DR management, fenofibrate could be a useful adjunctive treatment to modifiable risk factor control and regular ophthalmic review. Its incorporation into clinical practice should be continually revised as more information becomes available.

Figures

FIG. 1.
FIG. 1.
Chemical structure of fenofibrate. Lines indicate carbon bonds. O, oxygen; Cl, chlorine.
FIG. 2.
FIG. 2.
Identified lipid and nonlipid molecular actions of fenofibrate and its active metabolite, fenofibric acid. IGF1R, IGF-1 receptor; LRP-6, LDL receptor–related protein-6; NF-κB, nuclear factor-κB.
FIG. 3.
FIG. 3.
Classical fenofibrate signaling pathway. Fenofibrate is rapidly converted to fenofibric acid (FA) in vivo by tissue and plasma esterases before entering the cell. Fenofibric acid binds to PPARα and forms a heterodimer complex with retinoid X receptor (RXR). This complex then binds to specific peroxisome proliferator response elements (PPREs) to activate target gene transcription. RA, 9-cis retinoic acid.
FIG. 4.
FIG. 4.
Canonical Wnt signaling in an endothelial cell. A: Wnt ligand induces recruitment of dishevelled (Dsh) and Axin to the Frizzled receptor and LDL receptor–related protein (LRP) coreceptor, respectively, and inhibition of the β-catenin destruction complex. This allows β-catenin to accumulate in the cytoplasm and translocate to the nucleus, where it binds with T-cell factor (TCF) and activates transcription of proangiogenic genes. B: Fenofibrate (F) appears to inhibit Wnt signaling through inhibition of LRP6 phosphorylation. The mechanism is not completely understood but may involve upregulation of the VLDL receptor (VLDLR). APC, adenomatous polyposis coli; GSK3, glycogen synthase kinase-3; P, phosphate.

References

    1. Keech AC, Mitchell P, Summanen PA, et al. FIELD study investigators Effect of fenofibrate on the need for laser treatment for diabetic retinopathy (FIELD study): a randomised controlled trial. Lancet 2007;370:1687–1697
    1. Chew EY, Ambrosius WT, Davis MD, et al. ACCORD Study Group. ACCORD Eye Study Group Effects of medical therapies on retinopathy progression in type 2 diabetes. N Engl J Med 2010;363:233–244
    1. Balfour JA, McTavish D, Heel RC. Fenofibrate. A review of its pharmacodynamic and pharmacokinetic properties and therapeutic use in dyslipidaemia. Drugs 1990;40:260–290
    1. Lefebvre P, Chinetti G, Fruchart J-C, Staels B. Sorting out the roles of PPARα in energy metabolism and vascular homeostasis. J Clin Invest 2006;116:571–580
    1. Guérin M, Bruckert E, Dolphin PJ, Turpin G, Chapman MJ. Fenofibrate reduces plasma cholesteryl ester transfer from HDL to VLDL and normalizes the atherogenic, dense LDL profile in combined hyperlipidemia. Arterioscler Thromb Vasc Biol 1996;16:763–772
    1. Nigon F, Lesnik P, Rouis M, Chapman MJ. Discrete subspecies of human low density lipoproteins are heterogeneous in their interaction with the cellular LDL receptor. J Lipid Res 1991;32:1741–1753
    1. Dejager S, Bruckert E, Chapman MJ. Dense low density lipoprotein subspecies with diminished oxidative resistance predominate in combined hyperlipidemia. J Lipid Res 1993;34:295–308
    1. Delerive P, De Bosscher K, Besnard S, et al. Peroxisome proliferator-activated receptor α negatively regulates the vascular inflammatory gene response by negative cross-talk with transcription factors NF-κB and AP-1. J Biol Chem 1999;274:32048–32054
    1. Staels B, Koenig W, Habib A, et al. Activation of human aortic smooth-muscle cells is inhibited by PPARα but not by PPARγ activators. Nature 1998;393:790–793
    1. Marx N, Sukhova GK, Collins T, Libby P, Plutzky J. PPARα activators inhibit cytokine-induced vascular cell adhesion molecule-1 expression in human endothelial cells. Circulation 1999;99:3125–3131
    1. Pasceri V, Cheng JS, Willerson JT, Yeh ETH. Modulation of C-reactive protein-mediated monocyte chemoattractant protein-1 induction in human endothelial cells by anti-atherosclerosis drugs. Circulation 2001;103:2531–2534
    1. Shu H, Wong B, Zhou G, et al. Activation of PPARα or γ reduces secretion of matrix metalloproteinase 9 but not interleukin 8 from human monocytic THP-1 cells. Biochem Biophys Res Commun 2000;267:345–349
    1. Panigrahy D, Kaipainen A, Huang S, et al. PPARα agonist fenofibrate suppresses tumor growth through direct and indirect angiogenesis inhibition. Proc Natl Acad Sci U S A 2008;105:985–990
    1. Meissner M, Stein M, Urbich C, et al. PPARα activators inhibit vascular endothelial growth factor receptor-2 expression by repressing Sp1-dependent DNA binding and transactivation. Circ Res 2004;94:324–332
    1. Yokoyama Y, Xin B, Shigeto T, et al. Clofibric acid, a peroxisome proliferator-activated receptor alpha ligand, inhibits growth of human ovarian cancer. Mol Cancer Ther 2007;6:1379–1386
    1. Chen Y, Hu Y, Mott R, et al. Mechanisms for the therapeutic effect of fenofibrate on diabetic retinopathy in type 1 diabetes models. Presented at the 71st Scientific Sessions of the American Diabetes Association, 24–28 June 2011, San Diego, California
    1. Tomizawa A, Hattori Y, Inoue T, Hattori S, Kasai K. Fenofibrate suppresses microvascular inflammation and apoptosis through adenosine monophosphate-activated protein kinase activation. Metabolism 2011;60:513–522
    1. Kadian S, Mahadevan N, Balakumar P. Differential effects of low-dose fenofibrate treatment in diabetic rats with early onset nephropathy and established nephropathy. Eur J Pharmacol 2013;698:388–396
    1. Ibarra-Lara L, Hong E, Soria-Castro E, et al. Clofibrate PPARα activation reduces oxidative stress and improves ultrastructure and ventricular hemodynamics in no-flow myocardial ischemia. J Cardiovasc Pharmacol 2012;60:323–334
    1. Duncan LJ, Cullen JF, Ireland JT, Nolan J, Clarke BF, Oliver MF. A three-year trial of atromid therapy in exudative diabetic retinopathy. Diabetes 1968;17:458–467
    1. Harrold BP, Marmion VJ, Gough KR. A double-blind controlled trial of clofibrate in the treatment of diabetic retinopathy. Diabetes 1969;18:285–291
    1. Cullen JF, Town SM, Campbell CJ. Double-blind trial of Atromid-S in exudative diabetic retinopathy. Trans Ophthalmol Soc U K 1974;94:554–562
    1. Dorne PA. [Exudative diabetic retinopathy. The use of clofibrate in the treatment of hard exudates using a reduced but prolonged dosage over several years (author’s transl)]. Arch Ophtalmol (Paris) 1977;37:393–400
    1. Freyberger H, Schifferdecker E, Schatz H. Regression of hard exudates in diabetic background retinopathy in therapy with etofibrate antilipemic agent. Med Klin (Munich) 1994;89:594–597, 633
    1. Emmerich KH, Poritis N, Stelmane I, et al. [Efficacy and safety of etofibrate in patients with non-proliferative diabetic retinopathy]. Klin Monatsbl Augenheilkd 2009;226:561–567 [in German]
    1. Havel E, Rencova E, Novak J, et al. Serum lipoproteins lowering and diabetic exudative retinopathy. Atherosclerosis 1997;134:309
    1. Morita H, Nagai R. Retinopathy progression in type 2 diabetes. N Engl J Med 2010;363:2171–; author reply 2173–2174.
    1. Early Treatment Diabetic Retinopathy Study Research Group Fundus photographic risk factors for progression of diabetic retinopathy. ETDRS report number 12. Ophthalmology 1991;98(Suppl.):823–833
    1. Early Treatment Diabetic Retinopathy Study Research Group Early photocoagulation for diabetic retinopathy. ETDRS report number 9. Ophthalmology 1991;98(Suppl.):766–785
    1. Ho AC, Scott IU, Kim SJ, et al. Anti-vascular endothelial growth factor pharmacotherapy for diabetic macular edema: a report by the American Academy of Ophthalmology. Ophthalmology 2012;119:2179–2188
    1. Davis TME, Ting R, Best JD, et al. Fenofibrate Intervention and Event Lowering in Diabetes Study investigators Effects of fenofibrate on renal function in patients with type 2 diabetes mellitus: the Fenofibrate Intervention and Event Lowering in Diabetes (FIELD) Study. Diabetologia 2011;54:280–290
    1. Mychaleckyj JC, Craven T, Nayak U, et al. Reversibility of fenofibrate therapy-induced renal function impairment in ACCORD type 2 diabetic participants. Diabetes Care 2012;35:1008–1014
    1. Rajamani K, Colman PG, Li LP, et al. FIELD study investigators Effect of fenofibrate on amputation events in people with type 2 diabetes mellitus (FIELD study): a prespecified analysis of a randomised controlled trial. Lancet 2009;373:1780–1788
    1. Chew EY, Ambrosius WT, Howard LT, et al. ACCORD Study Group Rationale, design, and methods of the Action to Control Cardiovascular Risk in Diabetes Eye Study (ACCORD-EYE). Am J Cardiol 2007;99:103i–111i
    1. Keech A, Simes RJ, Barter P, et al. FIELD study investigators Effects of long-term fenofibrate therapy on cardiovascular events in 9795 people with type 2 diabetes mellitus (the FIELD study): randomised controlled trial. Lancet 2005;366:1849–1861
    1. Ting RD, Keech AC, Drury PL, et al. FIELD Study Investigators Benefits and safety of long-term fenofibrate therapy in people with type 2 diabetes and renal impairment: the FIELD Study. Diabetes Care 2012;35:218–225
    1. Ginsberg HN, Elam MB, Lovato LC, et al. ACCORD Study Group Effects of combination lipid therapy in type 2 diabetes mellitus. N Engl J Med 2010;362:1563–1574
    1. Taskinen MR, Barter PJ, Ehnholm C, et al. FIELD study investigators Ability of traditional lipid ratios and apolipoprotein ratios to predict cardiovascular risk in people with type 2 diabetes. Diabetologia 2010;53:1846–1855
    1. Simo R, Garcia-Ramirez M, Higuera M, Hernandez C. Apolipoprotein A1 is overexpressed in the retina of diabetic patients. Am J Ophthalmol 2009;147:319–325.e311
    1. Sasongko MB, Wong TY, Nguyen TT, et al. Serum apolipoprotein AI and B are stronger biomarkers of diabetic retinopathy than traditional lipids. Diabetes Care 2011;34:474–479
    1. Kim J, Ahn J-H, Kim J-H, et al. Fenofibrate regulates retinal endothelial cell survival through the AMPK signal transduction pathway. Exp Eye Res 2007;84:886–893
    1. Chen Y, Hu Y, Lin M, et al. Therapeutic effects of PPARα agonists on diabetic retinopathy in type 1 diabetes models. Diabetes 2013;62:261–272
    1. Cacicedo JM, Benjachareonwong S, Chou E, Yagihashi N, Ruderman NB, Ido Y. Activation of AMP-activated protein kinase prevents lipotoxicity in retinal pericytes. Invest Ophthalmol Vis Sci 2011;52:3630–3639
    1. Lyons TJ, Li W, Wells-Knecht MC, Jokl R. Toxicity of mildly modified low-density lipoproteins to cultured retinal capillary endothelial cells and pericytes. Diabetes 1994;43:1090–1095
    1. Wu M, Chen Y, Wilson K, et al. Intraretinal leakage and oxidation of LDL in diabetic retinopathy. Invest Ophthalmol Vis Sci 2008;49:2679–2685
    1. Barth JL, Yu Y, Song W, et al. Oxidised, glycated LDL selectively influences tissue inhibitor of metalloproteinase-3 gene expression and protein production in human retinal capillary pericytes. Diabetologia 2007;50:2200–2208
    1. Villarroel M, Garcia-Ramírez M, Corraliza L, Hernández C, Simó R. Fenofibric acid prevents retinal pigment epithelium disruption induced by interleukin-1β by suppressing AMP-activated protein kinase (AMPK) activation. Diabetologia 2011;54:1543–1553
    1. Trudeau K, Roy S, Guo W, et al. Fenofibric acid reduces fibronectin and collagen type IV overexpression in human retinal pigment epithelial cells grown in conditions mimicking the diabetic milieu: functional implications in retinal permeability. Invest Ophthalmol Vis Sci 2011;52:6348–6354
    1. Miranda S, González-Rodríguez Á, García-Ramírez M, et al. Beneficial effects of fenofibrate in retinal pigment epithelium by the modulation of stress and survival signaling under diabetic conditions. J Cell Physiol 2012;227:2352–2362
    1. Chen Y, Hu Y, Lu K, Flannery JG, Ma JX. Very low density lipoprotein receptor, a negative regulator of the wnt signaling pathway and choroidal neovascularization. J Biol Chem 2007;282:34420–34428

Source: PubMed

3
購読する