PPARA Polymorphism Influences the Cardiovascular Benefit of Fenofibrate in Type 2 Diabetes: Findings From ACCORD-Lipid

Mario Luca Morieri, Hetal S Shah, Jennifer Sjaarda, Petra A Lenzini, Hannah Campbell, Alison A Motsinger-Reif, He Gao, Laura Lovato, Sabrina Prudente, Assunta Pandolfi, Marcus G Pezzolesi, Ronald J Sigal, Guillaume Paré, Santica M Marcovina, Daniel M Rotroff, Elisabetta Patorno, Luana Mercuri, Vincenzo Trischitta, Emily Y Chew, Peter Kraft, John B Buse, Michael J Wagner, Sharon Cresci, Hertzel C Gerstein, Henry N Ginsberg, Josyf C Mychaleckyj, Alessandro Doria, Mario Luca Morieri, Hetal S Shah, Jennifer Sjaarda, Petra A Lenzini, Hannah Campbell, Alison A Motsinger-Reif, He Gao, Laura Lovato, Sabrina Prudente, Assunta Pandolfi, Marcus G Pezzolesi, Ronald J Sigal, Guillaume Paré, Santica M Marcovina, Daniel M Rotroff, Elisabetta Patorno, Luana Mercuri, Vincenzo Trischitta, Emily Y Chew, Peter Kraft, John B Buse, Michael J Wagner, Sharon Cresci, Hertzel C Gerstein, Henry N Ginsberg, Josyf C Mychaleckyj, Alessandro Doria

Abstract

The cardiovascular benefits of fibrates have been shown to be heterogeneous and to depend on the presence of atherogenic dyslipidemia. We investigated whether genetic variability in the PPARA gene, coding for the pharmacological target of fibrates (PPAR-α), could be used to improve the selection of patients with type 2 diabetes who may derive cardiovascular benefit from addition of this treatment to statins. We identified a common variant at the PPARA locus (rs6008845, C/T) displaying a study-wide significant influence on the effect of fenofibrate on major cardiovascular events (MACE) among 3,065 self-reported white subjects treated with simvastatin and randomized to fenofibrate or placebo in the ACCORD-Lipid trial. T/T homozygotes (36% of participants) experienced a 51% MACE reduction in response to fenofibrate (hazard ratio 0.49; 95% CI 0.34-0.72), whereas no benefit was observed for other genotypes (P interaction = 3.7 × 10-4). The rs6008845-by-fenofibrate interaction on MACE was replicated in African Americans from ACCORD (N = 585, P = 0.02) and in external cohorts (ACCORD-BP, ORIGIN, and TRIUMPH, total N = 3059, P = 0.005). Remarkably, rs6008845 T/T homozygotes experienced a cardiovascular benefit from fibrate even in the absence of atherogenic dyslipidemia. Among these individuals, but not among carriers of other genotypes, fenofibrate treatment was associated with lower circulating levels of CCL11-a proinflammatory and atherogenic chemokine also known as eotaxin (P for rs6008845-by-fenofibrate interaction = 0.003). The GTEx data set revealed regulatory functions of rs6008845 on PPARA expression in many tissues. In summary, we have found a common PPARA regulatory variant that influences the cardiovascular effects of fenofibrate and that could be used to identify patients with type 2 diabetes who would derive benefit from fenofibrate treatment, in addition to those with atherogenic dyslipidemia.

© 2020 by the American Diabetes Association.

Figures

Figure 1
Figure 1
Regional plot of the PPARA gene region. Each point represents one SNP. The base pair position on chromosome (chr) 22 (from 46.5 to 46.7 Mb) is on the x-axis, and the negative log transformation of the P value for interaction between each SNP and fenofibrate on the primary outcome is on the y-axis. The significance threshold (P = 6.2 × 10−4) after adjustment for multiple comparisons is indicated by the dashed line. Colors indicate the amount of linkage disequilibrium between plotted SNPs and rs6008845 (specific information of top SNPs can be found in Supplementary Table 2). Plots were generated using LocusZoom v1.1 (Abecasis Laboratory, University of Michigan School of Public Health).
Figure 2
Figure 2
rs6008845 association with primary outcome stratified by fenofibrate treatment in discovery and validation cohorts of subjects with type 2 diabetes and high cardiovascular risk. Note: the association between rs6008845 and the primary outcome is depicted as the effect per each T allele copy.
Figure 3
Figure 3
Fenofibrate cardiovascular effectiveness according to rs6008845 genotypes. Top panel: among all self-reported white subjects randomized to fenofibrate or placebo in the ACCORD-Lipid study. Middle and bottom panels: in the same population according to absence or presence of atherogenic dyslipidemia at baseline (a few subjects, N = 15, were not included due to missing data on lipid profile at baseline).
Figure 4
Figure 4
Effects of fenofibrate on changes in lipid levels among self-reported whites in ACCORD-Lipid, stratified by rs6008845 genotypes. Error bars represent SEs. One SD is equal to 5.4 mg/dL for HDL-c, 87.1 mg/dL for triglycerides, 35.2 mg/dL for total cholesterol, and 29.8 mg/dL for LDL cholesterol. To convert cholesterol values to millimoles per liter, multiply by 0.02586. To convert the values for triglycerides to millimoles per liter, multiply by 0.01129. Chol, cholesterol.
Figure 5
Figure 5
Fenofibrate effects on chemokine levels according to rs6008845 genotypes among self-reported whites from the ACCORD-MIND biomarker study. Error bars represent SEs.

References

    1. American Diabetes Association 10. Cardiovascular disease and risk management: Standards of Medical Care in Diabetes–2019. Diabetes Care 2019;42(Suppl. 1):S103–S123
    1. Taskinen MR, Borén J. New insights into the pathophysiology of dyslipidemia in type 2 diabetes. Atherosclerosis 2015;239:483–495
    1. Fruchart JC, Sacks F, Hermans MP, et al. . The Residual Risk Reduction Initiative: a call to action to reduce residual vascular risk in patients with dyslipidemia. Am J Cardiol 2008;102(Suppl.):1K–34K
    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. 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. Effect of fenofibrate on progression of coronary-artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet 2001;357:905–910
    1. Rubins HB, Robins SJ, Collins D, et al. .; Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group . Gemfibrozil for the secondary prevention of coronary heart disease in men with low levels of high-density lipoprotein cholesterol. N Engl J Med 1999;341:410–418
    1. Belfort R, Berria R, Cornell J, Cusi K. Fenofibrate reduces systemic inflammation markers independent of its effects on lipid and glucose metabolism in patients with the metabolic syndrome. J Clin Endocrinol Metab 2010;95:829–836
    1. Staels B, Koenig W, Habib A, et al. . Activation of human aortic smooth-muscle cells is inhibited by PPARalpha but not by PPARgamma activators. Nature 1998;393:790–793
    1. Keene D, Price C, Shun-Shin MJ, Francis DP. Effect on cardiovascular risk of high density lipoprotein targeted drug treatments niacin, fibrates, and CETP inhibitors: meta-analysis of randomised controlled trials including 117,411 patients. BMJ 2014;349:g4379.
    1. Jun M, Foote C, Lv J, et al. . Effects of fibrates on cardiovascular outcomes: a systematic review and meta-analysis. Lancet 2010;375:1875–1884
    1. Sacks FM, Carey VJ, Fruchart JC. Combination lipid therapy in type 2 diabetes. N Engl J Med 2010;363:692–694
    1. Bruckert E, Labreuche J, Deplanque D, Touboul PJ, Amarenco P. Fibrates effect on cardiovascular risk is greater in patients with high triglyceride levels or atherogenic dyslipidemia profile: a systematic review and meta-analysis. J Cardiovasc Pharmacol 2011;57:267–272
    1. Kim NH, Han KH, Choi J, Lee J, Kim SG. Use of fenofibrate on cardiovascular outcomes in statin users with metabolic syndrome: propensity matched cohort study. BMJ 2019;366:l5125.
    1. NICE Clinical Guideline Centre Lipid Modification: Cardiovascular Risk Assessment and the Modification of Blood Lipids for the Primary and Secondary Prevention of Cardiovascular Disease. London, National Institute for Health and Care Excellence, 2014
    1. Ferrari R, Aguiar C, Alegria E, et al. . Current practice in identifying and treating cardiovascular risk, with a focus on residual risk associated with atherogenic dyslipidaemia. Eur Heart J Suppl 2016;18(Suppl. C):C2–C12
    1. Frazier-Wood AC, Ordovas JM, Straka RJ, et al. . The PPAR alpha gene is associated with triglyceride, low-density cholesterol and inflammation marker response to fenofibrate intervention: the GOLDN study. Pharmacogenomics J 2013;13:312–317
    1. Smith JA, Arnett DK, Kelly RJ, et al. . The genetic architecture of fasting plasma triglyceride response to fenofibrate treatment. Eur J Hum Genet 2008;16:603–613
    1. Gerstein HC, Miller ME, Byington RP, et al. .; Action to Control Cardiovascular Risk in Diabetes Study Group . Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008;358:2545–2559
    1. Cushman WC, Evans GW, Byington RP, et al. .; ACCORD Study Group . Effects of intensive blood-pressure control in type 2 diabetes mellitus. N Engl J Med 2010;362:1575–1585
    1. Bosch J, Gerstein HC, Dagenais GR, et al. .; ORIGIN Trial Investigators . n-3 fatty acids and cardiovascular outcomes in patients with dysglycemia. N Engl J Med 2012;367:309–318
    1. Arnold SV, Chan PS, Jones PG, et al. .; Cardiovascular Outcomes Research Consortium . Translational Research Investigating Underlying Disparities in Acute Myocardial Infarction Patients’ Health Status (TRIUMPH): design and rationale of a prospective multicenter registry. Circ Cardiovasc Qual Outcomes 2011;4:467–476
    1. Samaropoulos XF, Light L, Ambrosius WT, Marcovina SM, Probstfield J, Goff DC Jr. The effect of intensive risk factor management in type 2 diabetes on inflammatory biomarkers. Diabetes Res Clin Pract 2012;95:389–398
    1. Shah HS, Morieri ML, Marcovina SM, et al. . Modulation of GLP-1 levels by a genetic variant that regulates the cardiovascular effects of intensive glycemic control in ACCORD. Diabetes Care 2018;41:348–355
    1. Shah HS, Gao H, Morieri ML, et al. . Genetic predictors of cardiovascular mortality during intensive glycemic control in type 2 diabetes: findings from the ACCORD clinical trial. Diabetes Care 2016;39:1915–1924
    1. Gao X, Starmer J, Martin ER. A multiple testing correction method for genetic association studies using correlated single nucleotide polymorphisms. Genet Epidemiol 2008;32:361–369
    1. Altman DG, Andersen PK. Calculating the number needed to treat for trials where the outcome is time to an event. BMJ 1999;319:1492–1495
    1. GTEx Consortium Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 2015;348:648–660
    1. Han B, Eskin E. Random-effects model aimed at discovering associations in meta-analysis of genome-wide association studies. Am J Hum Genet 2011;88:586–598
    1. Willer CJ, Schmidt EM, Sengupta S, et al. .; Global Lipids Genetics Consortium . Discovery and refinement of loci associated with lipid levels. Nat Genet 2013;45:1274–1283
    1. Ahola-Olli AV, Würtz P, Havulinna AS, et al. . Genome-wide association study identifies 27 loci influencing concentrations of circulating cytokines and growth factors. Am J Hum Genet 2017;100:40–50
    1. Morieri ML, Shah H, Doria A; Action to Control Cardiovascular Risk in Diabetes (ACCORD) Genetic Study Group . Variants in ANGPTL4 and the risk of coronary artery disease. N Engl J Med 2016;375:2304–2305
    1. Chanock SJ, Manolio T, Boehnke M, et al. .; NCI-NHGRI Working Group on Replication in Association Studies . Replicating genotype-phenotype associations. Nature 2007;447:655–660
    1. Robins SJ, Collins D, Wittes JT, et al. .; VA-HIT Study Group. Veterans Affairs High-Density Lipoprotein Intervention Trial . Relation of gemfibrozil treatment and lipid levels with major coronary events: VA-HIT: a randomized controlled trial. JAMA 2001;285:1585–1591
    1. Ponath PD, Qin S, Ringler DJ, et al. . Cloning of the human eosinophil chemoattractant, eotaxin. Expression, receptor binding, and functional properties suggest a mechanism for the selective recruitment of eosinophils. J Clin Invest 1996;97:604–612
    1. Sallusto F, Mackay CR, Lanzavecchia A. Selective expression of the eotaxin receptor CCR3 by human T helper 2 cells. Science 1997;277:2005–2007
    1. Chen J, Akyürek LM, Fellström B, Häyry P, Paul LC. Eotaxin and capping protein in experimental vasculopathy. Am J Pathol 1998;153:81–90
    1. Haley KJ, Lilly CM, Yang JH, et al. . Overexpression of eotaxin and the CCR3 receptor in human atherosclerosis: using genomic technology to identify a potential novel pathway of vascular inflammation. Circulation 2000;102:2185–2189
    1. Raghuraman G, Hsiung J, Zuniga MC, et al. . Eotaxin augments calcification in vascular smooth muscle cells. J Cell Biochem 2017;118:647–654
    1. Canouï-Poitrine F, Luc G, Mallat Z, et al. .; PRIME Study Group . Systemic chemokine levels, coronary heart disease, and ischemic stroke events: the PRIME study. Neurology 2011;77:1165–1173
    1. Zee RY, Cook NR, Cheng S, et al. . Threonine for alanine substitution in the eotaxin (CCL11) gene and the risk of incident myocardial infarction. Atherosclerosis 2004;175:91–94
    1. Cross DS, McCarty CA, Hytopoulos E, et al. . Coronary risk assessment among intermediate risk patients using a clinical and biomarker based algorithm developed and validated in two population cohorts. Curr Med Res Opin 2012;28:1819–1830
    1. Cameron AR, Morrison VL, Levin D, et al. . Anti-inflammatory effects of metformin irrespective of diabetes status. Circ Res 2016;119:652–665
    1. Loughrey BV, McGinty A, Young IS, McCance DR, Powell LA. Increased circulating CC chemokine levels in the metabolic syndrome are reduced by low-dose atorvastatin treatment: evidence from a randomized controlled trial. Clin Endocrinol (Oxf) 2013;79:800–806
    1. Staumont-Sallé D, Abboud G, Brénuchon C, et al. . Peroxisome proliferator-activated receptor alpha regulates skin inflammation and humoral response in atopic dermatitis. J Allergy Clin Immunol 2008;121:962–968.e6
    1. Marx N, Sukhova GK, Collins T, Libby P, Plutzky J. PPARalpha activators inhibit cytokine-induced vascular cell adhesion molecule-1 expression in human endothelial cells. Circulation 1999;99:3125–3131
    1. Delerive P, Gervois P, Fruchart JC, Staels B. Induction of IkappaBalpha expression as a mechanism contributing to the anti-inflammatory activities of peroxisome proliferator-activated receptor-alpha activators. J Biol Chem 2000;275:36703–36707
    1. Huber MA, Denk A, Peter RU, Weber L, Kraut N, Wirth T. The IKK-2/Ikappa Balpha /NF-kappa B pathway plays a key role in the regulation of CCR3 and eotaxin-1 in fibroblasts. A critical link to dermatitis in Ikappa Balpha -deficient mice. J Biol Chem 2002;277:1268–1275
    1. Seidel P, Roth M, Ge Q, Merfort I, S’ng CT, Ammit AJ. IκBα glutathionylation and reduced histone H3 phosphorylation inhibit eotaxin and RANTES. Eur Respir J 2011;38:1444–1452
    1. Wang WR, Liu EQ, Zhang JY, et al. . Activation of PPAR alpha by fenofibrate inhibits apoptosis in vascular adventitial fibroblasts partly through SIRT1-mediated deacetylation of FoxO1. Exp Cell Res 2015;338:54–63
    1. Ali FY, Armstrong PC, Dhanji AR, et al. . Antiplatelet actions of statins and fibrates are mediated by PPARs. Arterioscler Thromb Vasc Biol 2009;29:706–711
    1. Paumelle R, Staels B. Cross-talk between statins and PPARalpha in cardiovascular diseases: clinical evidence and basic mechanisms. Trends Cardiovasc Med 2008;18:73–78
    1. Balakumar P, Mahadevan N. Interplay between statins and PPARs in improving cardiovascular outcomes: a double-edged sword? Br J Pharmacol 2012;165:373–379
    1. U.S. Food and Drug Administration FDA notice: 81 FR 22612; docket no. FDA-2016-N-1127, 2016. Available from . Accessed 6 February 2020

Source: PubMed

3
Subscribe