Empagliflozin and uric acid metabolism in diabetes: A post hoc analysis of the EMPA-REG OUTCOME trial

João Pedro Ferreira, Silvio E Inzucchi, Michaela Mattheus, Thomas Meinicke, Dominik Steubl, Christoph Wanner, Bernard Zinman, João Pedro Ferreira, Silvio E Inzucchi, Michaela Mattheus, Thomas Meinicke, Dominik Steubl, Christoph Wanner, Bernard Zinman

Abstract

Aim: To evaluate the effect of empagliflozin on uric acid (UA) levels, antigout medication and gout episodes in the EMPA-REG OUTCOME trial (NCT01131676).

Materials and methods: A total of 7020 patients with type 2 diabetes (T2D) were randomized to either empagliflozin (10 or 25 mg) or placebo. The effects of empagliflozin versus placebo on UA concentration were assessed using mixed linear models. A composite outcome of new prescription of antigout medication or gout episode was studied with Cox proportional hazards models.

Results: Empagliflozin reduced serum UA levels versus placebo: week 52 adjusted mean treatment difference = -0.37 (95% confidence interval [CI] -0.42, -0.31) mg/dL; this was more pronounced in patients with baseline UA ≥ 7.0 mg/dL versus <7.0 mg/dL: week 52 adjusted mean treatment difference = -0.56 (95% CI -0.68, -0.43) and -0.30 (95% CI -0.37, -0.24) mg/dL, respectively. Among 6607 patients not taking antigout medications at baseline, 5.2% had a gout episode or initiated antigout treatment versus 3.6% in the placebo and empagliflozin groups, respectively: hazard ratio 0.67 (95% CI 0.53, 0.85; P = 0.001). Both components of the composite outcome contributed to the reduction with empagliflozin in the composite. Risk reduction was similar with both empagliflozin doses.

Conclusions: Empagliflozin reduced UA levels and the composite of gout episodes or prescription of antigout medication. These clinically important findings expand the utility of empagliflozin as a potential antigout treatment in patients with T2D, beyond its well-established cardio-renal benefits.

Keywords: antigout treatment; empagliflozin; gout; type 2 diabetes; uric acid.

Conflict of interest statement

J.P.F. is a consultant for Boehringer Ingelheim. S.E.I. reports membership on scientific/research advisory boards for Boehringer Ingelheim, AstraZeneca, Intarcia, Lexicon, Janssen, Sanofit, Merck & Co. and Novo Nordisk, has received research supplies to Yale University from Takeda, and has participated in medical educational projects, for which unrestricted funding from Boehringer Ingelheim, Eli Lilly, and Merck & Co. was received by Yale University. C.W. has received honoraria for consultancy and lecturing from AstraZeneca, Bayer, BI, GlaxoSmithKline, Eli Lilly and Company, Merck Sharp & Dome, Mundipharma, Sanofi Genzyme and Takeda. B.Z. received consulting fees from Boehringer Ingelheim, Novo Nordisk and Eli Lilly. M.M., T.M. and D.S. are Boehringer Ingelheim employees.

© 2021 The Authors. Diabetes, Obesity and Metabolism published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Effect of empagliflozin (10 and 25 mg doses pooled) versus placebo on uric acid (UA) levels (mg/dL) over time (on‐treatment). Mixed‐effect model repeated measures analyses included the continuous fixed effects of baseline UA and baseline glycated haemoglobin at each visit and discrete fixed effects for treatment at each visit, sex, region, baseline estimated glomerular filtration rate, baseline body mass index and last projected visit
FIGURE 2
FIGURE 2
Time to new onset of gout or antigout medication during on‐treatment phase, for patients not on antigout medication at baseline. Hazard ratio (HR) based on Cox proportional hazards model with terms for age, sex, geographic region, baseline glycated haemoglobin, baseline estimated glomerular filtration rate, baseline body mass index and treatment. CI, confidence interval
FIGURE 3
FIGURE 3
Effect of empagliflozin on new onset of gout or initiation of antigout medication during follow‐up (on‐treatment). †Per 1000 patient‐years. ‡Based on Cox regression with terms for age, sex, geographic region, baseline body mass index, baseline glycated haemoglobin, baseline estimated glomerular filtration rate, and treatment. §Analysis performed in patients not on antigout medication at baseline (n = 6607). ¶Analysis performed in all patients (n = 7020). CI, confidence interval; HR, hazard ratio

References

    1. Verma S, Ji Q, Bhatt DL, et al. Association between uric acid levels and cardio‐renal outcomes and death in patients with type 2 diabetes: a subanalysis of EMPA‐REG OUTCOME. Diabetes Obes Metab. 2020;22(7):1207‐1214.
    1. Yuan H, Yu C, Li X, et al. Serum uric acid levels and risk of metabolic syndrome: a dose‐response meta‐analysis of prospective studies. J Clin Endocrinol Metab. 2015;100(11):4198‐4207.
    1. Zoppini G, Targher G, Chonchol M, et al. Serum uric acid levels and incident chronic kidney disease in patients with type 2 diabetes and preserved kidney function. Diabetes Care. 2012;35(1):99‐104.
    1. Kim IY, Lee DW, Lee SB, Kwak IS. The role of uric acid in kidney fibrosis: experimental evidences for the causal relationship. Biomed Res Int. 2014;2014:638732‐638739.
    1. Bailey CJ. Uric acid and the cardio‐renal effects of SGLT2 inhibitors. Diabetes Obes Metab. 2019;21(6):1291‐1298.
    1. Clarson LE, Chandratre P, Hider SL, et al. Increased cardiovascular mortality associated with gout: a systematic review and meta‐analysis. Eur J Prev Cardiol. 2015;22(3):335‐343.
    1. Maiuolo J, Oppedisano F, Gratteri S, Muscoli C, Mollace V. Regulation of uric acid metabolism and excretion. Int J Cardiol. 2016;213:8‐14.
    1. Chino Y, Samukawa Y, Sakai S, et al. SGLT2 inhibitor lowers serum uric acid through alteration of uric acid transport activity in renal tubule by increased glycosuria. Biopharm Drug Dispos. 2014;35(7):391‐404.
    1. McNally JS, Saxena A, Cai H, Dikalov S, Harrison DG. Regulation of xanthine oxidoreductase protein expression by hydrogen peroxide and calcium. Arterioscler Thromb Vasc Biol. 2005;25(8):1623‐1628.
    1. Packer M. Cardioprotective effects of Sirtuin‐1 and its downstream effectors: potential role in mediating the heart failure benefits of SGLT2 (sodium‐glucose cotransporter 2) inhibitors. Circ Heart Fail. 2020;13:e007197.
    1. Zhao Y, Xu L, Tian D, et al. Effects of sodium‐glucose co‐transporter 2 (SGLT2) inhibitors on serum uric acid level: a meta‐analysis of randomized controlled trials. Diabetes Obes Metab. 2018;20(2):458‐462.
    1. McGuire DK, Shih WJ, Cosentino F, et al. Association of SGLT2 inhibitors with cardiovascular and kidney outcomes in patients with type 2 diabetes: a meta‐analysis. JAMA Cardiol. 2020;6:148.
    1. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta‐analysis of cardiovascular outcome trials. Lancet. 2019;393(10166):31‐39.
    1. Neuen BL, Young T, Heerspink HJL, et al. SGLT2 inhibitors for the prevention of kidney failure in patients with type 2 diabetes: a systematic review and meta‐analysis. Lancet Diabetes Endocrinol. 2019;7(11):845‐854.
    1. Zannad F, Ferreira JP, Pocock SJ, et al. SGLT2 inhibitors in patients with heart failure with reduced ejection fraction: a meta‐analysis of the EMPEROR‐reduced and DAPA‐HF trials. Lancet. 2020;396:819‐829.
    1. Inzucchi SE, Zinman B, Fitchett D, et al. How does empagliflozin reduce cardiovascular mortality? Insights from a mediation analysis of the EMPA‐REG OUTCOME Trial. Diabetes Care. 2018;41(2):356‐363.
    1. Kuo CF, Grainge MJ, Zhang W, Doherty M. Global epidemiology of gout: prevalence, incidence and risk factors. Nat Rev Rheumatol. 2015;11(11):649‐662.
    1. Li J, Badve S, Zhou Z. The effects of canagliflozin on gout in type 2 diabetes: a post‐hoc analysis of the CANVAS program. Lancet Rheumatol. 2019;1(4):E220–E228.
    1. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117‐2128.
    1. Waldman B, Ansquer JC, Sullivan DR, et al. Effect of fenofibrate on uric acid and gout in type 2 diabetes: a post‐hoc analysis of the randomised, controlled FIELD study. Lancet Diabetes Endocrinol. 2018;6(4):310‐318.
    1. Beyersmann J, Allignol A, Schumacher M. Competing Risks and Multistate Models with R. Springer; 2011.
    1. Ferreira JP, Kraus BJ, Zwiener I, et al. Cardio/kidney composite end points: a post hoc analysis of the EMPA‐REG OUTCOME trial. J Am Heart Assoc. 2021;10:e020053.
    1. Cutolo M, Cimmino MA, Perez‐Ruiz F. Potency on lowering serum uric acid in gout patients: a pooled analysis of registrative studies comparing febuxostat vs. allopurinol. Eur Rev Med Pharmacol Sci. 2017;21(18):4186‐4195.
    1. Schumacher HR Jr, Becker MA, Wortmann RL, et al. Effects of febuxostat versus allopurinol and placebo in reducing serum urate in subjects with hyperuricemia and gout: a 28‐week, phase III, randomized, double‐blind, parallel‐group trial. Arthritis Rheum. 2008;59(11):1540‐1548.
    1. Taylor TH, Mecchella JN, Larson RJ, Kerin KD, Mackenzie TA. Initiation of allopurinol at first medical contact for acute attacks of gout: a randomized clinical trial. Am J Med. 2012;125(11):1126‐1134.e7.
    1. Dalbeth N, Saag KG, Palmer WE, et al. Effects of febuxostat in early gout: a randomized, double‐blind, placebo‐controlled study. Arthritis Rheumatol. 2017;69(12):2386‐2395.
    1. Solomon DH, Glynn RJ, MacFadyen JG, et al. Relationship of interleukin‐1β blockade with incident gout and serum uric acid levels: exploratory analysis of a randomized controlled trial. Ann Intern Med. 2018;169(8):535‐542.
    1. Uetake D, Ohno I, Ichida K, et al. Effect of fenofibrate on uric acid metabolism and urate transporter 1. Intern Med. 2010;49(2):89‐94.
    1. Nakanishi T, Ohya K, Shimada S, Anzai N, Tamai I. Functional cooperation of URAT1 (SLC22A12) and URATv1 (SLC2A9) in renal reabsorption of urate. Nephrol Dial Transplant. 2013;28(3):603‐611.
    1. Würzner G, Gerster JC, Chiolero A, et al. Comparative effects of losartan and irbesartan on serum uric acid in hypertensive patients with hyperuricaemia and gout. J Hypertens. 2001;19(10):1855‐1860.
    1. Lee JW, Bajwa PJ, Carson MJ, et al. Fenofibrate represses interleukin‐17 and interferon‐gamma expression and improves colitis in interleukin‐10‐deficient mice. Gastroenterology. 2007;133(1):108‐123.
    1. Heerspink HJL, Perco P, Mulder S, et al. Canagliflozin reduces inflammation and fibrosis biomarkers: a potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia. 2019;62(7):1154‐1166.
    1. Mancini SJ, Boyd D, Katwan OJ, et al. Canagliflozin inhibits interleukin‐1β‐stimulated cytokine and chemokine secretion in vascular endothelial cells by AMP‐activated protein kinase‐dependent and ‐independent mechanisms. Sci Rep. 2018;8(1):5276.
    1. Maayah ZH, Ferdaoussi M, Takahara S, Soni S, Dyck JRB. Empagliflozin suppresses inflammation and protects against acute septic renal injury. Inflammopharmacology. 2021;29(1):269‐279.
    1. Huang XF, Li HQ, Shi L, Xue JY, Ruan BF, Zhu HL. Synthesis of resveratrol analogues, and evaluation of their cytotoxic and xanthine oxidase inhibitory activities. Chem Biodivers. 2008;5(4):636‐642.
    1. Rodgers JT, Lerin C, Haas W, Gygi SP, Spiegelman BM, Puigserver P. Nutrient control of glucose homeostasis through a complex of PGC‐1alpha and SIRT1. Nature. 2005;434(7029):113‐118.

Source: PubMed

3
구독하다