Short-Term Changes in Albuminuria and Risk of Cardiovascular and Renal Outcomes in Type 2 Diabetes Mellitus: A Post Hoc Analysis of the EMPA-REG OUTCOME Trial

Simke W Waijer, Di Xie, Silvio E Inzucchi, Bernard Zinman, Audrey Koitka-Weber, Michaela Mattheus, Maximillian von Eynatten, Lesley A Inker, Christoph Wanner, Hiddo J L Heerspink, Simke W Waijer, Di Xie, Silvio E Inzucchi, Bernard Zinman, Audrey Koitka-Weber, Michaela Mattheus, Maximillian von Eynatten, Lesley A Inker, Christoph Wanner, Hiddo J L Heerspink

Abstract

Background Early reduction in albuminuria with an SGLT2 (sodium-glucose cotransporter 2) inhibitor may be a positive indicator of long-term cardiovascular and renal benefits. We assessed changes in albuminuria during the first 12 weeks of treatment and subsequent long-term cardiovascular and renal risks associated with the SGLT2 inhibitor, empagliflozin, in the EMPA-REG OUTCOME (Empagliflozin Cardiovascular Outcome Event Trial in Type 2 diabetes Mellitus Patients) trial. Methods and Results We calculated the percentage urinary albumin creatinine ratio (UACR) change from baseline to week 12 in 6820 participants who did not experience a cardiovascular outcome (including 3-point major cardiovascular events and cardiovascular death or hospitalization for heart failure) or renal outcome (defined as 40% decline in estimated glomerular filtration rate from baseline, estimated glomerular filtration rate <15 mL/min per 1.73 m2, need for continuous renal-replacement therapy, or renal death) during the first 12 weeks. Multivariable Cox regression models were used to estimate the hazard ratio (HR) for each 30% reduction in UACR with outcomes. Empagliflozin reduced UACR by 18% (95% CI, 14-22) at week 12 compared with placebo, and increased the likelihood of a >30% reduction in UACR (odds ratio, 1.42; 95% CI, 1.27-1.58; P<0.001). During 3.0 years of follow-up, 704 major cardiovascular events, 440 cardiovascular deaths/hospitalizations for heart failure, and 168 renal outcomes were observed. Each 30% decrease in UACR during the first 12 weeks was statistically significantly associated with a lower hazard for major cardiovascular events (HR, 0.96; 95% CI, 0.93-0.99; P=0.012), cardiovascular deaths/hospitalizations for heart failure (HR, 0.94; 95% CI, 0.91-0.98; P=0.003), and renal outcomes (HR, 0.83; 95% CI, 0.78-0.89; P<0.001). Conclusions Short-term reduction in UACR was more common with empagliflozin and was statistically significantly associated with a decreased risk of long-term cardiovascular and renal outcomes. Registration URL: https://www.clinicaltrials.gov. Unique identifier: NCT01131676.

Keywords: cardiovascular outcomes; empagliflozin; kidney (diabetes); sodium‐glucose cotransporter 2 inhibitors.

Conflict of interest statement

Dr Inzucchi has participated on clinical trial executive/steering/publications committees and/or served as an advisor for Boehringer Ingelheim, AstraZeneca, Novo Nordisk, Sanofi/Lexicon, Abbott/Alere, and vTv Therapeutics. He has delivered lectures supported by Boehringer Ingelheim and Merck. Dr Zinman reports consultations and honoraria from AstraZeneca, Boehringer Ingelheim, Eli Lilly, Janssen, Merck, Novo Nordisk, and Sanofi. Dr Inker reports funding from National Institutes of Health, National Kidney Foundation, Retrophin, Omeros, Dialysis Clinics, Inc., and Reata Pharmaceuticals for research and contracts to Tufts Medical Center and consulting agreements with Tricida and Omeros Corp. Dr Wanner reports serving on advisory boards for Bayer, Boehringer Ingelheim, and Merck and received speaker's honoraria from Boehringer Ingelheim, Merck Sharp & Dohme, Eli Lilly, and AstraZeneca. Dr Koitka‐Weber and M. Mattheus are Boehringer Ingelheim company employees. Dr von Eynatten was a Boehringer Ingelheim employee at the time the analysis was conducted. Dr Heerspink is supported by a VIDI (917.15.306) grant from the Netherlands Organisation for Scientific Research and has served as a consultant for AbbVie, Astellas, AstraZeneca, Boehringer Ingelheim, Fresenius, Gilead, Janssen, Merck, Mundipharma, Mitsubishi‐Tanabe, and Retrophin and reports grants for research support from AbbVie, AstraZeneca, Boehringer Ingelheim, and Janssen. The remaining authors have no disclosures to report.

Figures

Figure 1. Relationship between baseline UACR and…
Figure 1. Relationship between baseline UACR and (A) major adverse cardiovascular event, (B) cardiovascular death/hospitalization for heart failure, and (C) renal outcome and the event rate of (D) major adverse cardiovascular event, (E) cardiovascular death/hospitalization for heart failure, and (F) renal outcome across the entire patient cohort.
The numbers above each circle (A through C) represent the number (percentage) of outcomes for each UACR category. The numbers above each bar represent the event rate (1000 patient×years). Cox regression models were adjusted for age, sex, smoking status, body mass index, systolic blood pressure, diastolic blood pressure, use of angiotensin‐converting enzyme inhibitor/angiotensin II receptor blocker, use of diuretics, treatment assignment (empagliflozin/placebo), region of residence, baseline glycated hemoglobin, estimated glomerular filtration rate, low‐density lipoprotein cholesterol, and high‐density lipoprotein cholesterol. UACR indicates urinary albumin creatinine ratio.
Figure 2. Relationship between change in UACR…
Figure 2. Relationship between change in UACR at week 12 and (A) major adverse cardiovascular event, (B) cardiovascular death/hospitalization for heart failure, and (C) renal outcome compared with the referent group (–30% to +30%).
The numbers above each circle represent the number (percentage) of outcomes for each change in UACR category. Cox regression models were adjusted for age, sex, smoking status, body mass index, baseline systolic and diastolic blood pressure, treatment assignment (empagliflozin/placebo), use of angiotensin‐converting enzyme inhibitor/angiotensin II receptor blocker, use of diuretics, region of residence, baseline UACR, glycated hemoglobin, estimated glomerular filtration rate, low‐density lipoprotein cholesterol and high‐density lipoprotein cholesterol, and percentage changes in estimated glomerular filtration rate, systolic blood pressure, glycated hemoglobin, and body weight at week 12. UACR indicates urinary albumin creatinine ratio.
Figure 3. Relationship between change in UACR…
Figure 3. Relationship between change in UACR at week 12 and (A) major adverse cardiovascular event, (B) cardiovascular death/hospitalization for heart failure, and (C) renal outcome in the placebo and empagliflozin groups.
Each point represents the median of each quartile change in albuminuria within the treatment group. Cox regression models were adjusted for age, sex, smoking status, body mass index, systolic blood pressure, diastolic blood pressure, use of angiotensin‐converting enzyme inhibitor/angiotensin II receptor blocker, use of diuretics, region of residence, baseline UACR, glycated hemoglobin, estimated glomerular filtration rate, low‐density lipoprotein cholesterol and high‐density lipoprotein cholesterol, percentage changes in estimated glomerular filtration rate, systolic blood pressure, glycated hemoglobin, and body weight at 12 weeks. UACR indicates urinary albumin creatinine ratio.
Figure 4. Adjusted HR for the association…
Figure 4. Adjusted HR for the association between >30% reduction in UACR from baseline to week 12 and cardiovascular and renal outcomes in all patients and within different subgroups.
Cox regression models were adjusted for age, sex, smoking status, body mass index, baseline systolic and diastolic blood pressure, treatment assignment (empagliflozin/placebo), use of ACEi/ARB, use of diuretics, region of residence, baseline UACR, glycated hemoglobin, eGFR, low‐density lipoprotein cholesterol and high‐density lipoprotein cholesterol, and percentage changes in eGFR, systolic blood pressure, glycated hemoglobin, and body weight at week 12. ACEi indicates angiotensin‐converting enzyme inhibitor; ARB, angiotensin II receptor blocker; CVD, cardiovascular death; eGFR, estimated glomerular filtration rate; HHF, hospitalization for heart failure; HR, hazard ratio; MACE, major adverse cardiovascular event; and UACR, urinary albumin creatinine ratio. *P value is the test of interaction between each subgroup.
Figure 5. Relationship between UACR at week…
Figure 5. Relationship between UACR at week 12 and (A) major adverse cardiovascular event, (B) cardiovascular death/hospitalization for heart failure, and (C) renal outcome, and the event rate of (D) major adverse cardiovascular event, (E) cardiovascular death/hospitalization for heart failure, and (F) renal outcome in both the empagliflozin and placebo groups.
The numbers above each circle (A through C) represent the number (percentage) of outcomes for each UACR category. The numbers above each bar represent the event rate (1000 patient×years). The <30 mg/g category in the placebo group was used as a reference for both the empagliflozin and placebo groups. Cox regression models were adjusted for age, sex, smoking status, body mass index, systolic blood pressure, diastolic blood pressure, use of angiotensin‐converting enzyme inhibitor/angiotensin II receptor blocker, use of diuretics, region of residence, baseline glycated hemoglobin, estimated glomerular filtration rate, low‐density lipoprotein cholesterol, and high‐density lipoprotein cholesterol. UACR indicates urinary albumin creatinine ratio.

References

    1. Emerging Risk Factors Collaboration , Di Angelantonio E, Kaptoge S, Wormser D, Willeit P, Butterworth AS, Bansal N, O’Keeffe LM, Gao P, Wood AM, Burgess S, et al. Association of cardiometabolic multimorbidity with mortality. JAMA. 2015;314:52–60.
    1. Kosiborod M, Gomes MB, Nicolucci A, Pocock S, Rathmann W, Shestakova MV, Watada H, Shimomura I, Chen H, Cid‐Ruzafa J, et al. Vascular complications in patients with type 2 diabetes: prevalence and associated factors in 38 countries (the DISCOVER study program). Cardiovasc Diabetol. 2018;17:150.
    1. Fox CS, Matsushita K, Woodward M, Bilo HJ, Chalmers J, Heerspink HJ, Lee BJ, Perkins RM, Rossing P, Sairenchi T, et al. Associations of kidney disease measures with mortality and end‐stage renal disease in individuals with and without diabetes: a meta‐analysis. Lancet. 2013;380:1662–1673.
    1. Ninomiya T, Perkovic V, de Galan BE, Zoungas S, Pillai A, Jardine M, Patel A, Cass A, Neal B, Poulter N, et al. Albuminuria and kidney function independently predict cardiovascular and renal outcomes in diabetes. J Am Soc Nephrol. 2009;20:1813–1821.
    1. Heerspink HJ, Ninomiya T, Persson F, Brenner BM, Brunel P, Chaturvedi N, Desai AS, Haffner SM, McMurray JJ, Solomon SD, et al. Is a reduction in albuminuria associated with renal and cardiovascular protection? A post hoc analysis of the ALTITUDE trial. Diabetes Obes Metab. 2016;18:169–177.
    1. Roscioni SS, Lambers Heerspink HJ, de Zeeuw D. Microalbuminuria: target for renoprotective therapy PRO. Kidney Int. 2014;86:40–49.
    1. de Zeeuw D, Remuzzi G, Parving HH, Keane WF, Zhang Z, Shahinfar S, Snapinn S, Cooper ME, Mitch WE, Brenner BM. Proteinuria, a target for renoprotection in patients with type 2 diabetic nephropathy: lessons from RENAAL. Kidney Int. 2004;65:2309–2320.
    1. Atkins RC, Briganti EM, Lewis JB, Hunsicker LG, Braden G, Champion de Crespigny PJ, DeFerrari G, Drury P, Locatelli F, Wiegmann TB, et al. Proteinuria reduction and progression to renal failure in patients with type 2 diabetes mellitus and overt nephropathy. Am J Kidney Dis. 2005;45:281–287.
    1. Heerspink HJL, Greene T, Tighiouart H, Gansevoort RT, Coresh J, Simon AL, Chan TM, Hou FF, Lewis JB, Locatelli F, et al. Change in albuminuria as a surrogate endpoint for progression of kidney disease: a meta‐analysis of treatment effects in randomised clinical trials. Lancet Diabetes Endocrinol. 2019;7:128–139.
    1. Coresh J, Heerspink HJL, Sang Y, Matsushita K, Arnlov J, Astor BC, Black C, Brunskill NJ, Carrero JJ, Feldman HI, et al. Change in albuminuria and subsequent risk of end‐stage kidney disease: an individual participant‐level consortium meta‐analysis of observational studies. Lancet Diabetes Endocrinol. 2019;7:115–127.
    1. Lea J, Greene T, Hebert L, Lipkowitz M, Massry S, Middleton J, Rostand SG, Miller E, Smith W, Bakris GL. The relationship between magnitude of proteinuria reduction and risk of end‐stage renal disease: results of the African American study of kidney disease and hypertension. Arch Intern Med. 2005;165:947–953.
    1. Ruggenenti P, Perna A, Remuzzi G. Retarding progression of chronic renal disease: the neglected issue of residual proteinuria. Kidney Int. 2003;63:2254–2261.
    1. van den Belt SM, Heerspink HJL, Gracchi V, de Zeeuw D, Wühl E, Schaefer F; ESCAPE Trial Group . Early proteinuria lowering by angiotensin‐converting enzyme inhibition predicts renal survival in children with CKD. J Am Soc Nephrol. 2018;29:2225–2233.
    1. Grempler R, Thomas L, Eckhardt M, Himmelsbach F, Sauer A, Sharp DE, Bakker RA, Mark M, Klein T, Eickelmann P. Empagliflozin, a novel selective sodium glucose cotransporter‐2 (SGLT‐2) inhibitor: characterisation and comparison with other SGLT‐2 inhibitors. Diabetes Obes Metab. 2012;14:83–90.
    1. Heise T, Seewaldt‐Becker E, Macha S, Hantel S, Pinnetti S, Seman L, Woerle HJ. Safety, tolerability, pharmacokinetics and pharmacodynamics following 4 weeks' treatment with empagliflozin once daily in patients with type 2 diabetes. Diabetes Obes Metab. 2013;15:613–621.
    1. Barnett AH, Mithal A, Manassie J, Jones R, Rattunde H, Woerle HJ, Broedl UC. Efficacy and safety of empagliflozin added to existing antidiabetes treatment in patients with type 2 diabetes and chronic kidney disease: a randomised, double‐blind, placebo‐controlled trial. Lancet Diabetes Endocrinol. 2013;2:369–384.
    1. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128.
    1. Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, Johansen OE, Woerle HJ, Broedl UC, Zinman B, et al. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med. 2016;375:323–334.
    1. Zinman B, Inzucchi SE, Lachin JM, Wanner C, Ferrari R, Fitchett D, Bluhmki E, Hantel S, Kempthorne‐Rawson J, Newman J, et al. Rationale, design, and baseline characteristics of a randomized, placebo‐controlled cardiovascular outcome trial of empagliflozin (EMPA‐REG OUTCOME). Cardiovasc Diabetol. 2014;13:102.
    1. Levey AS, Bosch JP, Lewis JB, Greene T, Rogers N, Roth D. A more accurate method to estimate glomerular filtration rate from serum creatinine: a new prediction equation. Modification of Diet in Renal Disease Study Group. Ann Intern Med. 1999;130:461–470.
    1. Cherney DZI, Zinman B, Inzucchi SE, Koitka‐Weber A, Mattheus M, von Eynatten M, Wanner C. Effects of empagliflozin on the urinary albumin‐to‐creatinine ratio in patients with type 2 diabetes and established cardiovascular disease: an exploratory analysis from the EMPA‐REG OUTCOME randomised, placebo‐controlled trial. Lancet Diabetes Endocrinol. 2017;5:610–621.
    1. Levey AS, Stevens LA, Schmid CH, Zhang YL, Castro AF III, Feldman HI, Kusek JW, Eggers P, Van Lente F, Greene T, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150:604–612.
    1. Inker LA, Lambers Heerspink HJ, Mondal H, Schmid CH, Tighiouart H, Noubary F, Coresh J, Greene T, Levey AS. GFR decline as an alternative end point to kidney failure in clinical trials: a meta‐analysis of treatment effects from 37 randomized trials. Am J Kidney Dis. 2014;64:848–859.
    1. Herrington WG, Preiss D, Haynes R, von Eynatten M, Staplin N, Hauske SJ, George JT, Green JB, Landray MJ, Baigent C, et al. The potential for improving cardio‐renal outcomes by sodium‐glucose co‐transporter‐2 inhibition in people with chronic kidney disease: a rationale for the EMPA‐KIDNEY study. Clin Kidney J. 2018;11:749–761.
    1. MacMahon S, Peto R, Cutler J, Collins R, Sorlie P, Neaton J, Abbott R, Godwin J, Dyer A, Stamler J. Blood pressure, stroke, and coronary heart disease. Part 1, prolonged differences in blood pressure: prospective observational studies corrected for the regression dilution bias. Lancet. 1990;335:765–774.
    1. Scirica BM, Mosenzon O, Bhatt DL, Udell JA, Steg PG, McGuire DK, Im K, Kanevsky E, Stahre C, Sjostrand M, et al. Cardiovascular outcomes according to urinary albumin and kidney disease in patients with type 2 diabetes at high cardiovascular risk: observations from the SAVOR‐TIMI 53 trial. JAMA Cardiol. 2018;3:155–163.
    1. Brouwers FP, de Boer RA, van der Harst P, Voors AA, Gansevoort RT, Bakker SJ, Hillege HL, van Veldhuisen DJ, van Gilst WH. Incidence and epidemiology of new onset heart failure with preserved vs. reduced ejection fraction in a community‐based cohort: 11‐year follow‐up of PREVEND. Eur Heart J. 2013;34:1424–1431.
    1. Bailey LN, Levitan EB, Judd SE, Sterling MR, Goyal P, Cushman M, Safford MM, Gutierrez OM. Association of urine albumin excretion with incident heart failure hospitalization in community‐dwelling adults. JACC Heart Fail. 2019;7:394–401.
    1. Litvin CB, Hyer JM, Ornstein SM. Use of clinical decision support to improve primary care identification and management of chronic kidney disease (CKD). J Am Board Fam Med. 2016;29:604–612.
    1. Perkins RM, Chang AR, Wood KE, Coresh J, Matsushita K, Grams M. Incident chronic kidney disease: trends in management and outcomes. Clin Kidney J. 2016;9:432–437.
    1. Peralta CA, Frigaard M, Rubinsky AD, Rolon L, Lo L, Voora S, Seal K, Tuot D, Chao S, Lui K, et al. Implementation of a pragmatic randomized trial of screening for chronic kidney disease to improve care among non‐diabetic hypertensive veterans. BMC Nephrol. 2017;18:132.
    1. Stevens PE, O'Donoghue DJ, de Lusignan S, Van Vlymen J, Klebe B, Middleton R, Hague N, New J, Farmer CK. Chronic kidney disease management in the United Kingdom: NEOERICA project results. Kidney Int. 2007;72:92–99.
    1. Witte EC, Lambers Heerspink HJ, de Zeeuw D, Bakker SJ, de Jong PE, Gansevoort R. First morning voids are more reliable than spot urine samples to assess microalbuminuria. J Am Soc Nephrol. 2009;20:436–443.
    1. Selvin E, Juraschek SP, Eckfeldt J, Levey AS, Inker LA, Coresh J. Within‐person variability in kidney measures. Am J Kidney Dis. 2013;61:716–722.
    1. Jun M, Ohkuma T, Zoungas S, Colagiuri S, Mancia G, Marre M, Matthews D, Poulter N, Williams B, Rodgers A, et al. Changes in albuminuria and the risk of major clinical outcomes in diabetes: results from ADVANCE‐ON. Diabetes Care. 2018;41:163–170.
    1. Smith M, Herrington WG, Weldegiorgis M, Hobbs FR, Bankhead C, Woodward M. Change in albuminuria and risk of renal and cardiovascular outcomes: natural variation should be taken into account. Kidney Int Rep. 2018;3:939–949.
    1. Kröpelin TF, de Zeeuw D, Andress DL, Bijlsma MJ, Persson F, Parving H‐H, Heerspink HJL. Number and frequency of albuminuria measurements in clinical trials in diabetic nephropathy. Clin J Am Soc Nephrol. 2015;10:410–416.
    1. Perkovic V, de Zeeuw D, Mahaffey KW, Fulcher G, Erondu N, Shaw W, Barrett TD, Weidner‐Wells M, Deng H, Matthews DR, et al. Canagliflozin and renal outcomes in type 2 diabetes: results from the CANVAS Program randomised clinical trials. Lancet Diabetes Endocrinol. 2018;6:691–704.
    1. van Raalte DH, Bjornstad P, Persson F, Powell DR, de Cassia CR, Wang PS, Liu M, Heerspink HJL, Cherney D. The impact of sotagliflozin on renal function, albuminuria, blood pressure, and hematocrit in adults with type 1 diabetes. Diabetes Care. 2019;42:1921–1929.
    1. Heerspink HJL, Perkins BA, Fitchett DH, Husain M, Cherney DZI. Sodium glucose cotransporter 2 inhibitors in the treatment of diabetes mellitus: cardiovascular and kidney effects, potential mechanisms, and clinical applications. Circulation. 2016;134:752–772.
    1. Dekkers CCJ, Gansevoort RT, Heerspink HJL. New diabetes therapies and diabetic kidney disease progression: the role of SGLT‐2 inhibitors. Curr Diab Rep. 2018;18:27.
    1. Martens P, Mathieu C, Verbrugge FH. Promise of SGLT2 inhibitors in heart failure: diabetes and beyond. Curr Treat Options Cardiovasc Med. 2017;19:23.

Source: PubMed

3
Sottoscrivi