The Effect of Dapagliflozin on Albuminuria in DECLARE-TIMI 58

Ofri Mosenzon, Stephen D Wiviott, Hiddo J L Heerspink, Jamie P Dwyer, Avivit Cahn, Erica L Goodrich, Aliza Rozenberg, Meir Schechter, Ilan Yanuv, Sabina A Murphy, Thomas A Zelniker, Ingrid A M Gause-Nilsson, Anna Maria Langkilde, Martin Fredriksson, Peter A Johansson, Deepak L Bhatt, Lawrence A Leiter, Darren K McGuire, John P H Wilding, Marc S Sabatine, Itamar Raz, Ofri Mosenzon, Stephen D Wiviott, Hiddo J L Heerspink, Jamie P Dwyer, Avivit Cahn, Erica L Goodrich, Aliza Rozenberg, Meir Schechter, Ilan Yanuv, Sabina A Murphy, Thomas A Zelniker, Ingrid A M Gause-Nilsson, Anna Maria Langkilde, Martin Fredriksson, Peter A Johansson, Deepak L Bhatt, Lawrence A Leiter, Darren K McGuire, John P H Wilding, Marc S Sabatine, Itamar Raz

Abstract

Objective: Sodium-glucose cotransporter 2 inhibitors (SGLT2i) improve albuminuria in patients with high cardiorenal risk. We report albuminuria change in the Dapagliflozin Effect on Cardiovascular Events (DECLARE-TIMI 58) cardiovascular outcome trial, which included populations with lower cardiorenal risk.

Research design and methods: DECLARE-TIMI 58 randomized 17,160 patients with type 2 diabetes, creatinine clearance >60 mL/min, and either atherosclerotic cardiovascular disease (CVD; 40.6%) or risk-factors for CVD (59.4%) to dapagliflozin or placebo. Urinary albumin-to-creatinine ratio (UACR) was tested at baseline, 6 months, 12 months, and yearly thereafter. The change in UACR over time was measured as a continuous and categorical variable (≤15, >15 to <30, ≥30 to ≤300, and >300 mg/g) by treatment arm. The composite cardiorenal outcome was a ≥40% sustained decline in the estimated glomerular filtration rate (eGFR) to <60 mL/min/1.73 m2, end-stage kidney disease, and cardiovascular or renal death; specific renal outcome included all except cardiovascular death.

Results: Baseline UACR was available for 16,843 (98.15%) participants: 9,067 (53.83%) with ≤15 mg/g, 2,577 (15.30%) with >15 to <30 mg/g, 4,030 (23.93%) with 30-300 mg/g, and 1,169 (6.94%) with >300 mg/g. Measured as a continuous variable, UACR improved from baseline to 4.0 years with dapagliflozin, compared with placebo, across all UACR and eGFR categories (all P < 0.0001). Sustained confirmed ≥1 category improvement in UACR was more common in dapagliflozin versus placebo (hazard ratio 1.45 [95% CI 1.35-1.56], P < 0.0001). Cardiorenal outcome was reduced with dapagliflozin for subgroups of UACR ≥30 mg/g (P < 0.0125, P interaction = 0.033), and the renal-specific outcome was reduced for all UACR subgroups (P < 0.05, P interaction = 0.480).

Conclusions: In DECLARE-TIMI 58, dapagliflozin demonstrated a favorable effect on UACR and renal-specific outcome across baseline UACR categories, including patients with normal albumin excretion. The results suggest a role for SGLT2i also in the primary prevention of diabetic kidney disease.

Trial registration: ClinicalTrials.gov NCT01730534.

© 2021 by the American Diabetes Association.

Figures

Figure 1
Figure 1
Change in UACR over time by treatment arm at baseline, 6 months, and 1, 2, 3, and 4 years in the group of patients with baseline UACR ≤15 mg/g (A), baseline UACR >15 to <30 mg/g (B), baseline UACR ≥30 to ≤300 mg/g (C) and baseline UACR >300 mg/g (D), and in the group of patients with baseline eGFR ≥90 mL/min/1.73 m2 (E), baseline eGFR <90 to ≥60 mL/min/1.73 m2 (F), and baseline eGFR <60 mL/min/1.73 m2 (G). Shown are point estimates and 95% confidence intervals of geometric mean back-transformed to the original scale.
Figure 1
Figure 1
Change in UACR over time by treatment arm at baseline, 6 months, and 1, 2, 3, and 4 years in the group of patients with baseline UACR ≤15 mg/g (A), baseline UACR >15 to <30 mg/g (B), baseline UACR ≥30 to ≤300 mg/g (C) and baseline UACR >300 mg/g (D), and in the group of patients with baseline eGFR ≥90 mL/min/1.73 m2 (E), baseline eGFR <90 to ≥60 mL/min/1.73 m2 (F), and baseline eGFR <60 mL/min/1.73 m2 (G). Shown are point estimates and 95% confidence intervals of geometric mean back-transformed to the original scale.
Figure 2
Figure 2
Change in confirmed sustained categorical UACR (mg/g) from baseline (BL) to EOT in dapagliflozin vs. placebo arm. A: Improvement in UACR categories. B: Deterioration in UACR categories.
Figure 3
Figure 3
Treatment effect of dapagliflozin vs. placebo on composite cardiorenal and renal-specific outcomes according to baseline UACR categories of ≤15, >15 to 300 mg/g. Cox model with stratification factor (baseline hematuria status and eASCVD or MRF status). KM, Kaplan-Meier.

References

    1. Wanner C, Inzucchi SE, Lachin JM, et al. .; EMPA-REG OUTCOME Investigators . Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 2016;375:323–334
    1. Neal B, Perkovic V, Mahaffey KW, et al. .; CANVAS Program Collaborative Group . Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 2017;377:644–657
    1. Wiviott SD, Raz I, Bonaca MP, et al. .; DECLARE–TIMI 58 Investigators . Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2019;380:347–357
    1. Mosenzon O, Wiviott SD, Cahn A, et al. . Effects of dapagliflozin on development and progression of kidney disease in patients with type 2 diabetes: an analysis from the DECLARE-TIMI 58 randomised trial. Lancet Diabetes Endocrinol 2019;7:606–617
    1. Perkovic V, Jardine MJ, Neal B, et al. .; CREDENCE Trial Investigators . Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 2019;380:2295–2306
    1. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. .; DAPA-CKD Trial Committees and Investigators . Dapagliflozin in patients with chronic kidney disease. N Engl J Med 2020;383:1436–1446
    1. Cannon CP, Pratley R, Dagogo-Jack S, et al. .; VERTIS CV Investigators . Cardiovascular outcomes with ertugliflozin in type 2 diabetes. N Engl J Med 2020;383:1425–1435
    1. Tuttle KR, Bakris GL, Bilous RW, et al. . Diabetic kidney disease: a report from an ADA Consensus Conference. Diabetes Care 2014;37:2864–2883
    1. Yamanouchi M, Furuichi K, Hoshino J, et al. .; Research Group of Diabetic Nephropathy, the Ministry of Health, Labour and Welfare, and the Japan Agency for Medical Research and Development . Nonproteinuric versus proteinuric phenotypes in diabetic kidney disease: a propensity score-matched analysis of a nationwide, biopsy-based cohort study. Diabetes Care 2019;42:891–902
    1. Chronic Kidney Disease Prognosis Consortium, Matsushita K, van der Velde M, Astor BC, et al. . Association of estimated glomerular filtration rate and albuminuria with all-cause and cardiovascular mortality in general population cohorts: a collaborative meta-analysis. Lancet 2010;375:2073–2081
    1. van der Velde M, Matsushita K, Coresh J, et al. .; Chronic Kidney Disease Prognosis Consortium . Lower estimated glomerular filtration rate and higher albuminuria are associated with all-cause and cardiovascular mortality. A collaborative meta-analysis of high-risk population cohorts. Kidney Int 2011;79:1341–1352
    1. Coresh J, Heerspink HJL, Sang Y, et al. .; Chronic Kidney Disease Prognosis Consortium and Chronic Kidney Disease Epidemiology Collaboration . 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. Heerspink HJL, Greene T, Tighiouart H, et al. .; Chronic Kidney Disease Epidemiology Collaboration . 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. Schmieder RE, Mann JFE, Schumacher H, et al. .; ONTARGET Investigators . Changes in albuminuria predict mortality and morbidity in patients with vascular disease. J Am Soc Nephrol 2011;22:1353–1364
    1. American Diabetes Association . 11. Microvascular complications and foot care: Standards of Medical Care in Diabetes—2021. Diabetes Care 2021;44(Suppl. 1):S151–S167
    1. Wiviott SD, Raz I, Bonaca MP, et al. . The design and rationale for the Dapagliflozin Effect on Cardiovascular Events (DECLARE)-TIMI 58 Trial. Am Heart J 2018;200:83–89
    1. Raz I, Mosenzon O, Bonaca MP, et al. . DECLARE-TIMI 58: participants’ baseline characteristics. Diabetes Obes Metab 2018;20:1102–1110
    1. Schwandt A, Denkinger M, Fasching P, et al. . Comparison of MDRD, CKD-EPI, and Cockcroft-Gault equation in relation to measured glomerular filtration rate among a large cohort with diabetes. J Diabetes Complications 2017;31:1376–1383
    1. Levey AS, de Jong PE, Coresh J, et al. . The definition, classification, and prognosis of chronic kidney disease: a KDIGO Controversies Conference report. Kidney Int 2011;80:17–28
    1. Cherney DZI, Zinman B, Inzucchi SE, et al. . 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. Perkovic V, de Zeeuw D, Mahaffey KW, 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. Heerspink HJL, Johnsson E, Gause-Nilsson I, Cain VA, Sjöström CD. Dapagliflozin reduces albuminuria in patients with diabetes and hypertension receiving renin-angiotensin blockers. Diabetes Obes Metab 2016;18:590–597
    1. Fioretto P, Stefansson BV, Johnsson E, Cain VA, Sjöström CD. Dapagliflozin reduces albuminuria over 2 years in patients with type 2 diabetes mellitus and renal impairment. Diabetologia 2016;59:2036–2039
    1. Cherney D, Lund SS, Perkins BA, et al. . The effect of sodium glucose cotransporter 2 inhibition with empagliflozin on microalbuminuria and macroalbuminuria in patients with type 2 diabetes. Diabetologia 2016;59:1860–1870
    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. Tonneijck L, Muskiet MHA, Smits MM, et al. . Glomerular hyperfiltration in diabetes: mechanisms, clinical significance, and treatment. J Am Soc Nephrol 2017;28:1023–1039
    1. van Bommel EJM, Muskiet MHA, van Baar MJB, et al. . The renal hemodynamic effects of the SGLT2 inhibitor dapagliflozin are caused by post-glomerular vasodilatation rather than pre-glomerular vasoconstriction in metformin-treated patients with type 2 diabetes in the randomized, double-blind RED trial. Kidney Int 2020;97:202–212
    1. Lytvyn Y, Bjornstad P, van Raalte DH, Heerspink HL, Cherney DZI. The new biology of diabetic kidney disease—mechanisms and therapeutic implications. Endocr Rev 2020;41:202–231
    1. Vallon V, Thomson SC. Targeting renal glucose reabsorption to treat hyperglycaemia: the pleiotropic effects of SGLT2 inhibition. Diabetologia 2017;60:215–225
    1. Tsimihodimos V, Filippatos TD, Elisaf MS. SGLT2 inhibitors and the kidney: effects and mechanisms. Diabetes Metab Syndr 2018;12:1117–1123
    1. O’Neill J, Fasching A, Pihl L, Patinha D, Franzén S, Palm F. Acute SGLT inhibition normalizes O2 tension in the renal cortex but causes hypoxia in the renal medulla in anaesthetized control and diabetic rats. Am J Physiol Renal Physiol 2015;309:F227–F234
    1. Parving HH, Lewis JB, Ravid M, Remuzzi G; DEMAND investigators . Prevalence and risk factors for microalbuminuria in a referred cohort of type II diabetic patients: a global perspective. Kidney Int 2006;69:2057–2063
    1. Young BA, Katon WJ, Von Korff M, et al. . Racial and ethnic differences in microalbuminuria prevalence in a diabetes population: the pathways study. J Am Soc Nephrol 2005;16:219–228
    1. Newman DJ, Mattock MB, Dawnay ABS, et al. . Systematic review on urine albumin testing for early detection of diabetic complications. Health Technol Assess 2005;9:iii–vi, xiii-163
    1. Scirica BM, Mosenzon O, Bhatt DL, 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. Blecker S, Matsushita K, Köttgen A, et al. . High-normal albuminuria and risk of heart failure in the community. Am J Kidney Dis 2011;58:47–55
    1. Astor BC, Matsushita K, Gansevoort RT, et al. .; Chronic Kidney Disease Prognosis Consortium . Lower estimated glomerular filtration rate and higher albuminuria are associated with mortality and end-stage renal disease. A collaborative meta-analysis of kidney disease population cohorts. Kidney Int 2011;79:1331–1340
    1. Kröpelin TF, de Zeeuw D, Andress DL, et al. . Number and frequency of albuminuria measure ments in clinical trials in diabetic nephropathy. Clin J Am Soc Nephrol 2015;10:410–416
    1. Kröpelin TF, de Zeeuw D, Remuzzi G, Bilous R, Parving H-H, Heerspink HJL. Determining the optimal protocol for measuring an albuminuria class transition in clinical trials in diabetic kidney disease. J Am Soc Nephrol 2016;27:3405–3412
    1. EMPA-KIDNEY (The Study of Heart and Kidney Protection With Empagliflozin). . Accessed 9 May 2020. Available from
    1. Vart P, Powe NR, McCulloch CE, et al. .; Centers for Disease Control and Prevention Chronic Kidney Disease Surveillance Team . National trends in the prevalence of chronic kidney disease among racial/ethnic and socioeconomic status groups, 1988-2016. JAMA Netw Open 2020;3:e207932.

Source: PubMed

3
Se inscrever