Liraglutide and Renal Outcomes in Type 2 Diabetes

Similar documents
Supplementary Appendix

CANVAS Program Independent commentary

MOA: Long acting glucagon-like peptide 1 receptor agonist

LEADER Liraglutide and cardiovascular outcomes in type 2 diabetes

Gli endpoint micro-vascolari nei trial di outcome cardiovascolare

HEART FAILURE AND DIABETES MELLITUS: DANGEROUS LIASONS MICHEL KOMAJDA, MD

PROTEZIONE DAL DANNO RENALE NEL DIABETE TIPO 2: RUOLO DEI NUOVI FARMACI. Massimo Boemi UOC Malattie Metaboliche e Diabetologia IRCCS INRCA Ancona

Can We Reduce Heart Failure by Treating Diabetes? CVOT Data on SGLT2 Inhibitors and GLP-1Receptor Agonists

No Increased Cardiovascular Risk for Lixisenatide in ELIXA

Update on Diabetes Cardiovascular Outcome Trials

1. Albuminuria an early sign of glomerular damage and renal disease. albuminuria

Diabete: terapia nei pazienti a rischio cardiovascolare

Soo LIM, MD, PHD Internal Medicine Seoul National University Bundang Hospital

Cardiologists and HbA1c: Novel Diabetes Drugs and Cardiovascular Disease Outcomes

Medical therapy advances London/Manchester RCP February/June 2016

Cardiovascular Benefits of Two Classes of Antihyperglycemic Medications

Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes

Current principles of diabetes management

egfr > 50 (n = 13,916)

LATE BREAKING STUDIES IN DM AND CAD. Will this change the guidelines?

Management of Type 2 Diabetes Cardiovascular Outcomes Trials Tom Blevins MD Texas Diabetes and Endocrinology Austin, Texas

Top HF Trials to Impact Your Practice

ADVANCE post trial ObservatioNal Study

Cardiovascular outcome studies with glucagon-like peptide 1 receptor agonists what will REWIND add?

Alogliptin after Acute Coronary Syndrome in Patients with Type 2 Diabetes

Prevention And Treatment of Diabetic Nephropathy. MOH Clinical Practice Guidelines 3/2006 Dr Stephen Chew Tec Huan

SGLT2 inhibition in diabetes: extending from glycaemic control to renal and cardiovascular protection

Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes

Diabetes and Heart Failure: The Role of SGLT2 Inhibitors

Empagliflozin (Jardiance ) for the treatment of type 2 diabetes mellitus, the EMPA REG OUTCOME study

Clinical Relevance of Blood Pressure Lowering Effect of Modern Antidiabetic Drugs

Diabetes new challenges, new agents, new order

Variability in drug response: towards more personalized diabetes care Petrykiv, Sergei

TREATMENT OF DIABETES AFTER METFORMIN GREGG GERETY, MD ALBANY MEDICAL COLLEGE, DIVISION OF COMMUNITY ENDOCRINOLOGY JULY 14, 2017

Drug Class Monograph

Help the Heart. An Update on GLP-1 Agonists and SGLT2 Inhibitors. Tara Hawley, PharmD PGY1 Pharmacy Resident Mayo Clinic Health System Eau Claire

Liraglutide and Glycaemic Outcomes in the LEADER Trial

GLP 1 agonists Winning the Losing Battle. Dr Bernard SAMIA. KCS Congress: Impact through collaboration

Effect of Sitagliptin on Cardiovascular Outcomes in Type 2 Diabetes

CARDIOVASCULAR RISK FACTOR CONTROL IN TYPE 2 DIABETES MELLITUS AND NEW TRIAL EVIDENCE

Risk Factors, Mortality, and Cardiovascular Outcomes in Patients with Type 2 Diabetes

01/09/2017. Outline. SGLT 2 inhibitor? Diabetes Patients: Complex and Heterogeneous. Association between diabetes and cardiovascular events

Drug Class Monograph

Bariatric Surgery versus Intensive Medical Therapy for Diabetes 3-Year Outcomes

GUIDANCE UPDATE TYPE 2 DIABETES AN OVERVIEW OF THE SIGN GUIDELINE ON PHARMACOLOGICAL MANAGEMENT OF GLYCAEMIC CONTROL.

abstract n engl j med 375;4 nejm.org July 28,

Intensive Diabetes Therapy and Glomerular Filtration Rate in Type 1 Diabetes

Update on Cardiovascular Outcome Trials in Diabetes Jay S. Skyler, MD, MACP

Educational Objectives

Effects of Intensive Blood-Pressure Control in Type 2 Diabetes Mellitus

2782 Diabetes Care Volume 37, October 2014

SMJ Singapore Medical Journal

Endocrinologist Sweetgrass Endocrinology

Updates in Diabetes and Cardiovascular Disease Management: Are You Making the Link?

Diabetic Nephropathy. Objectives:

Update on Cardiovascular Outcome Trials in Diabetes. Rury R. Holman, FMedSci NIHR Senior Investigator 11 th February 2013

Cardiovascular Outcomes With Newer Diabetes Drugs: Results From The EMPA-REG and LEADER Trials

Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes

Type 2 diabetes affects an estimated 25.8 million

The EMPA-REG OUTCOME trial: Design and results. David Fitchett, MD University of Toronto, Canada

Dapagliflozin and cardiovascular outcomes in type 2

CV outcomes Studies and Implications for diabetes management. Seraj Abualnaja, MD, FRCPC Consultant Interventional cardiologist DSFH

Effect of SGLT-2 Inhibitors on the Heart. Robert Zimmerman MD Vice Chairman Endocrinology Director Diabetes Center Cleveland Clinic

Can Treating Diabetes with SGLT2 inhibitors Prevent Heart Failure?

Disclosures. Objectives. Bryan Cardiology Conference DM2 & Cardiovascular Outcome Trials 8/28/2017

Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes

Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes

sitagliptin, 25mg, 50mg and 100mg film-coated tablets (Januvia ) SMC No. (1083/15) Merck Sharp and Dohme UK Ltd

Diabetes and kidney disease.

The Role of Vildagliptin in the Therapy of Type 2 Diabetic Patients with Renal Dysfunction

This clinical study synopsis is provided in line with Boehringer Ingelheim s Policy on Transparency and Publication of Clinical Study Data.

The CARI Guidelines Caring for Australasians with Renal Impairment. Protein Restriction to prevent the progression of diabetic nephropathy GUIDELINES

Follow-up of Blood-Pressure Lowering and Glucose Control in Type 2 Diabetes

Aggressive blood pressure reduction and renin angiotensin system blockade in chronic kidney disease: time for re-evaluation?

DPP-4 inhibitor use and risk of diabetic retinopathy: a new safety issue of a safe drug Nam Hoon Kim

Microvascular Disease in Type 1 Diabetes

ESC GUIDELINES ON DIABETES AND CARDIOVASCULAR DISEASES

KDIGO Controversies Conference on Management of Patients with Diabetes and Chronic Kidney Disease

Glycemic Control Patterns and Kidney Disease Progression among Primary Care Patients with Diabetes Mellitus

Cedars Sinai Diabetes. Michael A. Weber

Multi-factor approach to reduce cardiovascular risk in diabetes

Case Studies in Type 2 Diabetes Mellitus: Focus on Cardiovascular Outcomes Trials

Obesity, Insulin Resistance, Metabolic Syndrome, and the Natural History of Type 2 Diabetes

The Seventh Report of the Joint National Commission

OBJECTIVE RESEARCH DESIGN AND METHODS RESULTS CONCLUSIONS. Diabetes Care Volume 39, July CARDIOVASCULAR AND METABOLIC RISK

Supplementary Online Content

Diabetic Kidney Disease Tripti Singh MD Department of Nephrology University of Wisconsin

Oral Hypoglycemics and Risk of Adverse Cardiac Events: A Summary of the Controversy

Differentiating Among the SGLT-2 Inhibitors: Considering Cardiovascular and Other Safety Outcomes

Hot Topics in Diabetic Kidney Disease a primary care perspective

Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes

ALLHAT RENAL DISEASE OUTCOMES IN HYPERTENSIVE PATIENTS STRATIFIED INTO 4 GROUPS BY BASELINE GLOMERULAR FILTRATION RATE (GFR)

Canagliflozin in combination therapy for treating type 2 diabetes

NATIONAL INSTITUTE FOR HEALTH AND CARE EXCELLENCE. Single Technology Appraisal. Canagliflozin in combination therapy for treating type 2 diabetes

Supplementary Appendix

Cardiovascular outcome trials in diabetes: what have we learnt?

Empagliflozin: Role in Treatment Options for Patients with Type 2 Diabetes Mellitus

ABSTRACT ORIGINAL RESEARCH. Elizabeth S. Ommen Lei Xu Edward A. O Neill Barry J. Goldstein Keith D. Kaufman Samuel S. Engel

SGLT2 and cardiovascular events: Why did EMPA-REG Outcomes surprise and what were the likely mechanisms? David Preiss, 3 John J McMurray 1

KDIGO conference on high CV risk associated with CKD. The role of BP in CKD stage 1-4

Transcription:

The new england journal of medicine Original Article and Renal Outcomes in Type 2 Diabetes Johannes F.E. Mann, M.D., David D. Ørsted, M.D., Ph.D., Kirstine Brown Frandsen, M.D., Steven P. Marso, M.D., Neil R. Poulter, F.Med.Sci., Søren Rasmussen, Ph.D., Karen Tornøe, M.D., Ph.D., Bernard Zinman, M.D., and John B. Buse, M.D., Ph.D., for the LEADER Steering Committee and Investigators* ABSTRACT BACKGROUND In a randomized, controlled trial that compared liraglutide, a glucagon-like peptide 1 analogue, with placebo in patients with type 2 diabetes and high cardiovascular risk who were receiving usual care, we found that liraglutide resulted in lower risks of the primary end point (nonfatal myocardial infarction, nonfatal stroke, or death from cardiovascular causes) and death. However, the long-term effects of liraglutide on renal outcomes in patients with type 2 diabetes are unknown. METHODS We report the prespecified secondary renal outcomes of that randomized, controlled trial in which patients were assigned to receive liraglutide or placebo. The secondary renal outcome was a composite of new-onset persistent macroalbuminuria, persistent doubling of the serum creatinine level, end-stage renal disease, or death due to renal disease. The risk of renal outcomes was determined with the use of time-to-event analyses with an intention-to-treat approach. Changes in the estimated glomerular filtration rate and albuminuria were also analyzed. RESULTS A total of 934 patients underwent randomization, and the median follow-up of the patients was 3.84 years. The renal outcome occurred in fewer participants in the liraglutide group than in the placebo group (268 of 4668 patients vs. 337 of 4672; hazard ratio,.78; 95% confidence interval [CI],.67 to.92; P =.3). This result was driven primarily by the new onset of persistent macroalbuminuria, which occurred in fewer participants in the liraglutide group than in the placebo group (161 vs. 215 patients; hazard ratio,.74; 95% CI,.6 to.91; P =.4). The rates of renal adverse events were similar in the liraglutide group and the placebo group (15.1 events and 16.5 events per 1 patient-years), including the rate of acute kidney injury (7.1 and 6.2 events per 1 patient-years, respectively). From KfH Kidney Center, Munich, and Friedrich Alexander University of Erlangen, Erlangen both in Germany (J.F.E.M.); Novo Nordisk, Bagsvaerd, Denmark (D.D.Ø., K.B.-F., S.R., K.T.); University of Texas Southwestern Medical Center, Dallas (S.P.M.); Imperial College London, London (N.R.P.); Lunenfeld Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto (B.Z.); and University of North Carolina School of Medicine, Chapel Hill (J.B.B.). Address reprint requests to Dr. Mann at the KfH Kidney Center, 15 Isoldenstr., Munich 884, Germany, or at prof. j. mann@ gmail. com. * A complete list of the steering committee and investigators in the Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial is provided in the Supplementary Appendix, available at NEJM.org. N Engl J Med 217;377:839-48. DOI: 1.156/NEJMoa161611 Copyright 217 Massachusetts Medical Society. CONCLUSIONS This prespecified secondary analysis shows that, when added to usual care, liraglutide resulted in lower rates of the development and progression of diabetic kidney disease than placebo. (Funded by Novo Nordisk and the National Institutes of Health; LEADER ClinicalTrials.gov number, NCT117948.) n engl j med 377;9 nejm.org August 31, 217 839

The new england journal of medicine Persons with type 2 diabetes mellitus, particularly those with evidence of cardiovascular disease, are at high risk for chronic kidney disease. 1,2 In most countries, diabetic kidney disease is reported to be the leading cause of dialysis-dependent chronic kidney disease. 3 Efforts to blunt the global increase in the prevalence of end-stage renal disease therefore include the prevention of new-onset diabetic nephropathy and the prevention of progression of established diabetic nephropathy. Improved glycemic control has been shown to reduce the occurrence of early diabetic nephropathy. 4-7 In patients with established diabetic nephropathy, blood-pressure control and inhibition of the renin angiotensin system are pivotal to slowing a progressive reduction in the glomerular filtration rate. 8 Some glucose-lowering agents appear to have glucose-independent effects on diabetic nephropathy and its progression. Data from studies investigating the dipeptidyl peptidase 4 (DPP-4) inhibitor linagliptin and the human glucagon-like peptide 1 (GLP-1) analogue liraglutide, as well as sodium glucose cotransporter 2 (SGLT2) inhibitors, suggest that these drugs exert beneficial effects in experimental models of kidney injury. 9-11 However, in long-term studies, no clinically meaningful benefits on renal outcomes have emerged with these medications as compared with placebo, except for a convincing benefit among patients who have received the SGLT2 inhibitor empagliflozin. 11 In short-term studies, liraglutide improved glycemic control, reduced urinary albumin excretion, and increased urinary sodium excretion. 12,13 However, the long-term effects of liraglutide on the development and progression of diabetic kidney disease are unknown. The Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial recently showed lower risks of cardiovascular outcomes, death from any cause, and microvascular outcomes with liraglutide than with placebo. 14 The lower risk of microvascular outcomes was driven by renal outcomes. Therefore, we report here the results of a prespecified analysis of the effects of liraglutide on renal outcomes. Methods Trial Design and Oversight The trial design and methods have been published previously. 15 The trial protocol, available with the full text of this article at NEJM.org, was approved by the institutional review board or ethics committee at each participating center, and all the patients provided written informed consent. In brief, in this double-blind, placebocontrolled trial, patients with type 2 diabetes and a high risk of cardiovascular disease were randomly assigned, in a 1:1 ratio, to receive liraglutide or matching placebo, in addition to usual care. The disposition of the trial participants has been published previously. 15 Given the regulatory request to provide data on the use of liraglutide in patients with stage 4 chronic kidney disease (estimated glomerular filtration rate [GFR], 15 to <3 ml per minute per 1.73 m 2 of body-surface area), approximately 22 such patients were included in the trial. The minimum planned follow-up of the patients receiving the trial regimen was 42 months, and the maximum planned followup was 6 months, with an additional 3 days of follow-up after the trial regimen was completed. All the authors had access to the final trial results and vouch for the fidelity of the trial to the protocol. The first two authors drafted the manuscript of this article, which was revised and approved by all the authors, who also vouch for the accuracy and completeness of the data and analyses reported. Microvascular and Renal Outcomes The prespecified secondary microvascular outcome was a composite of renal and retinal outcomes. 15 In this article, we focus on the renal outcomes, which were more common than the retinal events (Table S1 in the Supplementary Appendix, available at NEJM.org). The composite renal outcome consisted of new-onset persistent macroalbuminuria, persistent doubling of the serum creatinine level and an estimated GFR of 45 ml or less per minute per 1.73 m 2 of bodysurface area (hereafter referred to as persistent doubling of the serum creatinine level), the need for continuous renal-replacement therapy (end- 84 n engl j med 377;9 nejm.org August 31, 217

and Renal Outcomes in Type 2 Diabetes stage renal disease) with no reversible cause of the renal disease, or death from renal disease. Macroalbuminuria was defined as a concentration of more than 3 mg of albumin in a 24-hour urine sample or as a concentration of more than 3 mg of albumin per gram of creatinine in a first morning sample. To confirm the presence of persistent macroalbuminuria or doubling of the serum creatinine level, a second measurement was mandatory. Renal outcomes were ascertained every 12 months and at the end of the trial. All the abovementioned renal outcomes were adjudicated by an independent committee of experts who were unaware of the trial-group assignments, as described previously. 14,15 Additional nonadjudicated renal outcomes and adverse-event reporting are described in the Supplementary Methods section in the Supplementary Appendix. Laboratory Assessments Laboratory tests were performed centrally (at ICON in Warrington, PA). Creatinine levels (both urinary and serum) were measured enzymatically, calibrated to isotope dilution mass spectrometry values (Roche Hitachi), and the urinary albumin concentration was assessed by means of immunoprecipitation (Roche Hitachi). The serum creatinine level and the albumin-to-creatinine ratio were measured and calculated at randomization, annually, and at trial completion; the serum creatinine level was also measured at month 6. The GFR was estimated with the four-component Modification of Diet in Renal Disease formula, as specified in the trial protocol. 16 Microalbuminuria (or a high urinary albumin level) was defined as a concentration of 3 to 3 mg of albumin per gram of creatinine, and macroalbuminuria (or a very high urinary albumin level) as a concentration of more than 3 mg of albumin per gram of creatinine. If the urinary albumin measurements were below the lower limit of quantification (5 mg per liter), a value of 2.5 mg per liter was imputed for statistical analysis. Statistical Analysis The statistical analysis plan is available with the protocol, 14 and additional details are provided in the Supplementary Methods section in the Supplementary Appendix. In this secondary analysis, the required sample size was based on the primary cardiovascular outcome. 15 Although the trial was not powered for renal outcomes, we calculated post hoc that the trial had 85% power to detect a risk of the composite renal outcome that was 22% lower in the liraglutide group than in the placebo group. Renal outcomes were not included in the hierarchical testing strategy that was used for the primary cardiovascular outcome. All the analyses were performed at the nominal alpha level of.5 without correction for multiple hypothesis testing. In prespecified time-to-event analyses we applied a Cox proportional-hazards model with trial group as a covariate. Sensitivity analyses were performed with death as a competing risk. All the patients who underwent randomization were included and followed from the time of randomization until death or the end of followup, whichever came first. Prespecified between-group comparisons were performed at 36 months, which was the last trial visit at which laboratory testing was performed at the same time point in the entire population. We estimated trial-group ratios for the estimated GFR and the urinary albumin-tocreatinine ratio using a mixed-effects model for repeated measures on log-transformed values (see the Supplementary Methods section in the Supplementary Appendix). Subgroups of patients who were at increased renal risk (according to the estimated GFR and urinary albumin-to-creatinine ratio at baseline) were defined before unblinding. Results Trial Population A total of 934 patients underwent randomization, with 4668 patients being assigned to receive liraglutide and 4672 to receive placebo. The median follow-up of the patients was 3.84 years, and the mean percentage of time that patients were taking the trial regimen was 83.%. The characteristics of the patients at baseline were generally balanced between the trial groups (Tables S2 and S3 in the Supplementary Appen- n engl j med 377;9 nejm.org August 31, 217 841

The new england journal of medicine Table 1. Composite Renal Outcome and Individual Components of the Composite Outcome.* Outcome (N = 4668) (N = 4672) Total (N = 934) Hazard Ratio (95% CI) P Value no. of patients (rate per 1 patient-yr of observation) Composite renal outcome 268 (15.) 337 (19.) 65 (17.).78 (.67.92).3 Components of composite renal outcome New-onset persistent macroalbuminuria 161 (9.) 215 (12.1) 376 (1.6).74 (.6.91).4 Persistent doubling of serum creatinine 87 (4.9) 97 (5.5) 184 (5.2).89 (.67 1.19).43 level Renal-replacement therapy 56 (3.1) 64 (3.6) 12 (3.4).87 (.61 1.24).44 Death due to renal disease 8 (.4) 5 (.3) 13 (.4) 1.59 (.52 4.87).41 * There were 17,822 patient-years of observation in the liraglutide group and 17,741 in the placebo group. All the events were adjudicated. Hazard ratios and P values were estimated with the use of a Cox proportional-hazards model with trial group as a covariate. The composite renal outcome consisted of new-onset persistent macroalbuminuria, persistent doubling of the serum creatinine level and an estimated glomerular filtration rate of 45 ml or less per minute per 1.73 m 2 of body-surface area (referred to as persistent doubling of the serum creatinine level), the need for continuous renal-replacement therapy (end-stage renal disease), or death due to renal disease. One patient who had macroalbuminuria at baseline had an event of new-onset persistent macroalbuminuria that was confirmed by adjudication after the patient had regression to microalbuminuria earlier in the trial. dix). The mean age of the patients was 64 years, and the mean blood pressure 136/77 mm Hg. The mean estimated GFR was 8 ml per minute per 1.73 m 2 ; a total of 2.7% of the patients had an estimated GFR of 3 to 59 ml per minute per 1.73 m 2, and 2.4% had an estimated GFR of less than 3 ml per minute per 1.73 m 2. Microalbuminuria and macroalbuminuria, as assessed at randomization, were present in 26.3% and 1.5% of the patients, respectively. A total of 8632 patients received antihypertensive agents, and 7741 received renin angiotensin system blockers. Composite Renal Outcome The prespecified composite renal outcome occurred in fewer patients in the liraglutide group than in the placebo group (268 patients [5.7%] vs. 337 [7.2%]; hazard ratio,.78; 95% confidence interval [CI],.67 to.92; P =.3) (Table 1 and Fig. 1A). New-onset persistent macroalbuminuria occurred in fewer patients in the liraglutide group than in the placebo group (161 patients [3.4%] vs. 215 [4.6%]; hazard ratio,.74; 95% CI,.6 to.91; P =.4) (Fig. 1B). The risks of persistent doubling of the serum creatinine level and of end-stage renal disease (use of renalreplacement therapy) did not differ significantly between groups (Table 1 and Fig. 1C and 1D). There were few cases of death due to renal disease (8 patients in the liraglutide group and 5 in the placebo group). Sensitivity analyses, in which death from any cause was considered to be a competing risk, yielded similar results (Table S4 in the Supplementary Appendix). Similarly, analyses that took into account the differences between groups in the use of inhibitors of the renin angiotensin system, the glycated hemoglobin level, body weight, and blood pressure did not materially alter the results (Tables S5 through S8 in the Supplementary Appendix). Composite Renal Outcome in Subgroups The prespecified subgroups of patients with an elevated renal risk at baseline included 3422 patients with microalbuminuria or macroalbuminuria, 2158 with an estimated GFR of less than 6 ml per minute per 1.73 m 2, and 113 with both an estimated GFR of less than 6 ml per minute per 1.73 m 2 and microalbuminuria or macroalbuminuria (Fig. 2, and Table S9 in the Supplementary Appendix). The effect of liraglutide on the composite renal outcome appeared to be independent of subgroups (Fig. 2). In the subgroup of patients with microalbuminuria or macroalbuminuria, the composite renal outcome occurred in fewer patients in the liraglutide group than in the placebo group (23 of 1684 patients [13.7%] vs. 283 of 1738 [16.3%]; hazard ratio,.81; 95% CI,.68 to.96; P =.2). In the subgroup of patients with an estimated GFR of less than 6 ml per minute per 1.73 m 2, the 842 n engl j med 377;9 nejm.org August 31, 217

and Renal Outcomes in Type 2 Diabetes A Composite Renal Outcome 1 1 Hazard ratio,.78 (95% CI,.67.92) 8 P=.3 8 6 6 4 4 2 Patients with an Event (%) Patients with an Event (%) 2 6 12 18 24 3 36 42 48 54 6 12 18 24 3 36 42 48 54 4672 4668 4643 4635 454 4561 4428 4492 4316 44 4196 434 494 421 399 4114 1613 1632 433 454 C Persistent Doubling of Serum Creatinine Level 1 1 Hazard ratio,.89 (95% CI,.67 1.19) 8 P=.43 8 6 6 4 4 2 2 6 12 18 24 3 36 42 48 54 6 12 18 24 3 36 42 48 54 4672 4668 4647 4639 4596 4591 4529 4544 4447 4476 4367 443 4282 4332 4196 4264 1682 1692 456 475 B New Onset of Persistent Macroalbuminuria 1 1 Hazard ratio,.74 (95% CI,.6.91) 8 P=.4 8 6 6 4 4 2 Patients with an Event (%) Patients with an Event (%) 2 6 12 18 24 3 36 42 48 54 6 12 18 24 3 36 42 48 54 4672 4668 4646 4638 4551 457 4455 458 4359 4437 4252 4353 4162 4268 473 4182 1642 1662 D Continuous Renal-Replacement Therapy 1 1 Hazard ratio,.87 (95% CI,.61 1.24) 8 P=.44 8 6 6 4 4 2 2 442 461 6 12 18 24 3 36 42 48 54 4672 4668 4645 464 6 12 18 24 3 36 42 48 54 459 4596 4527 4547 4454 4484 437 4416 4299 4349 4227 4282 1699 171 461 483 Figure 1. Composite Renal Outcome and Components of the Composite Outcome. The primary composite renal outcome in the time-to-event analysis was a composite (Panel A) of the first occurrence of persistent macroalbuminuria (Panel B), persistent doubling of the serum creatinine level and an estimated glomerular filtration rate of 45 ml or less per minute per 1.73 m 2 of body-surface area (referred to as persistent doubling of the serum creatinine level; Panel C), the need for continuous renal-replacement therapy (for end-stage renal disease; Panel D), or death due to renal disease (data not shown). The component of death due to renal disease occurred in 13 patients (8 patients in the liraglutide group and 5 in the placebo group). Cumulative incidences were estimated with the use of the Kaplan Meier method, and the hazard ratios were calculated with the use of the Cox proportionalhazard regression model. The insets show the same data on an enlarged y axis. The data analyses are truncated at 54 months because less than 1% of the participants had an observation time beyond 54 months. All the events were adjudicated. One patient with macroalbuminuria at baseline had an event of new-onset persistent macroalbuminuria that was confirmed by adjudication after the patient had regression to microalbuminuria earlier in the trial. composite renal outcome occurred in 146 of 1116 patients (13.1%) in the liraglutide group and in 156 of 142 (15.%) in the placebo group (hazard ratio,.84; 95% CI,.67 to 1.5; P =.13). In patients with both an estimated GFR of less than 6 ml per minute per 1.73 m 2 and microalbuminuria or macroalbuminuria, the composite renal outcome occurred in 131 of 583 patients (22.5%) in the liraglutide group and in 141 of 547 (25.8%) in the placebo group (hazard ratio,.81; 95% CI,.64 to 1.3; P =.9). Renal Function over Time The estimated GFR declined continuously (Fig. 3A), but the decline was slightly slower in the liraglutide group than in the placebo group (estimated trial-group ratio at 36 months, 1.2; 95% CI, 1. to 1.3; P =.1, corresponding to a 2% less decrease with liraglutide). The decrease in the estimated GFR at 36 months was 7.44 ml per minute per 1.73 m 2 in the liraglutide group, as compared with 7.82 ml per minute per 1.73 m 2 in the placebo group. The urinary albumin-to- n engl j med 377;9 nejm.org August 31, 217 843

The new england journal of medicine Subgroup No. of Patients Hazard Ratio (95% CI) P Value for Interaction All patients Estimated GFR <6 ml/min/1.73 m 2 6 ml/min/1.73 m 2 Microalbuminuria or macroalbuminuria Yes No Combined estimated GFR and albuminuria status Estimated GFR <6 ml/min/1.73 m 2 and microalbuminuria or macroalbuminuria Estimated GFR 6 ml/min/1.73 m 2 or no microalbuminuria or macroalbuminuria 934 2158 7182 3422 5918 113 821.78 (.67.92).84 (.67 1.5).68 (.54.86).81 (.68.96).69 (.46 1.4).81 (.64 1.3).7 (.56.87).2.5.36.4 1. 2. Better Better Figure 2. Composite Renal Outcome, According to Baseline Renal Risk. The estimated glomerular filtration rate (GFR) was calculated with the use of the Modification of Diet in Renal Disease formula. Hazard ratios and P values were estimated with the use of a Cox proportional-hazards model with interaction between trial group and renal risk at baseline. The composite renal outcome consisted of new-onset persistent macroalbuminuria, persistent doubling of the serum creatinine level and an estimated GFR of 45 ml or less per minute per 1.73 m 2, the need for continuous renal-replacement therapy (end-stage renal disease), or death due to renal disease. creatinine ratio increased less in the liraglutide group, yielding a 17% lower urinary albumin-tocreatinine ratio at 36 months in favor of liraglutide (estimated trial-group ratio,.83; 95% CI,.79 to.88; P<.1) (Fig. 3B). The estimated increase in the urinary albumin-to-creatinine ratio at 36 months was 1.8 mg of albumin per gram of creatinine in the liraglutide group, as compared with 6.3 mg of albumin per gram of creatinine in the placebo group (Fig. 3B); in an analysis that excluded values outside the quantification range, the results were similar (Table S1A and S1B in the Supplementary Appendix). The smaller increase in albuminuria at 36 months in the liraglutide group than in the placebo group was independent of the baseline estimated GFR and baseline albuminuria (Table S1A and S1B and Fig. S1A and S1B in the Supplementary Appendix). When results were stratified according to the estimated GFR at baseline, the decrease in the estimated GFR in patients with a baseline estimated GFR of 3 to 59 ml per minute per 1.73 m 2 was 2 ml per minute per 1.73 m 2 in the liraglutide group, as compared with 4 ml per minute per 1.73 m 2 in the placebo group (estimated trialgroup ratio in favor of liraglutide, 1.7; 95% CI, 1.4 to 1.1; P <.1) (Fig. 4, and Fig. S2 in the Supplementary Appendix). Results did not differ significantly between groups in patients with a baseline estimated GFR of 6 ml or more per minute per 1.73 m 2 or with an estimated GFR of less than 3 ml per minute per 1.73 m 2 (there were few patients in the latter group). When analyzed according to albuminuria at baseline, the decrease in the estimated GFR was smaller in the liraglutide group than in the placebo group in patients with macroalbuminuria (P =.1) but did not differ significantly in those with microalbuminuria (P =.24) or normoalbuminuria (P =.6) (data not shown). Other Outcomes and Adverse Events Outcomes that were not prespecified included the composite of the doubling of the serum creatinine level or the use of renal-replacement therapy; there were no significant differences between the randomized groups (Table S11 in the Supplementary Appendix). New-onset microalbuminuria occurred in fewer patients in the liraglutide group than in the placebo group (2293 patients [49.1%] vs. 2498 [53.5%]; hazard ratio,.87; 95% CI,.83 to.93; P<.1). The rates of renal adverse events were similar 844 n engl j med 377;9 nejm.org August 31, 217

and Renal Outcomes in Type 2 Diabetes in the liraglutide group and the placebo group (15.1 events and 16.5 events per 1 patientyears), including the rate of acute kidney injury (7.1 events and 6.2 events per 1 patient-years, respectively), according to adverse-event reporting. The similarity in the rates of adverse events also applies to subgroups defined according to the estimated GFR at baseline. Details are provided in Table S12A and S12B in the Supplementary Appendix. Discussion Among patients who were receiving usual care, liraglutide resulted in significantly lower rates of renal outcomes than placebo among patients with type 2 diabetes who were at high cardiovascular risk. This result was driven mainly by a lower incidence of macroalbuminuria in the liraglutide group than in the placebo group. There were nonsignificantly lower risks of the doubling of the serum creatinine level and of end-stage renal disease with liraglutide than with placebo during up to 5 years of follow-up. New-onset persistent macroalbuminuria is an effect that is typically associated with subsequent progressive reductions in the GFR in patients with type 2 diabetes. 17 For example, in ONTARGET (Ongoing Telmisartan Alone and in Combination with Ramipril Global Endpoint Trial) and TRANSCEND (Telmisartan Randomized Assessment Study in ACE Intolerant Subjects with Cardiovascular Disease), new-onset macroalbuminuria was associated with a risk of end-stage renal disease or doubling of the serum creatinine level that was 3 to 5 times as high as the risk among patients in whom new-onset macroalbuminuria did not develop. 18 Similar renal outcomes associated with new-onset macroalbuminuria were reported in the ADVANCE (Action in Diabetes and Vascular Disease: Preterax and Diamicron Modified Release Controlled Evaluation) trial 19 and supported by meta-regression analyses. 17 Macroalbuminuria is also a risk factor for cardiovascular events. 17,2 In this present trial, the risks of doubling of the serum creatinine level and end-stage renal disease did not differ significantly between the liraglutide group and the placebo group, possibly owing to the moderate decline in the estimated GFR observed in this cohort and the few patients who had advanced kidney disease at A Estimated GFR Mean Estimated GFR (ml/min/1.73 m 2 ) 85 8 75 7 65 6 6 12 24 36 48 4672 4668 B Urinary Albumin-to-Creatinine Ratio 35 Urinary Albumin-to-Creatinine Ratio 3 25 2 Estimated trial-group ratio at 36 mo, 1.2 (95% CI, 1. 1.3) P=.1 4356 4349 4237 4288 3911 431 Figure 3. Changes in the Estimated GFR and Urinary Albumin-to-Creatinine Ratio. Panel A shows the estimated GFR, and Panel B the urinary albumin-tocreatinine ratio (with albumin measured in milligrams and creatinine measured in grams). Geometric means were estimated for the urinary albuminto-creatinine ratio with the use of a linear mixed model for log-transformed assessment, with accounting for repeated measures. Trial-group ratios were estimated with the use of a mixed-effect model for repeated measures on log-transformed values. Interaction between visit and, respectively, trial group, sex, geographic region, and use of antidiabetic therapy at baseline were included as fixed effects, and interaction between visit and baseline log-estimated GFR or baseline urinary albumin-to-creatinine ratio and age at baseline were included as covariates. The values for the urinary albuminto-creatinine ratio that were outside the range of quantification were imputed (see the Supplementary Methods section in the Supplementary Appendix). randomization. The initial changes in the estimated GFR are difficult to interpret, 21 but there may be a potential slowing in the reduction in the estimated GFR with liraglutide in patients with a low baseline estimated GFR. The mechanism behind the effect of liraglutide on the renal outcomes is unclear. Statistical adjustment for the glycated hemoglobin level at 3634 386 755 812 15 Estimated trial-group ratio at 36 mo,.83 (95% CI,.79.88) 1 P<.1 12 24 36 48 4559 4578 413 4167 3789 3934 359 3686 73 786 n engl j med 377;9 nejm.org August 31, 217 845

The new england journal of medicine A Estimated GFR >9 ml/min/1.73 m 2 Estimated GFR (ml/min/1.73 m 2 ) 11 18 16 14 12 1 98 96 94 92 6 12 24 36 1616 1589 1542 159 1514 15 C Estimated GFR 3 59 ml/min/1.73 m 2 Estimated GFR (ml/min/1.73 m 2 ) 48.5 47.5 46.5 45.5 44.5 43.5 42.5 41.5 4.5 1415 1428 1332 1364 B Estimated GFR 6 9 ml/min/1.73 m 2 Estimated GFR (ml/min/1.73 m 2 ) 75.5 74.5 73.5 72.5 71.5 7.5 69.5 68.5 67.5 66.5. 6 12 24 36 1947 197 1856 1825 1817 1795 D Estimated GFR <3 ml/min/1.73 m 2 Estimated GFR (ml/min/1.73 m 2 ) 28 26 24 22 2 18 16 1696 1689 1574 167. 6 12 24 36 6 12 24 36 919 986 865 912 821 896 725 826 663 754 14 115 93 13 85 97 75 88 64 81 Figure 4. Mean Estimated GFR during the Trial in Subgroups Stratified According to Estimated GFR at Baseline. Data points are group geometric means from observed data during the trial. I bars indicate ±1 SD. Changes in the estimated GFR between the liraglutide group and the placebo group differed significantly in the subgroup of patients who had an estimated GFR between 3 and 59 ml per minute per 1.73 m 2 at baseline (Fig. S2 in the Supplementary Appendix). baseline and during the trial did not materially alter the effect of liraglutide on renal outcomes. However, in a meta-analysis, intensified glucose control was associated with a lower incidence of new macroalbuminuria than was usual care. 4 In the United Kingdom Prospective Diabetes Study (UKPDS), a 1-percentage-point difference in the glycated hemoglobin level between groups was associated with a consistent benefit on microvascular outcomes only, after 1 years of followup. 22 In the ACCORD (Action to Control Cardiovascular Risk in Diabetes) trial, a difference of.8 percentage points in the glycated hemoglobin level was associated with a much earlier benefit on macroalbuminuria but no change in the risks of doubling of the serum creatinine level or end-stage renal disease with 4 years of follow-up. 23 The ADVANCE trial showed a difference of.8 percentage points in the glycated hemoglobin level and a substantial benefit with regard to new-onset albuminuria over a 5-year follow-up with no significant change in the risk of a doubling of the serum creatinine level but a tendency toward a lower risk of end-stage renal disease. 24 Other large, randomized trials involving patients with type 2 diabetes have also reported microvascular events. 25-28 As in the present trial, these trials have shown between-group differences in the glycated hemoglobin level of between.2 and.4 percentage points, and benefits of active compounds on albuminuria, but not on other renal outcomes. In contrast, the EMPA-REG OUTCOME trial showed that empagliflozin resulted in significantly lower risks 846 n engl j med 377;9 nejm.org August 31, 217

and Renal Outcomes in Type 2 Diabetes than placebo of progression to macroalbuminuria, the doubling of the serum creatinine level, and despite small numbers, end-stage renal disease. 11,29 It appears to be unlikely that the moderate between-group differences in systolic blood pressure and body weight can fully explain the effect on renal outcomes. In an analysis that took into account those differences, the composite renal outcome still occurred less frequently with liraglutide than with placebo. The mechanisms behind the renal effects of liraglutide are probably multifactorial. Most studies investigating the effect of GLP-1 treatment on renal hemodynamic variables in patients with type 2 diabetes have shown neutral effects. 12,13 Preclinical studies have shown convincingly that GLP-1 therapy decreases the level of inflammation and oxidative stress and prevents diabetic nephropathy and acute kidney injury. 1,3 Furthermore, a recent randomized trial involving patients with type 2 diabetes and albuminuria showed that liraglutide treatment reduced levels of inflammatory biomarkers. 31 Hence, it is possible that preservation of renal function and the antialbuminuric effects of liraglutide may be due to antiinflammatory effects rather than renal hemodynamic effects. There are some limitations to the present analysis. Although the trial prespecified renal outcomes as secondary outcomes, it was not powered for analyses of the individual renal outcomes. Also, patients were followed for 3.5 to 5. years, and thus we cannot extrapolate to longer-term effects. The trial design prevents us from ascertaining the extent to which the active drug, glucose control, or other factors (including concomitant medications) explain outcomes. A significantly lower risk of renal outcomes was still observed when the analysis was adjusted for change in the glycated hemoglobin level, body weight, and blood pressure. That said, in the absence of head-to-head comparisons, other direct or indirect drug effects cannot be ruled out. Nonetheless, concomitant medications were more frequently added in the placebo group during the trial than in the liraglutide group. 14 In conclusion, in this secondary analysis, among patients with type 2 diabetes at high risk for cardiovascular disease who were receiving usual care, liraglutide resulted in a lower risk of the composite renal outcome than placebo, primarily owing to a lower rate of new-onset persistent macroalbuminuria. Supported by Novo Nordisk and by a grant (UL1TR1111, to Dr. Buse) from the National Institutes of Health. Disclosure forms provided by the authors are available with the full text of the article at NEJM.org. We thank Drs. Florian Baeres and Thomas Idorn (Novo Nordisk) for scientific input, and the participants and trial-site staff. References 1. Ritz E, Orth SR. Nephropathy in patients with type 2 diabetes mellitus. N Engl J Med 1999; 341: 1127-33. 2. Alegre-Díaz J, Herrington W, López- Cervantes M, et al. Diabetes and causespecific mortality in Mexico City. N Engl J Med 216; 375: 1961-71. 3. Molitch ME, Adler AI, Flyvbjerg A, et al. Diabetic kidney disease: a clinical update from Kidney Disease: Improving Global Outcomes. Kidney Int 215; 87: 2-3. 4. Coca SG, Ismail-Beigi F, Haq N, Krumholz HM, Parikh CR. Role of intensive glucose control in development of renal end points in type 2 diabetes mellitus: systematic review and meta-analysis. Arch Intern Med 212; 172: 761-9. 5. Perkovic V, Heerspink HL, Chalmers J, et al. Intensive glucose control improves kidney outcomes in patients with type 2 diabetes. Kidney Int 213; 83: 517-23. 6. UK Prospective Diabetes Study (UKPDS) Group. Effect of intensive bloodglucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet 1998; 352: 854-65. 7. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998; 352: 837-53. 8. Stevens PE, Levin A. Evaluation and management of chronic kidney disease: synopsis of the Kidney Disease: Improving Global Outcomes 212 clinical practice guideline. Ann Intern Med 213; 158: 825-3. 9. Zeisberg M, Zeisberg EM. Evidence for antifibrotic incretin-independent effects of the DPP-4 inhibitor linagliptin. Kidney Int 215; 88: 429-31. 1. Fujita H, Morii T, Fujishima H, et al. The protective roles of GLP-1R signaling in diabetic nephropathy: possible mechanism and therapeutic potential. Kidney Int 214; 85: 579-89. 11. Wanner C, Inzucchi SE, Lachin JM, et al. Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 216; 375: 323-34. 12. von Scholten BJ, Lajer M, Goetze JP, Persson F, Rossing P. Time course and mechanisms of the anti-hypertensive and renal effects of liraglutide treatment. Diabet Med 215; 32: 343-52. 13. Skov J, Pedersen M, Holst JJ, et al. Short-term effects of liraglutide on kidney function and vasoactive hormones in type 2 diabetes: a randomized clinical trial. Diabetes Obes Metab 216; 18: 581-9. 14. Marso SP, Daniels GH, Brown-Frandsen K, et al. and cardiovascular outcomes in type 2 diabetes. N Engl J Med 216; 375: 311-22. 15. Marso SP, Poulter NR, Nissen SE, et al. Design of the Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial. Am Heart J 213; 166(5): 823-3.e5. 16. Stevens LA, Coresh J, Greene T, Levey AS. Assessing kidney function measured and estimated glomerular filtration rate. N Engl J Med 26; 354: 2473-83. 17. Fox CS, Matsushita K, Woodward M, et al. Associations of kidney disease measures with mortality and end-stage renal disease in individuals with and without diabetes: a meta-analysis. Lancet 212; 38: 1662-73. 18. Schmieder RE, Mann JF, Schumacher H, et al. Changes in albuminuria predict n engl j med 377;9 nejm.org August 31, 217 847

and Renal Outcomes in Type 2 Diabetes mortality and morbidity in patients with vascular disease. J Am Soc Nephrol 211; 22: 1353-64. 19. Ninomiya T, Perkovic V, de Galan BE, et al. Albuminuria and kidney function independently predict cardiovascular and renal outcomes in diabetes. J Am Soc Nephrol 29; 2: 1813-21. 2. Gerstein HC, Mann JF, Yi Q, et al. Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA 21; 286: 421-6. 21. Clase CM, Barzilay J, Gao P, et al. Acute change in glomerular filtration rate with inhibition of the renin-angiotensin system does not predict subsequent renal and cardiovascular outcomes. Kidney Int 217; 91: 683-9. 22. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HAW. 1-Year followup of intensive glucose control in type 2 diabetes. N Engl J Med 28; 359: 1577-89. 23. The Action to Control Cardiovascular Risk in Diabetes Study Group. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 28; 358: 2545-59. 24. Zoungas S, Woodward M, Li Q, et al. Impact of age, age at diagnosis and duration of diabetes on the risk of macrovascular and microvascular complications and death in type 2 diabetes. Diabetologia 214; 57: 2465-74. 25. Green JB, Bethel MA, Armstrong PW, et al. Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med 215; 373: 232-42. 26. Scirica BM, Bhatt DL, Braunwald E, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 213; 369: 1317-26. 27. White WB, Cannon CP, Heller SR, et al. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med 213; 369: 1327-35. 28. Pfeffer MA, Claggett B, Diaz R, et al. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 215; 373: 2247-57. 29. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 215; 373: 2117-28. 3. Kodera R, Shikata K, Kataoka HU, et al. Glucagon-like peptide-1 receptor agonist ameliorates renal injury through its antiinflammatory action without lowering blood glucose level in a rat model of type 1 diabetes. Diabetologia 211; 54: 965-78. 31. von Scholten BJ, Persson F, Rosenlund S, et al. Effects of liraglutide on cardiovascular risk biomarkers in patients with type 2 diabetes and albuminuria: a subanalysis of a randomized, placebo-controlled, double-blind, crossover trial. Diabetes Obes Metab 217; 19: 91-5. Copyright 217 Massachusetts Medical Society. 848 n engl j med 377;9 nejm.org August 31, 217