Download as pdf or txt
Download as pdf or txt
You are on page 1of 34

1 Semaglutide and Diuretic Use in Obesity-Related Heart Failure with

2 Preserved Ejection Fraction: A Pooled Analysis of the STEP-HFpEF and

3 STEP-HFpEF-DM trials

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


PT
4
5 Sanjiv J. Shah 1 , Kavita Sharma 2 , Barry A. Borlaug3 , Javed Butler4 , Melanie Davies5 ,

RI
Dalane W Kitzman 6, Mark C. Petrie 7, Subodh Verma8 , Shachi Patel9 , Khaja M. Chinnakondepalli9 , Mette

7 N. Einfeldt10 , Thomas J. Jensen 10 , Søren Rasmussen 10 , Rabea Asleh 11 , Tuvia Ben-Gal12 ,

SC
8 Mikhail N. Kosiborod 9 * for the STEP-HFpEF Trial Committees and Investigators

U
10 1
Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine,

11 N
Chicago, IL, USA; 2 Division of Cardiology, The Johns Hopkins University School of Medicine,
A
12 Baltimore, MD, USA; 3 Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA;
M

13 4
Baylor Scott and White Research Institute, Dallas, TX, USA; and Department of Medicine, University of

14 Mississippi, Jackson, MS, USA; 5 Diabetes Research Centre, University of Leicester, Leicester, UK; and
D

15 NIHR Leicester Biomedical Research Centre, Leicester, UK; 6 Department of Internal Medicine, Sections
TE

16 on Cardiovascular Medicine and Geriatrics/Gerontology, Wake Forest University School of Medicine,

17 Winston-Salem, NC, USA; 7 School of Cardiovascular and Metabolic Health, University of Glasgow,
EP

18 Glasgow, UK; 8 Division of Cardiac Surgery, Li Ka Shing Knowledge Institute of St Michael’s Hospital,

19 Unity Health Toronto, University of Toronto, Toronto, ON, Canada; 9 Department of Cardiovascular
CC

20 Disease, Saint Luke’s Mid America Heart Institute, Kansas City, MO, USA; 10 Novo Nordisk A/S, Søborg,

21 Denmark; 11 Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew
A

22 University of Jerusalem, Jerusalem, Israel; and 12


Heart Failure Unit, Cardiology Department, Rabin

23 Medical Center, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel

24

© The Author(s) 2024. Published by Oxford University Press on behalf of the European Society of Cardiology. This
is an Open Access article distributed under the terms of the Creative Commons Attribution License
(https://creativecommons.org/licenses/by/4.0/), which permits unrestricted reuse, distribution, and reproduction in any
medium, provided the original work is properly cited. 1
1 *Corresponding author:

2 Mikhail N. Kosiborod

3 Saint Luke's Mid America Heart Institute, Kansas City, MO, USA.

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 Email: mkosiborod@saint-lukes.org

PT
5 Phone: + 1 8169323445

RI
6
7 Brief title: Semaglutide and diuretic use in the STEP-HFpEF Program
8

SC
9
10 ABSTRACT (359/250 words)

11 Background and Aims

U
12 In the STEP-HFpEF trial program, treatment with semaglutide resulted in multiple beneficial

13
N
effects in patients with obesity-related heart failure with preserved ejection fraction (HFpEF).
A
14 Efficacy may vary according to baseline diuretic use, and semaglutide treatment could modify
M

15 diuretic dose.
D

16 Methods
TE

17 In this pre-specified analysis of pooled data from the STEP-HFpEF and STEP-HFpEF-DM trials

18 (n=1145), which randomized participants with HFpEF and body mass index ≥30 kg/m2 to once
EP

19 weekly semaglutide 2.4 mg or placebo for 52 weeks, we examined whether efficacy and safety

20 endpoints differed by baseline diuretic use, as well as the effect of semaglutide on loop diuretic
CC

21 use and dose changes over the 52-week treatment period.

22
A

Results

23 At baseline, across no diuretic (n=220), non-loop diuretic only (n=223), and loop diuretic (<40

24 [n=219], 40 [n=309], and >40 [n=174] mg/day furosemide-equivalents) groups, there was

25 progressively higher prevalence of hypertension and atrial fibrillation; and severity of obesity

2
1 and heart failure. Over 52 weeks of treatment, semaglutide had a consistent beneficial effect on

2 change in body weight across diuretic use categories (adjusted mean difference vs. placebo

3 ranged from -8.8% [95% CI -10.3, -6.3] to -6.9% [95% CI -9.1, -4.7] from no diuretics to the

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 highest loop diuretic dose category; interaction P=0.39). Kansas City Cardiomyopathy

PT
5 Questionnaire clinical summary score improvement was greater in patients on loop diuretics

RI
6 compared to those not on loop diuretics (adjusted mean difference vs. placebo: +9.3 [6.5;12.1]

7 vs. +4.7 points [1.3, 8.2]; P=0.042). Semaglutide had consistent beneficial effects on all

SC
8 secondary efficacy endpoints (including 6-min walk distance) across diuretic subgroups

9 (interaction P=0.24-0.92). Safety also favored semaglutide versus placebo across the diuretic

U
10 subgroups. From baseline to 52 weeks, loop diuretic dose decreased by 17% in the semaglutide

11
N
group vs. a 2.4% increase in the placebo group (P<0.0001). Semaglutide (vs. placebo) was more
A
12 likely to result in loop diuretic dose reduction (odds ratio [OR] 2.67 [95% CI 1.70, 4.18]) and
M

13 less likely dose increase (OR 0.35 [95% CI 0.23, 0.53]; P<0.001 for both) from baseline to 52
D

14 weeks.
TE

15 Conclusions

16 In patients with obesity-related HFpEF, semaglutide improved heart failure-related symptoms


EP

17 and physical limitations across diuretic use subgroups, with more pronounced benefits among

18 patients receiving loop diuretics at baseline. Reductions in weight and improvements in exercise
CC

19 function with semaglutide versus placebo were consistent in all diuretic use categories.

20
A

Semaglutide also led to a reduction in loop diuretic use and dose between baseline and 52 weeks.

21 Key words: Glucagon-like peptide-1 receptor agonist, Clinical trial, Loop diuretics, Obesity,

22 Heart failure with preserved ejection fraction

23 ClinicalTrials.gov registration: NCT04788511 and NCT04916470

3
Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024
PT
RI
SC
U
N
A
M
D
TE
EP
CC

2
A

3 Abbreviations: CI, confidence interval; HR, hazard ratio; KCCQ-CSS, Kansas City

4 Cardiomyopathy Questionnaire clinical summary score

4
1 Introduction

2 Patients with heart failure (HF) and preserved ejection fraction (HFpEF) frequently

3 receive loop diuretics, which are first-line agents for decongestion but can cause electrolyte

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 abnormalities, worsening kidney function, and hypotension. 1 Furthermore, outpatient escalation

PT
5 in loop diuretic dose is associated with adverse outcomes and is increasingly viewed as a proxy

RI
6 for HF hospitalizations.2-9 In HFpEF, higher body mass index is associated with greater use and

7 doses of loop diuretics;10,11 and in patients with obesity-related HFpEF, loop diuretics appear to

SC
8 be less effective for decongestion and have an exaggerated unfavorable impact on kidney

U
9 function, as compared with those that have HFpEF but no obesity. 12 In the STEP-HFpEF trial

10
N
program (which included STEP-HFpEF and STEP-HFpEF-DM trials), the glucagon-like
A
11 peptide-1 receptor agonist (GLP1-RA) semaglutide, at a weight management dose of 2.4 mg
M
12 once weekly, improved HF-related symptoms and physical limitations (as measured by the

13 Kansas City Cardiomyopathy Questionnaire clinical summary score [KCCQ-CSS]), reduced


D

14 body weight, improved exercise function (6-min walk distance [6MWD]), and reduced markers
TE

15 of inflammation (C-reactive protein [CRP]) and myocardial end-diastolic wall stress (N-terminal

16 pro-B-type natriuretic peptide [NT-proBNP]) in obesity-related HFpEF compared with


EP

17 placebo.13-15 Use of semaglutide also led to fewer adjudicated HF events (pooled hazard ratio
CC

18 0.27, 95% confidence interval [CI] 0.12-0.56; P=0.0004).15

19 Whether the effects of semaglutide vary according to baseline diuretic use and dose, and
A

20 whether semaglutide (as compared with placebo) has an effect on loop diuretic use and dose over

21 time are important, clinically relevant questions. We therefore examined whether semaglutide

22 efficacy varies according to baseline diuretic use, and whether semaglutide results in changes in

5
1 loop diuretic use and dose over time in a pre-specified analysis of pooled data from the STEP-

2 HFpEF and STEP-HFpEF-DM trials.

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 Methods

PT
5 Trial program design

RI
6 We conducted a pre-specified analysis of the randomized, international, multicenter,

7 double-blind, placebo-controlled STEP-HFpEF program.13-15 The program comprised two trials:

SC
8 STEP-HFpEF, which was conducted in patients with obesity-related HFpEF (body mass index

U
9 [BMI] ≥30 kg/m2 , left ventricular ejection fraction [LVEF] ≥45%) without type 2 diabetes

10
N
(NCT04788511);13 and STEP-HFpEF-DM in patients with obesity-related HFpEF and type 2
A
11 diabetes (NCT04916470).14 The design and primary results of the individual trials, and the
M
12 overall program, have been published previously. 13,14,16 The two trials were conducted from 2021

13 to 2023 at 129 sites across 18 countries in Asia, Europe, and North and South America.
D

14 Institutional Review Board ethics approval was obtained at each study site, and all patients
TE

15 provided written, informed consent to participate in the trial. The steering committee, which

16 included both academic members and representatives from the sponsor (Novo Nordisk),
EP

17 designed both trials and was responsible for the academic publications. A global expert panel
CC

18 provided academic, medical, and operational input in each country. The sponsor of the trial

19 program was Novo Nordisk.


A

20

21 Trial participants

22 Participants were eligible if they had symptomatic HF, LVEF ≥45%, BMI ≥30 kg/m2 ,

23 New York Heart Association (NYHA) functional class II to IV, KCCQ-CSS <90 points, and at

6
1 least one of the following: (1) elevated filling pressures (based on right heart catheterization or

2 pulmonary artery pressure sensor technology); (2) elevated natriuretic peptide levels (with

3 thresholds stratified based on BMI) plus echocardiographic abnormalities; or (3) HF

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 hospitalization in the previous 12 months plus a requirement for ongoing diuretic treatment

PT
5 and/or echocardiographic abnormalities.

RI
6 Key exclusion criteria were prior or planned bariatric surgery, self-reported change in

7 body weight >5 kg within 90 days before randomization, or a systolic blood pressure of

SC
8 >160 mmHg at screening. In STEP-HFpEF-DM, patients with uncontrolled diabetic retinopathy

9 or maculopathy were also excluded.

U
10

11 Randomization and trial procedures


N
A
12 Eligible participants were randomized 1:1 to receive a once-weekly target dose of
M

13 semaglutide 2.4 mg subcutaneously or matching placebo on top of standard of care for 52 weeks.
D

14 Randomization was stratified by BMI <35 kg/m2 vs ≥35 kg/m2 . Among patients with type 2
TE

15 diabetes enrolled in the STEP-HFpEF-DM trial, semaglutide or placebo were added to

16 background glucose-lowering medications, which could comprise any drug other than GLP1-
EP

17 RAs. Modification of glucose-lowering treatment was at the discretion of the investigator.

18 Specific guidance regarding the adjustment of sulfonylurea and insulin doses was provided to
CC

19 mitigate the risk of hypoglycemia. The management of diuretic therapy was left to the discretion

20
A

of the investigators and treating physicians; no specific instructions regarding the use and dose of

21 diuretics was provided. Detailed ascertainment of diuretic use and types, and doses of loop

22 diuretics, was conducted at baseline, 20-, 36-, and 52-week visits in both trials. Loop diuretics,

23 thiazide diuretics, and mineralocorticoid receptor antagonists (MRAs) were considered diuretics;

7
1 and sodium-glucose cotransporter 2 (SGLT2) inhibitors were not considered diuretics for the

2 current analysis. All non-furosemide loop diuretic doses were converted to mg/day furosemide

3 equivalents based on published equivalent dose conversions, as listed in Supplemental Table

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 1.17 Participants not taking loop diuretics at baseline or at subsequent follow-up visits were

PT
5 coded using a dose of 0 mg/day furosemide equivalents.

RI
6

7 Outcomes

SC
8 The primary aims of the current analysis were to investigate the effects of semaglutide

9 2.4 mg once weekly, compared with placebo, on (1) efficacy and safety outcomes across baseline

U
10 diuretic use groups (no diuretics, non-loop diuretics only, and loop diuretics [<40, 40, and >40

11
N
mg/day furosemide equivalents]); and (2) changes in loop diuretic use and dose over 52 weeks.
A
12 The dual primary endpoints of the STEP-HFpEF program were: (1) change in KCCQ-
M

13 CSS from baseline to 52 weeks; and (2) percent change in body weight from baseline to
D

14 52 weeks. The confirmatory secondary endpoints were change in 6MWD from baseline to
TE

15 52 weeks; a hierarchical composite endpoint (comprised of all-cause death [from baseline to 57

16 weeks], HF events [from baseline to 57 weeks], differences in several thresholds [≥5, ≥10, ≥15
EP

17 points] of change in KCCQ-CSS from baseline to 52 weeks, and difference in 6MWD change

18 [≥30 m] from baseline to 52 weeks); and change in high-sensitivity CRP from baseline to
CC

19 52 weeks. HF events were adjudicated by a blinded Clinical Events Committee as previously

20
A

described.13 We also examined the additional supportive secondary endpoints, including change

21 in systolic blood pressure, waist circumference, and KCCQ overall summary score; and

22 exploratory endpoints of change in NT-proBNP levels, and change in the additional KCCQ

23 domains (total symptom score, physical limitations score [PLS], social limitations score, and

8
1 quality of life score) from baseline to 52 weeks. Safety endpoints included in the current analysis

2 were serious adverse events (SAEs), which included SAEs leading to permanent treatment

3 discontinuation, cardiac and gastrointestinal SAEs; and gastrointestinal adverse events leading to

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 discontinuation of study drug.

PT
5

RI
6

SC
8 Statistical analysis

9 Baseline characteristics were examined according to the five aforementioned baseline

U
10 diuretic use/dose groups. The Jonckheere–Terpstra trend test (for continuous variables), the

11
N
Cochran-Armitage trend test (for categorical variables) and Cochran-Mantel-Haenszel test (for
A
12 multinomial variables) were used to evaluate differences among the five groups.
M

13 All efficacy analyses (including change in loop diuretic use and dose) were done using
D

14 the full analysis set (all randomized participants according to the intention-to-treat principle,
TE

15 while in trial, regardless of treatment discontinuation). For change in KCCQ scores and 6MWD,

16 missing observations at week 52 caused by cardiovascular death or previous HF events were


EP

17 single imputed to the lowest observed value across both treatment arms and visits. Missing

18 values due to other reasons were imputed using multiple imputation from participants with non-
CC

19 missing values in the same randomized treatment arm. For other endpoints, missing observations

20
A

at week 52 were multiple imputed irrespective of death or prior HF events using the same

21 imputation method. Analyses were performed using analysis of covariance (ANCOVA) models

22 for continuous endpoints, with change in the corresponding endpoint at week 52 as the

23 dependent variable, with adjustment for the baseline value of the relevant continuous outcome

9
1 variable, and treatment, trial, and BMI stratification as fixed factors using 1000 imputations. To

2 determine whether efficacy endpoints were consistent across the diuretic groups, we included a

3 diuretic group × treatment interaction term in all models. Estimates were then combined using

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 Rubin’s rule. Interaction P-values were derived from an F-test of equality between the treatment

PT
5 differences across diuretic groups. Furthermore, trend P-values for difference in semaglutide

RI
6 versus placebo treatment across the diuretic medication groups were also derived for the various

7 endpoints. In sensitivity analyses, these analyses were repeated using the comparisons of (1) no

SC
8 diuretic vs. any diuretic use at baseline; and (2) no loop diuretic vs. loop diuretic (any dose) at

U
baseline.

10 To further explore the relationship between baseline loop diuretic dose and key efficacy

11
N
endpoints (changes in KCCQ-CSS, body weight, 6MWD, high-sensitivity CRP, NT-proBNP),
A
12 interaction P-values between the loop diuretic dose as a continuous variable (modeled as a
M

13 spline) and randomized treatment at week 52 were derived to assess potential heterogeneity of
D

14 treatment effects (semaglutide vs placebo) across the range of loop diuretic doses.
TE

15 Analyses of the hierarchical composite endpoint (win ratio 18 ) were performed stratified

16 by diuretic use groups, based on direct comparisons of each participant randomized to


EP

17 semaglutide vs each participant randomized to placebo. For each of the participant pairs, a

18 “treatment winner” based on similar observation time was declared based on the endpoint
CC

19 hierarchy. The win ratio (i.e., the proportion of winners randomized to semaglutide divided by
A

20 the winners randomized to placebo) was estimated independently within each diuretic group

21 (using 1000 imputations as described above). A win ratio of 1 indicates no difference between

22 treatment groups; a win ratio >1 favors the active treatment; and a win ratio <1 favors the

10
1 placebo. The test for equality of the diuretic groups for the win ratio was performed using a

2 Cochran’s Q test.

3 Safety endpoints across the diuretic groups were analyzed using the safety analysis set

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 (all randomized participants exposed to at least one dose of randomized treatment) and either on-

PT
5 treatment or in-trial data sets, depending on the type of safety event.

RI
6 The difference between treatment groups for change in loop diuretic dose from baseline

7 to week 52 was calculated using an ANCOVA model, with trial, treatment and BMI as fixed

SC
8 factors, and adjusted for baseline loop diuretic dose. Logistic regression was used to determine

9 the odds ratio (OR) and 95% CI for baseline to 52-week increase or decrease in loop diuretic

U
10 dose, with trial, treatment and BMI as fixed factors, and adjusted for baseline loop diuretic dose.

11
N
Supportive analyses were performed for baseline to 20-week, and baseline to 36-week increase
A
12 or decrease in loop diuretic dose, using the same methodology. Besides changes in loop diuretic
M

13 doses between baseline and 52 weeks, a new start of loop diuretic was considered a dose
D

14 increase, and discontinuation of loop diuretics was considered a dose decrease, and patients who
TE

15 died, withdrew from the study or were lost to follow-up, were excluded from these analyses.

16 Baseline characteristics were compared across the three loop diuretic change groups (dose
EP

17 decrease, no change, dose increase) using the same analytic techniques as those described above

18 for the comparison of baseline characteristics across diuretic groups. Logistic regression was
CC

19 then used to determine baseline characteristics associated with diuretic dose escalation.

20
A

No adjustment for multiple testing was performed. A two-sided P-value of <0.05 was

21 considered significant in all analyses except interaction testing, in which an interaction P-value

22 <0.10 was considered significant. Results are presented as estimated changes from baseline to

23 week 52 for continuous endpoints or a win ratio (for the hierarchical composite endpoint), with a

11
1 95% CI and a two-sided P-value. NT-proBNP and high-sensitivity CRP were log-transformed,

2 and hence, treatment ratios at week 52 are reported. Analyses were conducted by the independent

3 statistical group at Saint Luke’s Mid America Heart Institute in collaboration with Novo Nordisk,

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 using SAS vs. 9.4 (SAS Institute, Cary, NC, USA). All analyses were performed on anonymized

PT
5 data.

RI
6

7 Results

SC
8 Baseline characteristics

U
9 A total of 1145 patients were randomized across the STEP-HFpEF program (n=529 in

10
N
STEP-HFpEF and n=616 in STEP-HFpEF-DM). At baseline, 220 (19.2%) of the participants
A
11 were taking no diuretics, 223 (19.5%) were taking non-loop diuretic(s) only, and the remaining
M
12 702 (61.3%) were taking loop diuretics. Of the total 1145 trial participants, 219 (19.1%), 309

13 (30.0%), and 174 (15.2%) were taking a loop diuretic dose of <40, 40, and >40 mg/day
D

14 furosemide-equivalents, respectively. Supplemental Figure 1 displays the distribution of loop


TE

15 diuretic doses at baseline. Table 1 displays the baseline characteristics, stratified by baseline

16 diuretic use/dose. Across diuretic groups, there was a stepwise increase in proportion of White
EP

17 participants, NYHA functional class III symptoms, hypertension, and atrial fibrillation. BMI,
CC

18 waist circumference, high-sensitivity CRP, and NT-proBNP values also increased in a stepwise

19 fashion from no diuretics to the highest loop diuretic dose category. Greater loop diuretic
A

20 use/dose was associated with lower LVEF, KCCQ-CSS, and 6MWD. There were no differences

21 in SGLT2 inhibitor use and angiotensin receptor-neprilysin inhibitor use across diuretic groups.

22 MRA use was highest in the highest loop diuretic dose group. Insulin use increased, and

12
1 dipeptidyl peptidase-4 inhibitors decreased, in a stepwise fashion from no diuretics to the highest

2 dose loop diuretic group.

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 Effects of semaglutide vs. placebo on efficacy and safety endpoints by baseline diuretic use

PT
5 The effects of semaglutide on the dual primary, confirmatory secondary, supportive

RI
6 secondary and exploratory endpoints across the diuretic use subgroups are presented in Table 2.

7 The benefits of semaglutide were consistent across the health status, body weight, exercise

SC
8 function, and biomarker endpoints across the diuretic use categories (interaction P>0.10 for all

9 endpoints except for KCCQ-PLS [interaction p=0.092]; Table 2). However, progressively larger

U
10 increases (improvements) in the KCCQ-CSS (and PLS) domains occurred with semaglutide vs.

11
N
placebo from the no diuretic subgroup to the highest dose loop diuretic subgroup (Table 2).
A
12 These results were verified in sensitivity analyses comparing (1) no diuretic vs. any diuretic
M

13 groups (Supplemental Table 2) and (2) no loop diuretic vs. loop diuretic (any dose) groups
D

14 (Supplemental Table 3), which demonstrate that KCCQ-CSS improvements were larger in
TE

15 patients on any diuretic (compared to no diuretic), and any dose of loop diuretic (compared to no

16 loop diuretic), in particular (adjusted mean KCCQ-CSS change for semaglutide vs. placebo: +9.3
EP

17 points [6.5, 12.1] in participants on loop diuretics at baseline vs. +4.7 points [1.3, 8.2] in

18 participants not on loop diuretics at baseline; P for interaction =0.042). Win ratios were similar
CC

19 across diuretic use groups (no diuretics [1.17 (95% CI 0.85-1.63)]; non-loop diuretics only [1.64

20
A

(95% CI 1.17-2.30)]; <40 mg/day loop diuretic [1.73 (95% CI 1.19-2.52)]; 40 mg/day loop

21 diuretic [1.79 (95% CI 1.34-2.40)]; and >40 mg loop diuretic [2.06 (95% CI 1.39-3.06)]), with

22 no treatment heterogeneity (interaction P=0.24), indicating that semaglutide had consistent

23 efficacy on the hierarchical composite endpoint across diuretic groups.

13
1 Figure 1 displays the restrictive cubic spline curves of key efficacy outcomes over the

2 range of baseline loop diuretic dose (total daily mg furosemide-equivalents). When examined as

3 a continuous variable, KCCQ-CSS improvement was greater in the semaglutide vs. placebo

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 groups with increasing doses of loop diuretics (P=0.026). No significant interactions were

PT
5 observed for changes in body weight, 6MWD, CRP or NT-proBNP.

RI
6 There were fewer SAEs, and cardiac SAEs, in patients treated with semaglutide vs.

7 placebo across all diuretic groups (Table 3). Gastrointestinal SAEs occurred at similar rates in

SC
8 the semaglutide and placebo groups across diuretic groups (Table 3).

U
10 Effects of semaglutide vs. placebo on loop diuretic dose

11
N
From baseline to 52 weeks, loop diuretic dose decreased by 17% (from mean±SD
A
12 48.4±2.7 to 40.2±2.1 mg/day furosemide equivalents) in the semaglutide group vs. an increase of
M

13 2.4% (53.4±2.7 to 54.7±3.1 mg/day) in the placebo group, which resulted in a difference of 11.8
D

14 (95% CI 6.8, 16.8) mg/day lower loop diuretic dose in semaglutide vs. placebo groups
TE

15 (P<0.0001) (Figure 2). Compared with placebo, semaglutide-treated patients were also more

16 likely to experience a loop diuretic dose reduction (OR 2.67 [95% CI 1.70, 4.18]) and less likely
EP

17 to experience a dose increase (OR 0.35 [0.23, 0.53]) between baseline and 52 weeks; P<0.001 for

18 both (Figure 3). The results were consistent in the supportive analyses which examined loop
CC

19 diuretic dose reduction and dose increase at 20 weeks, and 36 weeks (Supplemental Tables 4A

20
A

and 4B, respectively). Semaglutide led to a lower incidence of new starts of loop diuretics in

21 those not on loop diuretics at baseline (HR 0.29 [95% CI 0.16, 0.52]; P<0.0001) compared with

22 placebo; and higher incidence of stopping loop diuretics in those on loop diuretics at baseline

23 (HR 2.69 [95% CI 1.19, 6.12]; P=0.02) (Figure 4). As shown in Tables 4-5, participants who

14
1 required a loop diuretic dose escalation (increase) were more frequently assigned to the placebo

2 group and were more likely to have the following at baseline: NYHA class III/IV symptoms,

3 atrial fibrillation, obstructive sleep apnea, and treatment with insulin. Higher LV ejection

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 fraction, SGLT2-inhibitor use, and renin angiotensin system blockade were all associated with

PT
5 lower likelihood of loop diuretic dose escalation. Figure 5 displays the correlation between

RI
6 changes in loop diuretic dose over time vs. changes in key efficacy endpoints, stratified by

7 treatment group. There was a linear relationship between reduction in loop diuretic dose and

SC
8 increase (improvement) in KCCQ-CSS, reduction in body weight, and reduction in high-

9 sensitivity CRP in the semaglutide group.

U
10

11
N
A
12
M

13
D

14 Discussion
TE

15 In a pre-specified analysis of the STEP-HFpEF program, we found that in patients with

16 obesity-related HFpEF, semaglutide 2.4 mg once weekly, compared with placebo, improved HF-
EP

17 related symptoms and physical limitations across diuretic use subgroups, with especially
CC

18 pronounced benefits among patients receiving loop diuretics at baseline. Semaglutide led to

19 consistent beneficial effects on body weight, exercise function, and biomarkers of inflammation
A

20 and congestion, across the subgroups of background diuretic therapy use and dose. Semaglutide

21 was also consistently well tolerated—with fewer SAEs and cardiac disorders compared with

22 placebo—irrespective of baseline diuretic therapy or dose. Furthermore, during 52 weeks of

23 treatment, compared with placebo, semaglutide treatment led to: (1) a nearly 20% reduction in

15
1 total daily loop diuretic dose; (2) more than a 2-fold increase in odds of loop diuretic dose

2 reduction; and (3) 66% lower odds of loop diuretic dose increase. Semaglutide use was also

3 associated with less loop diuretic starts (in those not already on loop diuretics at baseline) and

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 more frequent stopping of loop diuretics (in those on loop diuretics at baseline) compared with

PT
5 placebo. In the semaglutide group, reductions in daily loop diuretic dose were also linearly

RI
6 correlated with improvements in health status, reduction in body weight, and reduction in

7 systemic inflammation (Structured Graphical Abstract). These findings demonstrate that

SC
8 semaglutide is effective across the full spectrum of patients with obesity-related HFpEF, from

9 those who do not require loop diuretics to those with significant congestion, requiring high-dose

U
10 loop diuretic therapy, often with adjunctive MRA and SGLT2 inhibitor use. The results of this

11
N
study also complement the main findings of the STEP-HFpEF program, which demonstrated that
A
12 semaglutide results in disease modification (early and sustained lowering of NT-proBNP; and
M

13 fewer HF events) by showing that semaglutide leads to clinically relevant and statistically
D

14 significant reduction in daily loop diuretic dose over time.


TE

15 Although loop diuretics have been the de facto the first-line decongestive treatment for

16 HF across the LVEF spectrum for over 60 years, they can cause electrolyte abnormalities,
EP

17 worsening kidney function, and hypotension.1 In obesity-related HFpEF, with the availability of

18 SGLT2 inhibitors, MRAs, and now semaglutide, the need for loop diuretics, particularly at
CC

19 higher doses, may need to be reconsidered and substituted for these agents as first-line therapies.

20
A

It is notable that in the STEP-HFpEF program, there were fewer cardiac-related SAEs in the

21 semaglutide group compared with placebo, along with decongestive effects suggested by greater

22 reductions in NT-proBNP, fewer HF events, and lower loop diuretic dose requirements.

16
1 The search for alternative decongestive therapies is especially important in patients with

2 obesity-related HFpEF because of the blunted response to loop diuretics in these patients

3 compared to those with HFpEF but no obesity, and the greater frequency of worsening kidney

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 function in patients with obesity-related HFpEF during decongestion.12 Reassuringly, the HF

PT
5 benefits and safety of semaglutide were consistent across diuretic use/dose groups; thus, even in

RI
6 patients with minimal congestion, semaglutide is still effective and well-tolerated. Nevertheless,

7 it is notable and clinically relevant that for health status (i.e., KCCQ domains), the largest

SC
8 improvements with semaglutide were seen in the most congested patients (i.e., those who

9 required the highest dose of loop diuretics and often were also taking MRAs), who are especially

U
10 difficult to manage and have few efficacious treatment options. These results also underscore the

11
N
need for novel non-loop diuretic therapies to effectively decongest patients with obesity-related
A
12 HFpEF given their propensity for inadequate or poorly tolerated response to loop diuretics. 12
M

13 The discordance between the greater KCCQ benefit in the most congested patients (i.e.,
D

14 those treated with loop diuretics at high doses) despite a similar degree of percent body weight
TE

15 reduction across the diuretic use/dose groups is of substantial importance. The mean placebo-

16 corrected weight loss was 8.3% vs. 6.9%, whereas the mean placebo-corrected improvement in
EP

17 KCCQ-CSS was 3.2 vs. 11.6 points in the no diuretics and highest-dose loop diuretic groups,

18 respectively. These findings, coupled with the reduction in loop diuretic dose over time and the
CC

19 linear relationship between loop diuretic dose reductions and improvements in KCCQ-CSS and

20
A

CRP in the semaglutide arm point to potential weight-independent effects of semaglutide on

21 decongestion. Mechanisms underlying these findings remain speculative; possibilities include:

22 (1) selective reduction in epicardial, pericardial, and chest wall adipose tissue (which would

23 reduce pericardial constraint that is present in obesity-related HFpEF, thereby lowering filling

17
1 pressure19 ); (2) weight loss-independent, direct effects of semaglutide on vasorelaxation (via

2 GLP1 receptors on vascular smooth muscle cells20,21 ); or (3) beneficial kidney effects of GLP1-

3 RAs, including reduced tubulointerstitial damage, albuminuria, and glomerulosclerosis, with

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 improved podocyte architecture seen in preclinical studies, 22 which may be reasons why

PT
5 semaglutide has led to improved kidney outcomes in several clinical trials, 23 including the

RI
6 FLOW trial of semaglutide vs. placebo in patients with diabetes and chronic kidney disease,

7 which was stopped early due to overwhelming efficacy. 24 It is possible that all of these effects

SC
8 may be more pronounced in those patients with the greatest level of congestion at baseline.

9 Several studies have examined the effects of various HF medications on loop diuretic

U
10 changes over time in both HFpEF and in HF with reduced ejection fraction. 25-33 Of the studied

11
N
HF therapies, MRAs and SGLT2 inhibitors have been the agents with most consistent beneficial
A
12 effects on the reduction in loop diuretic dose over time; however, these changes have been
M

13 modest, with lower percent reduction in doses and lower odds of dose decreases. 25-27,29-31 For
D

14 example, in the DELIVER trial, the SGLT2 inhibitor dapagliflozin did not lead to loop diuretic
TE

15 reductions (HR 0.98; 95% CI 0.86-1.13, P=0.83) during follow-up.27

16 Semaglutide-induced reduction in loop diuretic dose is particularly relevant given the


EP

17 association of outpatient intensification of diuretics with increased risk for subsequent adverse

18 HF events in patients with HF across the LVEF spectrum. 2-8,34 In addition, multiple studies have
CC

19 demonstrated that higher loop diuretic doses are a proxy for disease severity and are associated

20
A

with worse outcomes in patients with HF.35,36 Given the relationship between high baseline loop

21 diuretic doses and dose intensification with subsequent HF events, it is therefore not surprising

22 that there were fewer HF events observed with semaglutide versus placebo in the STEP-HFpEF

23 program (although the program was not primarily designed to evaluate clinical events). 15 The 52-

18
1 week duration of the STEP-HFpEF program precludes determination of whether the magnitude

2 of reduction in loop diuretic dose is associated with future HF events, and the number of HF

3 events in the overall STEP-HFpEF program was small. For these reasons, dedicated

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 cardiovascular outcome trials of incretin-based therapies, in patients with obesity-related HFpEF

PT
5 should be undertaken.

RI
6

7 Strengths and limitations

SC
8 Strengths of this study include the pre-specified nature of the analysis, including

9 harmonization of the endpoints and study procedures across the two trials in the STEP-HFpEF

U
10 program, and the detailed recording of diuretic use, types, and doses throughout both trials,

11
N
which allowed us to examine the effects of semaglutide across groups stratified by baseline
A
12 diuretic use/dose on a variety of clinically relevant endpoints; and also to determine the effects of
M

13 semaglutide vs. placebo on loop diuretic doses during the 52-week duration of the trial.
D

14 Limitations of this study include the analysis of loop diuretic dose changes in isolation, without
TE

15 considering changes in other medications. However, such analyses are challenging given the

16 polypharmacy and hyper-polypharmacy commonly present in HFpEF,37 which makes accounting


EP

17 for all medication changes methodologically difficult. We were also unable to compare among

18 the types of loop diuretics given the relatively small sample sizes, although prior studies in HF
CC

19 have found no differences in outcomes in relation to type of loop diuretic. 38 Nonetheless, our

20
A

analytic approach was consistent with several other secondary analyses of loop diuretic dose

21 changes in previous HF trials.25,27-33 The study is also limited by lack of direct mechanistic

22 insights regarding changes in plasma volume, natriuresis, and cardiac structure/function. The

23 STEP-HFpEF program does include an echocardiographic substudy, which may shed additional

19
1 light on the mechanisms behind the benefits of semaglutide in future analyses. Interaction testing

2 may be underpowered and therefore could have missed significant differences in efficacy and

3 safety among diuretic groups. Finally, the STEP-HFpEF program included a paucity of non-

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 White participants, thereby limiting generalizability.

PT
5

RI
6 Conclusions

7 In STEP-HFpEF, the first clinical trial program to examine the role of the GLP1-RA

SC
8 semaglutide in the management of patients with obesity-related HFpEF, the clinical

9 characteristics of patients differed by baseline diuretic use and type, but the majority of

U
10 beneficial HF-related clinical effects and safety of semaglutide were consistent across diuretic

11
N
groups, with greater magnitude of improvement in HF-related symptoms and physical limitations
A
12 in patients taking loop diuretics. Semaglutide treatment led to a clinically meaningful and
M

13 significant reduction in loop diuretic dose over the 52-week treatment period, which along with
D

14 reductions in NT-proBNP and fewer HF events, suggests disease-modifying effects in obesity-


TE

15 related HFpEF.

16
EP

17 Declarations
CC

18 Funding

19 The STEP-HFpEF program was funded by Novo Nordisk.


A

20

21 Acknowledgements

22 We thank the STEP-HFpEF program trial participants, site investigators, and site staff.

23 Sanjiv J Shah, Kavita Sharma and Mikhail N. Kosiborod wrote the first draft of the manuscript.

20
1 Administrative support was provided by Casey McKeown RVN, FdSc of Apollo, OPEN Health

2 Communications, and funded by Novo Nordisk A/S, in accordance with Good Publication

3 Practice guidelines (https://www.ismpp.org/gpp-2022).

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4

PT
5 Disclosure of Interest

RI
6 S.J.S. was supported by research grants from the U.S. National Institutes of Health (NIH;

7 U54 HL160273; R01 HL140731; and R01 HL149423); has received research funding from

SC
8 AstraZeneca, Corvia, and Pfizer; and has received consulting fees from Abbott, Alleviant,

9 Amgen, Aria CV, AstraZeneca, Axon Therapies, Bayer, Boehringer Ingelheim, Boston Scientific,

U
10 BridgeBio, Bristol Myers Squibb, Corvia, Cytokinetics, Edwards Lifesciences, Eidos, Imara,

11
N
Impulse Dynamics, Intellia, Ionis, Lilly, Merck, NGM Biopharmaceuticals, Novartis, Novo
A
12 Nordisk, Pfizer, Prothena, Regeneron, Rivus, Sardocor, Shifamed, Tenax, Tenaya, and Ultromics.
M

13 K.S. is an advisory board member and consultant for Alleviant, AstraZeneca, Bayer, Boehringer
D

14 Ingelheim, Bristol Myers Squibb, Cytokinetics, Janssen, Novartis, Novo Nordisk, and Rivus, and
TE

15 receives honoraria.

16 B.A.B. receives research support from the National Institutes of Health (NIH) and the United
EP

17 States Department of Defense, as well as research grant funding from AstraZeneca, Axon,

18 GlaxoSmithKline, Medtronic, Mesoblast, Novo Nordisk, Rivus, and Tenax Therapeutics; has
CC

19 served as a consultant for Actelion, Amgen, Aria, Axon Therapies, BD, Boehringer Ingelheim,

20
A

Cytokinetics, Edwards Lifesciences, Eli Lilly, Imbria, Janssen, Merck, NGM, Novo Nordisk,

21 NXT, and VADovations; and is named inventor (US patent no. 10,307,179) for the tools and

22 approach for a minimally invasive pericardial modification procedure to treat heart failure.

21
1 J.B. is a consultant to 3live, Abbott, American Regent, Amgen, Applied Therapeutics,

2 AstraZeneca, Bayer, Boehringer Ingelheim, Bristol Myers Squibb, Cardiac Dimension, Cardior,

3 CVRx, Cytokinetics, Edwards, Element Science, Impulse Dynamics, Imbria, Innolife, Inventiva,

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 Janssen, Lexicon, Lilly, LivaNova, Medtronics, Merck, Novartis, Novo Nordisk, Occlutech,

PT
5 Pfizer, Pharmacosmos, PharmaIN, Roche, Sequana, SQ Innovation, and Vifor.

RI
6 M.J.D. has acted as consultant, advisory board member, and speaker for Boehringer Ingelheim,

7 Eli Lilly, Novo Nordisk, and Sanofi; an advisory board member and speaker for AstraZeneca; an

SC
8 advisory board member for Medtronic, Pfizer, and ShouTi Pharma; and speaker for Amgen,

9 Novartis, and Sanofi. M.J.D. has received grants as an investigator in support of investigator-

U
10 initiated trials from AstraZeneca, Boehringer Ingelheim, Eli Lilly, Janssen, Novo Nordisk, and

11 Sanofi-Aventis.
N
A
12 D.W.K. was supported in part by the Kermit Glenn Phillips II Chair in Cardiovascular Medicine
M

13 and NIH grants U01AG076928; R01AG078153; R01AG045551; R01AG18915; P30AG021332;


D

14 U24AG059624; and U01HL160272; and reports receiving honoraria as a consultant for


TE

15 AstraZeneca, Bayer, Boehringer Ingelheim, Corvia Medical, Ketyo, Novartis, Novo Nordisk,

16 Pfizer, and Rivus, grant funding from AstraZeneca, Bayer, Novartis, Novo Nordisk, Pfizer, and
EP

17 Rivus, and has stock ownership in Gilead Sciences.

18 M.C.P. has received research funding from AstraZeneca, Boehringer Ingelheim, Boston
CC

19 Scientific, Medtronic, Novo Nordisk, Novartis, Pharmacosmos, Roche, and SQ Innovations, and

20
A

served on committees or consulted for AbbVie, Akero, AnaCardio, Applied Therapeutics,

21 AstraZeneca, Bayer, Biosensors, Boehringer Ingelheim, Cardiorentis, Corvia, Eli Lilly, Horizon

22 Therapeutics, LIB Therapeutics, Moderna, New Amsterdam, Novartis, Novo Nordisk,

23 Pharmacosmos, Siemens, SQ Innovations, Takeda, Teikoku, and Vifor.

22
1 S.V. reports speaking honoraria and/or consulting fees from Abbott, Amarin, AstraZeneca, Bayer,

2 Boehringer Ingelheim, Canadian Medical and Surgical Knowledge Translation Research Group,

3 Eli Lilly, HLS Therapeutics, Janssen, Merck, Novartis, Novo Nordisk, Pfizer, PhaseBio, and

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 TIMI.

PT
5 S.P. reports no conflicts of interest.

RI
6 K.M.C. reports no conflicts of interest.

7 M.N.E. is an employee of Novo Nordisk A/S and a shareholder.

SC
8 T.J.J. is an employee of Novo Nordisk A/S and a shareholder.

9 S.R. is an employee of Novo Nordisk A/S and a shareholder.

U
10 R.A. reports no conflicts of interest.

11 T.B. reports no conflicts of interest.


N
A
12 M.N.K. served as a consultant or on an advisory board for 35Pharma, Alnylam, Amgen, Applied
M

13 Therapeutics, AstraZeneca, Bayer, Boehringer Ingelheim, Cytokinetics, Dexcom, Eli Lilly,


D

14 Esperion Therapeutics, Imbria Pharmaceuticals, Janssen, Lexicon Pharmaceuticals, Merck


TE

15 (Diabetes and Cardiovascular), Novo Nordisk, Pharmacosmos, Pfizer, Sanofi,

16 scPharmaceuticals, Structure Therapeutics, Vifor Pharma, and Youngene Therapeutics; has


EP

17 received research grants from AstraZeneca, Boehringer Ingelheim, and Pfizer; holds stocks in

18 Artera Health and Saghmos Therapeutics; and has received honoraria from AstraZeneca,
CC

19 Boehringer Ingelheim, and Novo Nordisk. He has also received other research support from

20
A

AstraZeneca.

21

22

23
1 Data Availability Statement

2 Data will be shared with bona fide researchers submitting a research proposal approved by the

3 independent review board. Access request proposals can be found at novonordisk-trials.com.

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 Data will be made available after research completion, and approval of the product and product

PT
5 use in the European Union and the USA. Individual participant data will be shared in data sets in

RI
6 a de-identified/anonymized format.

SC
8 REFERENCES
9
10 1. Pius R, Odukudu GO, Olorundare I, Makanjuola DI, Komolafe R, Njoku C, et al. A

U
11 Narrative Review on the Efficacy and Safety of Loop Diuretics in Patients With Heart

12 N
Failure With Reduced Ejection Fraction and Preserved Ejection Fraction. Cureus
A
13 2023;15:e45794. https://doi.org/10.7759/cureus.45794
M

14 2. Chatur S, Vaduganathan M, Claggett BL, Cunningham JW, Docherty KF, Desai AS, et al.
D

15 Outpatient Worsening Among Patients With Mildly Reduced and Preserved Ejection
TE

16 Fraction Heart Failure in the DELIVER Trial. Circulation 2023;148:1735-1745.

17 https://doi.org/10.1161/circulationaha.123.066506
EP

18 3. Ferreira JP, Liu J, Claggett BL, Vardeny O, Pitt B, Pfeffer MA, et al. Outpatient diuretic
CC

19 intensification as endpoint in heart failure with preserved ejection fraction trials: an

20 analysis from TOPCAT. Eur J Heart Fail 2022;24:378-384.


A

21 https://doi.org/10.1002/ejhf.2376

22 4. Greene SJ, Butler J. Expanding the Definition of Worsening Heart Failure and

23 Recognizing the Importance of Outpatient Escalation of Oral Diuretics. Circulation

24 2023;148:1746-1749. https://doi.org/10.1161/circulationaha.123.066915

24
1 5. Greene SJ, Mentz RJ, Felker GM. Outpatient Worsening Heart Failure as a Target for

2 Therapy: A Review. JAMA cardiology 2018;3:252-259.

3 https://doi.org/10.1001/jamacardio.2017.5250

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


PT
4 6. Khan MS, Butler J, Greene SJ. Recognizing the Significance of Outpatient Worsening

5 Heart Failure. J Am Heart Assoc 2020;9:e017485.

RI
6 https://doi.org/10.1161/jaha.120.017485

SC
7 7. Madelaire C, Gustafsson F, Stevenson LW, Kristensen SL, Køber L, Andersen J, et al.

8 One-Year Mortality After Intensification of Outpatient Diuretic Therapy. Journal of the

U
9 American Heart Association 2020;9:e016010-e016010.

10 https://doi.org/10.1161/JAHA.119.016010
N
A
M

11 8. Okumura N, Jhund PS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, et al.

12 Importance of Clinical Worsening of Heart Failure Treated in the Outpatient Setting:


D

13 Evidence From the Prospective Comparison of ARNI With ACEI to Determine Impact on
TE

14 Global Mortality and Morbidity in Heart Failure Trial (PARADIGM-HF). Circulation

15 2016;133:2254-2262. https://doi.org/10.1161/circulationaha.115.020729
EP

16 9. Wierda E, Dickhoff C, Handoko ML, Oosterom L, Kok WE, de Rover Y, et al. Outpatient
CC

17 treatment of worsening heart failure with intravenous and subcutaneous diuretics: a

18 systematic review of the literature. ESC Heart Fail 2020;7:892-902.


A

19 https://doi.org/10.1002/ehf2.12677

25
1 10. Litwin SE, Komtebedde J, Seidler T, Borlaug BA, Winkler S, Solomon SD, et al. Obesity

2 in heart failure with preserved ejection fraction: Insights from the REDUCE LAP-HF II

3 trial. Eur J Heart Fail 2024;26:177-189. https://doi.org/10.1002/ejhf.3092

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


PT
4 11. Morgen CS, Haase CL, Oral TK, Schnecke V, Varbo A, Borlaug BA. Obesity,

5 Cardiorenal Comorbidities, and Risk of Hospitalization in Patients With Heart Failure

RI
6 With Preserved Ejection Fraction. Mayo Clin Proc 2023;98:1458-1468.

SC
7 https://doi.org/10.1016/j.mayocp.2023.07.008

8 12. Reddy YNV, Obokata M, Testani JM, Felker GM, Tang WHW, Abou-Ezzeddine OF, et al.

U
9 Adverse Renal Response to Decongestion in the Obese Phenotype of Heart Failure With

10
N
Preserved Ejection Fraction. J Card Fail 2020;26:101-107.
A
11 https://doi.org/10.1016/j.cardfail.2019.09.015
M

12 13. Kosiborod MN, Abildstrøm SZ, Borlaug BA, Butler J, Rasmussen S, Davies M, et al.
D

13 Semaglutide in patients with heart failure with preserved ejection fraction and obesity. N
TE

14 Engl J Med 2023;389:1069–1084. https://doi.org/10.1056/NEJMoa2306963


EP

15 14. Kosiborod MN, Petrie MC, Borlaug BA, Butler J, Davies MJ, Kees Hovingh G, et al.

16 Semaglutide in patients with obesity-related heart failure with preserved ejection fraction
CC

17 and type 2 diabetes. N Engl J Med 2024;(in press).


A

18 15. Butler J, Shah SJ, Petrie MC, Borlaug BA, Abildstrom SZ, Davies M, et al. Semaglutide

19 and obesity-related heart failure with preserved ejection fraction: A pooled analysis of the

20 STEP-HFpEF and STEP-HFpEF DM randomised trials. The Lancet 2024;(in press).

26
1 16. Kosiborod MN, Abildstrøm SZ, Borlaug BA, Butler J, Christensen L, Davies M, et al.

2 Design and baseline characteristics of STEP-HFpEF program evaluating semaglutide in

3 patients with obesity HFpEF phenotype. JACC Heart Fail 2023;11:1000–1010.

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 https://doi.org/10.1016/j.jchf.2023.05.010

PT
5 17. Brater DC. Diuretic therapy. N Engl J Med 1998;339:387-395.

RI
6 https://doi.org/10.1056/NEJM199808063390607

SC
7 18. Pocock SJ, Ariti CA, Collier TJ, Wang D. The win ratio: a new approach to the analysis

8 of composite endpoints in clinical trials based on clinical priorities. European Heart

U
9 Journal 2011;33:176-182. https://doi.org/10.1093/eurheartj/ehr352
N
A
10 19. Obokata M, Reddy YNV, Pislaru SV, Melenovsky V, Borlaug BA. Evidence Supporting
M

11 the Existence of a Distinct Obese Phenotype of Heart Failure With Preserved Ejection

12 Fraction. Circulation 2017;136:6-19.


D

13 https://doi.org/10.1161/CIRCULATIONAHA.116.026807
TE

14 20. Pyke C, Heller RS, Kirk RK, Orskov C, Reedtz-Runge S, Kaastrup P, et al. GLP-1
EP

15 receptor localization in monkey and human tissue: novel distribution revealed with

16 extensively validated monoclonal antibody. Endocrinology 2014;155:1280-1290.


CC

17 https://doi.org/10.1210/en.2013-1934
A

18 21. Jensen EP, Poulsen SS, Kissow H, Holstein-Rathlou NH, Deacon CF, Jensen BL, et al.

19 Activation of GLP-1 receptors on vascular smooth muscle cells reduces the

20 autoregulatory response in afferent arterioles and increases renal blood flow. Am J

21 Physiol Renal Physiol 2015;308:F867-877. https://doi.org/10.1152/ajprenal.00527.2014

27
1 22. Wilbon SS, Kolonin MG. GLP1 Receptor Agonists-Effects beyond Obesity and Diabetes.

2 Cells 2023;13. https://doi.org/10.3390/cells13010065

3 23. Heerspink HJL, Apperloo E, Davies M, Dicker D, Kandler K, Rosenstock J, et al. Effects

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


PT
4 of semaglutide on albuminuria and kidney function in people with overweight or obesity

5 with or without type 2 diabetes: exploratory analysis from the STEP 1, 2, and 3 trials.

RI
6 Diabetes Care 2023;46:801–810. https://doi.org/10.2337/dc22-1889

SC
7 24. Gragnano F, De Sio V, Calabrò P. FLOW trial stopped early due to evidence of renal

8 protection with semaglutide. Eur Heart J Cardiovasc Pharmacother 2024;10:7-9.

U
9 https://doi.org/10.1093/ehjcvp/pvad080
N
A
10 25. Butler J, Usman MS, Filippatos G, Ferreira JP, Böhm M, Brueckmann M, et al. Safety
M

11 and Efficacy of Empagliflozin and Diuretic Use in Patients with Heart Failure and

12 Preserved Ejection Fraction: A Post Hoc Analysis of the EMPEROR-Preserved Trial.


D

13 JAMA Cardiol 2023;8:640-649. https://doi.org/10.1001/jamacardio.2023.1090


TE

14 26. Cavagnini ME, Best EE, Skersick PT, Truitt KP, Musick KL, Mangum BR, et al. Impact
EP

15 of Inpatient Initiation of SGLT2 Inhibitors on Diuretic Requirements in Patients With

16 Heart Failure. J Pharm Pract 2023:8971900231159739.


CC

17 https://doi.org/10.1177/08971900231159739
A

18 27. Chatur S, Vaduganathan M, Claggett B, Vardeny O, Desai AS, Jhund PS, et al.

19 Dapagliflozin and diuretic utilization in heart failure with mildly reduced or preserved

20 ejection fraction: the DELIVER trial. Eur Heart J 2023;44:2930-2943.

21 https://doi.org/10.1093/eurheartj/ehad283

28
1 28. Ezekowitz J, Alemayehu W, Edelmann F, Ponikowski P, Lam CSP, O'Connor CM, et al.

2 Diuretic use and outcomes in patients with heart failure with reduced ejection fraction:

3 Insights from the VICTORIA trial. Eur J Heart Fail 2024.

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 https://doi.org/10.1002/ejhf.3179

PT
5 29. Ferreira JP, Eschalier R, Duarte K, Damman K, Gustafsson F, Schou M, et al. Reduced

RI
6 Diuretic Dose in Patients Treated With Eplerenone. Circulation: Heart Failure 2020;13.

SC
7 https://doi.org/10.1161/circheartfailure.119.006597

8 30. Jackson AM, Dewan P, Anand IS, Belohlavek J, Bengtsson O, de Boer RA, et al.

U
9 Dapagliflozin and Diuretic Use in Patients With Heart Failure and Reduced Ejection

10
N
Fraction in DAPA-HF. Circulation 2020;142:1040-1054.
A
11 https://doi.org/10.1161/CIRCULATIONAHA.120.047077
M

12 31. Kalogeropoulos AP, Thankachen J, Butler J, Fang JC. Diuretic and renal effects of
D

13 spironolactone and heart failure hospitalizations: a TOPCAT Americas analysis. Eur J


TE

14 Heart Fail 2020;22:1600-1610. https://doi.org/10.1002/ejhf.1917


EP

15 32. Kido K, Bianco C, Caccamo M, Hashiguchi M, Choo LY, Sokos G. Sacubitril/Valsartan

16 Does Not Change the Use and Dose of Loop Diuretics in Patients With Heart Failure
CC

17 With Reduced Ejection Fraction. J Pharm Pract 2023:8971900231177202.

18 https://doi.org/10.1177/08971900231177202
A

19 33. Vardeny O, Claggett B, Kachadourian J, Desai AS, Packer M, Rouleau J, et al. Reduced

20 loop diuretic use in patients taking sacubitril/valsartan compared with enalapril: the

29
1 PARADIGM-HF trial. Eur J Heart Fail 2019;21:337-341.

2 https://doi.org/10.1002/ejhf.1402

3 34. Docherty KF, Jhund PS, Anand I, Bengtsson O, Böhm M, de Boer RA, et al. Effect of

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


PT
4 Dapagliflozin on Outpatient Worsening of Patients With Heart Failure and Reduced

5 Ejection Fraction: A Prespecified Analysis of DAPA-HF. Circulation 2020;142:1623-

RI
6 1632. https://doi.org/10.1161/CIRCULATIONAHA.120.047480

SC
7 35. Kapelios CJ, Laroche C, Crespo-Leiro MG, Anker SD, Coats AJS, Diaz-Molina B, et al.

8 Association between loop diuretic dose changes and outcomes in chronic heart failure:

U
9 observations from the ESC-EORP Heart Failure Long-Term Registry. Eur J Heart Fail

10
N
2020;22:1424-1437. https://doi.org/10.1002/ejhf.1796
A
M

11 36. Shah RV, McNulty S, O'Connor CM, Felker GM, Braunwald E, Givertz MM. Effect of

12 admission oral diuretic dose on response to continuous versus bolus intravenous diuretics
D

13 in acute heart failure: an analysis from diuretic optimization strategies in acute heart
TE

14 failure. Am Heart J 2012;164:862-868. https://doi.org/10.1016/j.ahj.2012.08.019


EP

15 37. Minamisawa M, Claggett B, Suzuki K, Hegde SM, Shah AM, Desai AS, et al.

16 Association of Hyper-Polypharmacy With Clinical Outcomes in Heart Failure With


CC

17 Preserved Ejection Fraction. Circ Heart Fail 2021;14:e008293.

18 https://doi.org/10.1161/CIRCHEARTFAILURE.120.008293
A

19 38. Mentz RJ, Anstrom KJ, Eisenstein EL, Sapp S, Greene SJ, Morgan S, et al. Effect of

20 Torsemide vs Furosemide After Discharge on All-Cause Mortality in Patients

30
1 Hospitalized With Heart Failure: The TRANSFORM-HF Randomized Clinical Trial.

2 JAMA 2023;329:214-223. https://doi.org/10.1001/jama.2022.23924

3 FIGURE LEGENDS

Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024


4 Figure 1. Restricted cubic spline curves of key efficacy outcomes over the range of baseline daily loop

PT
5 diuretic dose pooled across the STEP-HFpEF and STEP-HFpEF-DM trials.
6 Loop diuretic dose expressed in mg furosemide equivalents per day. 6MWD, 6-min walk distance; CRP,
7 C-reactive protein; KCCQ-CSS, Kansas City Cardiomyopathy Questionnaire clinical summary score; NT-

RI
8 proBNP, N-terminal pro-B-type natriuretic peptide; w, week.
9 Figure 2. Effect of semaglutide vs. placebo on loop diuretic dose from baseline to 52 weeks, pooled

SC
10 across the STEP-HFpEF and STEP-HFpEF-DM trials.

11 Error bars represent standard deviations. There was no significant difference in baseline loop diuretic dose

U
12 between the semaglutide and placebo groups (P=0.19).

13 N
Figure 3. Effect of semaglutide vs. placebo on loop diuretic dose changes from baseline to 52 weeks,
A
14 pooled across the STEP-HFpEF and STEP-HFpEF-DM trials.
M

15 Odds of loop diuretic dose changes over 52 weeks in response to treatment with semaglutide 2.4 mg: dose

16 increase (OR 0.34 [95% CI 0.23-0.52]), P<0.001; dose decrease (OR 2.09 [95% CI 1.39-3.15], P<0.001).
D

17
TE

18 Figure 4. Effects of semaglutide versus placebo on the time to first loop diuretic start among non-users at

19 baseline (A) and time to first loop diuretic stop in users at baseline (B), pooled across the STEP-HFpEF
EP

20 and STEP-HFpEF-DM trials

21
CC

22 Figure 5. Correlations of changes in loop diuretic dose vs. changes in efficacy endpoints, stratified by

23 treatment group, pooled across the STEP-HFpEF and STEP-HFpEF-DM trials.


A

24 R-values represent Pearson correlation coefficients. 6MWD, 6-min walk distance; CRP, C-reactive

25 protein; KCCQ-CSS, Kansas City Cardiomyopathy Questionnaire clinical summary score; NT-proBNP,

26 N-terminal pro-B-type natriuretic peptide; w, week.

27

31
Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024

32
PT
RI
SC
U
N
A
Figure 1
M
D
TE
EP
CC
A

1
2

3
4

5
Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024

33
PT
RI
SC
U
N
A

Figure 4
M
D
TE
EP
CC
A

2
3
Downloaded from https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehae322/7670783 by guest on 25 May 2024

34
PT
RI
SC
U
N
A
Figure 5
M
D
TE
EP
CC
A

1
2

You might also like