You are on page 1of 10

Circulation

ORIGINAL RESEARCH ARTICLE

Effects of Early Empagliflozin Initiation on Diuresis


and Kidney Function in Patients With Acute
Decompensated Heart Failure (EMPAG-HF)
P. Christian Schulze , MD, PhD; Jürgen Bogoviku, MD; Julian Westphal , MD; Pawel Aftanski, MD; Franz Haertel, MD;
Sissy Grund, MS; Stephan von Haehling, MD, PhD; Ulrike Schumacher, PhD; Sven Möbius-Winkler , MD, PhD; Martin Busch, MD

BACKGROUND: Effective diuretic regimens using loop diuretics in patients with acute decompensated heart failure are often
limited by the development of worsening kidney function. Sodium-glucose cotransporter-2 inhibitors induce glucosuria and
sodium excretion with nephroprotective effects in patients with stable heart failure but their role in acute decompensated
heart failure is unclear.

METHODS: In this single-center, prospective, double-blind, placebo-controlled, randomized study, we randomly assigned patients
with acute decompensated heart failure to empagliflozin 25 mg daily or placebo in addition to standard decongestive treatments
that included loop diuretics. The primary end point was cumulative urine output over 5 days. Secondary end points included
diuretic efficiency, dynamics in markers of kidney function and injury, and NT-proBNP (N-terminal pro-B-type natriuretic peptide).

RESULTS: Sixty patients were randomized within 12 hours of hospitalization for acute decompensated heart failure. Addition of
empagliflozin daily to standard medical treatment of acute decompensated heart failure resulted in a 25% increase in cumulative
urine output over 5 days (median 10.8 versus 8.7 L mL in placebo, group difference estimation 2.2 L [95% CI, 8.4 to 3.6];
P=0.003). Empagliflozin increased diuretic efficiency compared with placebo (14.1 mL urine per milligram furosemide equivalent
Downloaded from http://ahajournals.org by on July 8, 2023

[95% CI, 0.6–27.7]; P=0.041) without affecting markers of renal function (estimated glomerular filtration rate, 51±19 versus
54±17 mL/min per 1.73 m²; P=0.599) or injury (total urinary protein, 492±845 versus 503±847 mg/g creatinine; P=0.975;
and urinary α1-microglobulin, 55.4±38.6 versus 31.3±33.6 mg/g creatinine; P=0.066) with more pronounced decrease in NT-
proBNP in the empagliflozin group compared with placebo (−1861 versus −727.2 pg/mL after 5 days; quotient in slope, 0.89
[95% CI, 0.83–0.95]; P<0.001). There were no differences in the incidence of safety events between groups.

CONCLUSIONS: Early addition of empagliflozin to standard diuretic therapy increases urine output without affecting renal
function in patients with acute decompensated heart failure.

REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT04049045.

Key Words: diuretics ◼ heart failure ◼ kidney

Editorial, see p 299

H
eart failure (HF) has increased in incidence and impaired kidney function and diuretic resistance.3 The
prevalence worldwide and has high morbidity and only pharmacologic treatments available to augment
mortality.1,2 Acute decompensated HF (ADHF) diuresis are various combinations of diuretics added to
is a severe presentation of HF characterized by vol- loop diuretics or the addition of inotropes or vasodila-
ume retention and congestion, often accompanied by tors when decompensation is severe.2–4 The inability to

Correspondence to: P. Christian Schulze, MD, PhD, Department of Internal Medicine I, Division of Cardiology, University Hospital Jena, Am Klinikum 1, 07743 Jena,
Germany. Email christian.schulze@med.uni-jena.de
Supplemental Material, the podcast, and transcript are available with this article at https://www.ahajournals.org/doi/suppl/10.1161/CIRCULATIONAHA.122.059038.
Continuing medical education (CME) credit is available for this article. Go to http://cme.ahajournals.org to take the quiz.
For Sources of Funding and Disclosures, see page 297.
© 2022 American Heart Association, Inc.
Circulation is available at www.ahajournals.org/journal/circ

Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038 July 26, 2022 289


Schulze et al Empagliflozin in ADHF

benefit of these agents on outcomes after discharge,


Clinical Perspective highlighting a critical unmet need.5–8 The addition of
ORIGINAL RESEARCH

natriuretic peptides (nesiritide or ularitide),6,8 vasopres-


What Is New? sin antagonism,7,9 or ultrafiltration10,11to standard diuretic
ARTICLE

• The addition of synthetic natriuretic peptides, vaso- regimens have not shown consistent clinical benefit in
pressin antagonism, or ultrafiltration to standard patients with chronic HF or ADHF. Therefore, novel strat-
diuretic regimens has failed to improve outcomes in egies with the potential to improve diuresis in ADHF rep-
chronic or acute decompensated heart failure. resent a major therapeutic need.
• No new therapies for acute decompensated heart Sodium-glucose cotransporter-2 (SGLT2) inhibitors
failure, including diuretic drugs, have been intro- reduce the risk of cardiovascular death and hospitaliza-
duced in decades.
tion in ambulatory patients with stable chronic HF regard-
• The EMPAG-HF study (Empagliflozin in Acute
less of ejection fraction.12–15 However, the clinical effects
Decompensated Heart Failure) was designed
to test the hypothesis that early sodium-glucose of an early start of SGLT2 inhibition within the first 12
cotransporter-2 inhibition with empagliflozin added hours after clinical presentation in addition to standard
to standard medical therapy enhances diuresis loop diuretic–based decongestion strategies have not
without furthering kidney injury in patients with been studied in patients with ADHF. The EMPAG-HF
acute decompensated heart failure. study (Empagliflozin in Acute Decompensated Heart
Failure) was designed to test the hypothesis that early
What Are the Clinical Implications? SGLT2 inhibition using empagliflozin plus standard medi-
• Our study shows that in patients with acute decom- cal therapy enhances diuresis without furthering kidney
pensated heart failure, early initiation of the sodium- injury in patients with ADHF.
glucose cotransporter-2 inhibitor empagliflozin
within 12 hours of hospital presentation in addition
to standard treatment is safe and increases urine METHODS
output without affecting kidney function or injury
patterns. Study Design and Oversight
• EMPAG-HF is the first randomized study to show The EMPAG-HF study (URL: https://www.clinicaltrials.gov;
primary clinical benefits of a drug regimen added to Unique identifier: NCT04049045) randomly assigned patients
diuretic regimens in patients with acute decompen- with ADHF to receive empagliflozin or placebo in a double-blind,
placebo-controlled trial. The SGLT2 inhibitor empagliflozin at a
Downloaded from http://ahajournals.org by on July 8, 2023

sated heart failure.


• These observations suggest that addition of dose of 25 mg daily was administered within 12 hours of hospi-
sodium-glucose cotransporter-2 inhibition to stan- talization for 5 days compared with placebo added to standard
dard diuretic therapy is a promising strategy to decongestive treatment.
assist with conventional decongestive treatment in The local ethics committee of the University of Jena
patients with acute decompensated heart failure. approved the protocol. All patients gave written informed
consent. All authors had full access to the trial data and take
responsibility for its integrity and the data analysis. The data
that support the findings of the current study are available from
Nonstandard Abbreviations and Acronyms the corresponding author on reasonable request.

ADHF acute decompensated heart failure


AE adverse event Patient Cohort
BNP brain natriuretic peptide Patients with ADHF (18 to 85 years of age) with BNP (brain
natriuretic peptide) >100 pg/mL or NT-proBNP (N-terminal
CKD-EPI Chronic Kidney Disease Epidemiology
pro-BNP) >300 pg/mL were included. Patients with or
Collaboration
without type 2 diabetes or impaired glucose tolerance were
eGFR estimated glomerular filtration rate included. Patients did not have cognitive impairment. For
EMPAG-HF Empagliflozin in Acute Decompen- women of childbearing potential, a negative pregnancy test or
sated Heart Failure documentation of the correct use of a highly effective contra-
HF heart failure ceptive method was required.
NT-proBNP N-terminal probrain natriuretic peptide Patients were excluded for the following: type 1 diabetes,
chronic kidney disease with estimated glomerular filtration
SAE serious adverse event
rate (eGFR) <30 mL/min per 1.73 m² or end-stage kidney
SGLT2 sodium-glucose cotransporter-2 failure with the need for chronic dialysis treatment, acute kid-
ney injury (Acute Kidney Injury Network stage ≥2 or requiring
dialysis treatment), current medication with SGLT2 inhibitors,
achieve decongestion is associated with a worse prog- known intolerance or hypersensitivity to the active substance
nosis and a higher rate of rehospitalization for ADHF.2,3 empagliflozin or a contraindication or intolerance to furosemide,
Several randomized controlled trials testing novel acute HF without signs of congestion (“dry” case), indication
pharmacotherapies in patients with ADHF failed to show for urgent coronary angiography or any planned administration

290 July 26, 2022 Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038


Schulze et al Empagliflozin in ADHF

of an iodine-based contrast agent within the next 6 days, need drug exposure, and safety, laboratory, and efficacy measures
for hemofiltration or any other form of extracorporeal therapy, according to the intention-to-treat principle.

ORIGINAL RESEARCH
planned surgery, or recent participation in another clinical trial Patients without major protocol deviations were included
(within the 3 months before inclusion). Any cause of HF leading in the per-protocol analysis. A sensitivity analysis was per-

ARTICLE
to decompensation that needed urgent management (eg, acute formed for selected demographic and baseline characteris-
coronary syndrome, unstable arrhythmias, mechanical causes, tics (ie, age, sex), primary efficacy, and selected secondary
acute pulmonary embolism), inability to understand or provide efficacy measures (ie, daily urine volume and the diuretics-
written informed consent, or documented alcohol abuse (daily corrected urine volume).
alcohol intake of >12 g in women or >24 g in men) also led to A per-protocol analysis of the primary variable was per-
exclusion from the study. formed for patients without major protocol deviations as a
sensitivity analysis. Results of this analysis were in line with
the analysis on the basis of the intention-to-treat popula-
Primary and Secondary Outcomes tion. To keep the definition of the per-protocol analysis as
The primary outcome of EMPAG-HF was the total urine unaffected as possible, predictable protocol deviations (eg,
output measured and summed over 5 days. Secondary end time window deviations) were classified a priori as minor
points included markers of kidney function under treatment or major.
(increase in creatinine of >0.3 mg/dL, doubling of serum cre- The safety analysis set included all patients included in
atinine, need for renal replacement therapy) and the trajectory the full analysis. For safety purposes, patients were analyzed
of eGFR, cystatin-C, and changes in baseline kidney function. as treated.
Net urine output was assessed daily and urine output was For all data assessed, descriptive statistics including mean,
also controlled for diuretic dosage according to Clark et al.16 SD, minimum, quartiles with median and maximum for metric
and Oh and Han.4 Additional end points included worsening or data, and frequency analysis for nonmetric data were evaluated.
persistent HF defined by New York Heart Association class; For selected end points, change from baseline was evaluated.
patient-reported outcomes assessed by a visual analog scale Volume assessments sum values over the complete day 1 (ie,
for health status and a quality of life questionnaire (EQ-5D reflect effect of first treatment). Therefore, for these values, no
index)17; intermediate care, intensive care unit, and hospi- baseline is available. Comparison of treatment groups is on the
tal length of stay; markers of liver function (bilirubin, serum basis of time point values. Analysis was performed by treatment
aminotransferases, and relevant change in coagulation sta- group and in total.
tus); NT-proBNP; oxygen saturation without oxygen therapy
or need for oxygen, in liters per minute; presence of rales;
changes in chest X-ray; and number of patients alive at dis- Sample Size and Power Considerations
Downloaded from http://ahajournals.org by on July 8, 2023

charge and out of hospital after 30 days. A sample size of 52 participants (26 per treatment arm) was
estimated to provide a power of 80% at a 2-sided α level of
0.05 to detect a difference in urine volume between the 2
Safety End Points groups if the effect size is 0.8 assuming normal distribution
Prespecified safety end points included serious adverse events of means. To account for an attrition rate of ≈10% to 15%, 4
(SAEs), events related to study drug discontinuation, and participants per group were added, resulting in a total sample
adverse events (AEs) related to deteriorating kidney function. size of 60 patients (30 per treatment arm).
Other outcome measures included the number of AEs and Calculation of sample size and 80% power analysis was
SAEs including MedDRA SAE preferred terms and system and performed with nQuery Advisor 7.0.
organization controls in both groups within 30 days. All AEs
and SAEs including laboratory measures were documented
except for efficacy measures. Analysis of the Primary End Point
The clinical event adjudication committee consisted of V5 denotes the total urine volume over the study days 1 to 5:
3 independent clinicians blinded to study arm assignment V5E for the empagliflozin treatment group and V5P for the pla-
(Supplemental Material). The committee reviewed abstracted cebo treatment group.
clinical data to determine whether major events occurred. The null hypothesis H0: V5E=V5P was tested against the
alternative H0: V5E≠V5P by means of a nonparametric Mann-
Whitney U test, α=0.05, 2-sided. The data distribution was
Monitoring and Follow-Up checked by graphical means.
Blood and spot urine samples were taken daily from day 1 to Results of 2 patients with missing information on urine vol-
5, at hospital discharge, and at day 30 after hospital discharge ume at day 5 were regarded as missing data.
(safety end point). All laboratory values were measured at the
central laboratory of the institution. eGFR was on the basis
of serum creatinine and calculated using the Chronic Kidney Analysis of Secondary End Points
Disease Epidemiology Collaboration (CKD-EPI) equation. The diuretics-corrected daily urine volume (V1D) was evaluated
for every patient and day according to the following formula,
where V1 denotes the daily urine volume, Vnorm the standard
Statistical Analysis output of urine in the given patient population, D the daily body
All treated patients were included in the full analysis set and weight–adopted dose of furosemide, and Dmin the minimal daily
were analyzed as randomized with regard to demographic body weight–adopted dose of furosemide:
characteristics, protocol deviations, baseline characteristics, V1D=(V−Vnorm)/D at the respective day

Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038 July 26, 2022 291


Schulze et al Empagliflozin in ADHF

We assumed Vnorm=1.8 L according to Clark et al.16 and Baseline demographic characteristics of random-
Dmin=10 mg/70 kg body weight according to Oh and Han.4 In ized study participants are described in Table 1.
ORIGINAL RESEARCH

a case when no diuretics were administered, Dmin/2 was used Mean age was 74.7±9.9 years and 38% were
as denominator. female. A total of 53.3% of patients had de novo
ARTICLE

The daily urine volume V1 and the diuretics-corrected urine


HF and 68.3% of all patients had ischemic cardio-
volume V1D as well as weight, NT-proBNP (after logarithmic
myopathy. Median NT-proBNP concentrations were
transformation), and eGFR were analyzed by means of a mixed
model with the treatment, time, and treatment–time interaction 3386 (interquartile range, 2122–5344) pg/mL and
as fixed effects and the patient as random effect. mean systolic blood pressure was 135±23 mm Hg.
All tests of secondary measures are regarded as sensi- The mean eGFR at baseline was 60.2±18.7 mL/min
tivity analyses and are exploratory. For this reason, the sig- per 1.73 m2. A total of 21.1% had a left ventricular
nificance level was set to α=0.05 and no α correction for ejection fraction <30%.
multiple testing was done. A total of 63% of patients in the empagliflozin group
and 59% of patients in the placebo group had previous
treatment with loop diuretic medication (Table 1). No
RESULTS significant differences in baseline characteristics were
noted between the 2 groups.
Baseline Demographics
A total of 87 patients were screened between June
2019 and May 2021 at the University Hospital Jena, Primary Outcome
Germany, of whom 30 were randomized to empa- Cumulative urine output over 5 days was higher in the
gliflozin and 30 to placebo (Figure 1). One patient empagliflozin group compared with the placebo group
had to be excluded after randomization before the first (median, 10 775 versus 8650 mL in placebo; group dif-
dose of trial medication was administered because of ference, 2125 mL [95% CI 840–3550]; Figure 2). Thus,
acute ECG changes and need for urgent cardiac cath- patients in the empagliflozin group showed a 25% in-
eterization. Thus, only 29 patients were included in the crease in urine output over 5 days compared with pa-
placebo group for analysis. tients in the placebo group (P=0.003).
Downloaded from http://ahajournals.org by on July 8, 2023

Figure 1. Consolidated Standards of Reporting Trials diagram.

292 July 26, 2022 Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038


Schulze et al Empagliflozin in ADHF

Table 1.  Baseline Characteristics Table 1. Continued

ORIGINAL RESEARCH
Characteristics Empagliflozin (n=30) Placebo (n=29) Characteristics Empagliflozin (n=30) Placebo (n=29)
Age, y 72.9±11.2 76.5±8.3  Creatine kinase, µmol/ 1.9±2.2 (1.1–2.8) 1.6±1.0 (1.2–2.0)

ARTICLE
(68.8–77.1) (73.4–79.7) L×s

Female sex 11/30 (36.7) 12/29 (41.4)   Lactate, mmol/L 1.7±0.7 (1.4–2) 1.7±0.7 (1.4–2)

Type 2 diabetes 13/30 (43.3) 10/29 (34.5)   Leukocytes, Gpt/L 8.6±2 (7.8–9.3) 8.8±3.9 (7.3–10.3)

NYHA functional class   Hemoglobin, mmol/L 7.7±1.4 (7.2–8.2) 7.8±1.5 (7.2–8.3)

 II 3 (10) 5 (17.2)   Hematocrit, % 37.7±5.9 (35.5–39.9) 38.1±6.8 (35.4–40.7)

 III 20 (66.7) 13 (44.8)   Platelets, Gpt/L 218±61 (195–240) 226±66 (200–252)

 IV 7 (23.3) 11 (37.9)   NT-proBNP, pg/mL 4726±4516 4823±4995


(2939–6513) (2923–6724)
Body mass index, kg/m2 31.1±9.6 (27.5–34.7) 29.9±6.5 (27.1–32.6)
Heart rate, bpm 80±17 (73–87) 79±23 (70–88) Values are mean±SD (95% CI) or n/total N (%). eGFR indicates estimated
glomerular filtration rate; MELD-XI, Model for End-Stage Liver Disease Exclud-
Systolic blood pressure, 139±25 (129–148) 132±21 (124–140) ing International Normalized Ratio; NT-proBNP, N-terminal probrain natriuretic
mm Hg peptide; and NYHA, New York Heart Association.
Atrial fibrillation 13/27 (48.1) 14/27 (51.9)
Hypertension 27/30 (90) 25/29 (86.2) Secondary Outcomes
Left ventricular ejection 45±16 (39–51) 44±14 (30–50) Patients in the empagliflozin group showed no signifi-
fraction, % cant difference in body weight reduction compared
  ≤30% 6/29 (20.7) 6/28 (21.4) with patients in the placebo group over 5 days (mean
Pathogenesis of heart failure −4.2 versus −3.0 kg; difference, −1.2 kg [95% CI
 Ischemic 8/30 (27) 10/29 (34) −2.99 to 0.63]; P=0.198), which persisted until hos-
 Nonischemic 22/30 (73) 19/29 (66)
pital discharge with a trend toward lower levels at
follow-up on day 30 (difference in slope, −0.07 kg/d
De novo heart failure 18/30 (60) 14/29 (48)
[95% CI, −0.131 to 0.001]; P=0.054; Figure 3A).
Heart failure medication
This was accompanied by a greater reduction in mean
 Renin–angiotensin 23/30 (76.7) 20/29 (69) NT-proBNP from baseline in the empagliflozin group
inhibitor
compared with placebo (−1861 versus −727.2 pg/mL
Downloaded from http://ahajournals.org by on July 8, 2023

 Sacubitril/valsartan 5/30 (16.7) 5/29 (17.2) after 5 days; quotient in slope, 0.89 [95% CI 0.83–
 Mineralocorticoid re- 7/30 (23.3) 4/29 (13.8) 0.95]; P<0.001; Figure 2B). The treatment effect on
ceptor antagonist
NT-proBNP remains similar when the mixed model
  β-blocker 22/30 (73.3) 25/29 (86.2) analysis is adjusted for de novo HF and previous use
 Previous treatment with 19/30 (63) 17/29 (59) of loop diuretics (P=0.0006).
loop diuretics Both total and net urine output over 5 days were
Laboratory values higher in the empagliflozin group compared with the pla-
  Sodium, mmol/L 139±4 (138–141) 138±5 (136–140) cebo group (Table 2). Daily furosemide equivalent dose
  Potassium, mmol/L 4.1±0.5 (3.9–4.3) 3.9±0.6 (3.7–4.2) and the cumulative dose of loop diuretics were lower and
  Creatinine, µmol/L 107±29 (96–118) 98±28 (87–109)
diuretic efficiency expressed as milliliters urine produc-
tion per milligram furosemide equivalent was higher in
 eGFR, mL/min per 58.2±19.3 (51–66) 62.2±18.2 (55–69)
1.73 m2 the empagliflozin group compared with placebo (14.1
  Hemoglobin A1c, % 6.6±1.5 (6.0–7.2) 6.4±0.9 (6.0–6.7)
mL urine per milligram furosemide equivalent [95% CI,
0.6–27.7]; P<0.041; Table 2, Figure 2C, and Tables S2
 Fasting glucose, 8±1.5 (6.7–9.3) 7.8±2.3 (6.0–9.5)
mmol/L
and S3; Hodges-Lehmann estimate of group difference
43.7 mL urine per milligram furosemide equivalent [95%
  Uric acid, µmol/L 458±111 (411–505) 488±151 (421–555)
CI, 0.1–93]). Urine output and diuretic efficiency are
  Urea, mmol/L 7.9±3.7 (6.5–9.3) 7.7±3.7 (6.3–9.2)
independent of de novo HF or previous loop diuretic use
  Bilirubin, µmol/L 13.3±9.4 (9.8–16.8) 16.8±9.6 (13–20.6) (Tables S2 and S3).
 Alanine aminotransfer- 0.83±1.8 0.45±0.43 No differences in changes in eGFR during the treat-
ase, µmol/L×s (0.15–1.51) (0.28–0.61)
ment period were observed between the 2 groups
 Aspartate aminotrans- 0.92±1.6 (0.3–1.54) 0.50±0.2 (0.41–0.58) (eGFR, 51±19 versus 54±17 mL/min per 1.73 m²;
ferase, µmol/L×s
P=598). Previous use of loop diuretics or de novo HF
  MELD-XI score 12.3±2.5 (11.4–13.3) 11.9±2.5 (11.0–12.9) had no effect on eGFR levels (Table S3). At the 30-day
 Lactate dehydroge- 4.6±0.9 (4.3–5) 5.7±4.2 (3.8–7.3) follow-up visit, patients in the placebo group had lower
nase, µmol/L×s
eGFR compared with patients in the empagliflozin
(Continued ) group (Figure 3B). Additional serum markers of kidney

Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038 July 26, 2022 293


Schulze et al Empagliflozin in ADHF

the empagliflozin group had lower levels of serum uric


A Cumulative urine volume
acid during the 5-day study period.
ORIGINAL RESEARCH

12500
Mixed model analysis of daily urine volume sum
Difference of least square means (treatment effect):
387 mL (95% CI 50 to 724) Empagliflozin
ARTICLE

P=0.025
Urine volume sum cumulative [mL]

10000
Placebo
Quality of Life Questionnaires
Patients in the empagliflozin group had a greater abso-
7500
lute change in New York Heart Association class from
baseline to day 5 and until hospital discharge (Figure
5000
S1). The absolute improvement in EQ-5D index and vi-
sual analogue scale health status was numerically higher
2500
in the empagliflozin group compared with placebo but
day 1 day 2 day 3 day 4 day 5
not statistically significant (Figure 4).17
Number of patients
Empagliflozin 30 30 30 30 28
Placebo 29 29 29 29 29

Safety and Adverse Events


B Dynamics in NT-proBNP by day
No statistically significant differences in the occurrence
120 Mixed model analysis after logarithmic transformation
Quotient of slopes (treatment-time interaction)
0.888 (95% CI 0.830 to 0.950)
of safety events were observed between the 2 groups
P<0.001
during the study period or during follow-up of 30 days
NT-proBNP relative to baseline [%]

100
after randomization (Table 4). No early discontinuation of
trial drug was registered. No patient was lost to follow-
80 up. Two patients died in the placebo group and 1 patient
Placebo
died in the empagliflozin arm.
60

Empagliflozin
DISCUSSION
40
Baseline day 2 day 3 day 4 day 5 In patients with ADHF, early initiation of empagliflozin
Number of patients
Empagliflozin
Placebo
27
29
26
26
24
23
25
25
24
24
added to standard decongestive treatments led to a
25% increase in cumulative urine output over 5 days
of treatment. Empagliflozin also led to an increase in
Downloaded from http://ahajournals.org by on July 8, 2023

C Diuretic efficency

diuretic efficiency as well as greater reduction in NT-


Net urine output per mg furosemide [mL/mg]

20
Empagliflozin
proBNP and a trend toward lower body weight (Fig-
Placebo ures 2 and 3). These effects were not accompanied by
0 increased kidney dysfunction compared with placebo
(Figure 3B and Tables 2 and 3).
-20
The findings of the current study add to the increas-
ing evidence on effects of SGLT2 inhibition in patients
-40
Mixed model analysis of net urine output per mg furosemide equivalent
with cardiovascular diseases.12–15,18–21 The effects of
Difference of least square means (treatment effect):
14.1 mL/mg furosemide IV (95% CI 0.6 to 27.7)
empagliflozin in patients with ADHF are consistent with
-60 P=0.041
findings of previous studies in patients with kidney dys-
day 1
Number of patients
day 2 day 3 day 4 day 5
function and HF with preserved and reduced ejection
Empagliflozin
Placebo
30
29
30
29
30
29
30
29
28
29 fraction regardless of diabetes status that consistently
showed the clinical benefits of SGLT2 inhibition.12–15,19
Figure 2. Primary and secondary outcomes. First introduced as antihyperglycemic drugs, SGLT2
A, Cumulative urine output over 5 days (primary outcome). B, inhibitors have shown benefits in cardiovascular risk pre-
Dynamics in NT-proBNP levels in relation to baseline (mean and
SEM values evaluated under logarithmic transformation). C, Diuretic
vention and in patients with chronic stable HF.12–15,18,19
efficiency over 5 days. Error bars represent SEM. SGLT2 inhibitors reduce renal glucose reabsorption
and thus increase urinary glucose excretion. In addi-
function and injury including creatinine, urea, and cys- tion to glucosuric effects, empagliflozin is also associ-
tatin-C as well as urine markers of kidney injury includ- ated with osmotic diuresis and natriuresis.22 Chronic
ing urinary total protein, albumin, and α1-microglobulin use decreases body weight and blood pressure with-
(corrected for urine creatinine) were not different out increases in heart rate and has favorable effects on
between the 2 groups (total urinary protein, 492±845 markers of arterial stiffness and vascular resistance.21 In
versus 503±847 mg/g creatinine; P=0.975; and uri- patients with HF with reduced and preserved ejection
nary α1-microglobulin, 55.4±38.6 versus 31.3±33.6 fraction, SGLT2 inhibition prevented the decline in eGFR
mg/g creatinine; both P=0.066; Table 3). Patients in over time.14,15,18,21

294 July 26, 2022 Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038


Schulze et al Empagliflozin in ADHF

A Change from baseline in body weight by day B Change from baseline in eGFR by day

ORIGINAL RESEARCH
0 Mixed model analysis
Mixed model analysis
Change from baseline in actual body weight [kg]

Difference in slope (treatment-time interaction) Difference in slope (treatment-time interaction):


-0.066 kg per day (95% CI -0.131 to 0.001) 0.29 mL/min per day (95% CI 0.07 to 0.51)

Change from baseline in eGFR [mL/min]

ARTICLE
Empagliflozin
P=0.054 P=0.010
0
-2

-4
Placebo
-10

-6 Empagliflozin
Placebo

-20

Baseline day 2 day 3 day 4 day 5 Discharge day 30 Baseline day 2 day 3 day 4 day 5 Discharge day 30
Number of patients Number of patients
Empagliflozin 30 28 30 29 28 26 19 Empagliflozin 29 29 28 28 27 25 17
Placebo 26 24 26 23 25 25 15 Placebo 29 27 26 28 28 26 18

Figure 3. Secondary outcomes with extended 30-day follow-up.


A, Change in body weight from baseline in both groups. B, Dynamics in estimated glomerular filtration rate (eGFR) as change from baseline in
both groups. Error bars represent SEM.

The current study is distinct from previous clinical tri- urine output compared with 10 mg. We aimed to explore
als of SGLT2 inhibition in HF in several aspects. First, potentially negative effects of empagliflozin in ADHF in com-
no previous trial has recruited patients at such an early bination with high-dose loop diuretics and other HF drugs.
time period (<12 hours) after hospital admission with- Previous studies on the role of ultrafiltration in decom-
out hemodynamic stabilization. Second, this trial was pensated HF, such as UNLOAD (Ultrafiltration Versus
designed to monitor early effects of SGLT2 inhibition Intravenous Diuretics for Patients Hospitalized for Acute
including risks and benefits during the acute phase of Decompensated Congestive HF) and AVOID-HF (Aqua-
decongestive treatment in decompensated HF. Third, pheresis Versus Intravenous Diuretics and Hospitaliza-
patients were monitored during the most vulnerable tion for HF), have demonstrated that these therapies
Downloaded from http://ahajournals.org by on July 8, 2023

phase of acute HF treatment from admission to clinical may cause incremental weight loss comparable with the
stabilization (full study observation time was 5 days). effects of SGLT2 inhibition as shown in EMPAG-HF.10,11
The dosage of 25 mg empagliflozin in EMPAG-HF was Results from AQUAMARINE (Answering the Question
chosen to maximize potential diuretic effects in ADHF. We of Tolvaptan’s Efficacy for Patients With ADHF and Kid-
hypothesized that the higher dose would result in increased ney Failure) demonstrated that tolvaptan was associated

Table 2.  Primary and Secondary End Points

Estimation of group
Outcomes Empagliflozin Placebo difference* (95% CI)
Total urine output over 5 days, mL 10 775 (9100 to 12 925) 8650 (6450 to 10 350) 2125 (840 to 3550)†
Secondary outcomes
  Net urine output over 5 days, mL 3725 (2622 to 5830) 1480 (650 to 3826) 1950 (674 to 3250)
  Net fluid output over 5 days, mL 3925 (2825 to 6505) 1680 (850 to 4026) 2005 (700 to 3300)
  Change in body weight, kg (day 5) – 4.19±3.5 –3.02±2.9 –1.18 (–2.99 to 0.63)
  Cumulative dose of diuretics (in milligrams furosemide equivalent) 313±194.6 351.4±220.7 –38.4 (–176.7 to 70.0)
  Diuretic efficiency (mL/mg furosemide equivalent) 8.3 (–32.9 to 58.8) –25.9 (–80.3 to 16.8) 43.7 (0.1 to 93)
Renal function measures under treatment (day 5)
  Increase in serum creatinine of ≥26.5 µmol/L (0.3 mg/dL) 3/26 (11.5) 9/28 (32.1)
  Doubling of serum creatinine 0 of 26 0 of 28
  Need for renal replacement therapy 0 of 29 0 of 29
  Total urinary sodium excretion, mmol/L (day 5) 93±36.8 99.4±30.6 –6.4 (–27.6 to 14.8)
  Fractional excretion of sodium (day 5) 3170±2802.2 2439.1±2134.6 731 (–939 to 2400)

Values are mean±SD (95% CI), median (interquartile range), or n/total N (%).
*Hodges-Lehman estimation for median.
†Wilcoxon 2-sample test; P=0.003.

Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038 July 26, 2022 295


Schulze et al Empagliflozin in ADHF

Table 3.  Dynamics in Renal Function


ORIGINAL RESEARCH

Empagliflozin Placebo

Renal function measure Day 1 Day 3 Day 5 Day 30 Day 1 Day 3 Day 5 Day 30
ARTICLE

Daily urine output, mL 1688±972 2241±629 2555±1192 — 1580±1524 1963±871 1929±889 —


Daily net urine output, mL 737±971 784±636 1060±1027 — 616±1521 601±924 466±746 —
Diuretics-controlled urine output, −28.4±60.9 16.8±45.1 20.9±69 — −48.4±87.0 0.5±26.6 7.5±34.5 —
mL/mg furosemide
Serum markers of renal function
  Creatinine, µmol/L 107±28.8 115±34.8 119±33.9 120±42.1 97.8±28.2 107±24.7 111±29.4 118.4±29.4
  eGFR, mL/min per 1.73 m² 58.2±19.32 53.2±19.8 51.1±19.1 52.9±23.7 62.2±18.2 55.0±16.7 53.7±17.4 47.5±11.7
  Urea, mmol/L 7.9 C3.7 9.0±3.5 9.7±3.5 8.0±3.3 7.7±3.7 8.8±3.1 8.9±3.0 9.7±4.5
  Cystatin-C, mg/L 2±1.5 2.1±1.5 2.0±0.8 2.3±1.3 1.5±0.3 1.7±0.4 1.6±0.4 1.7±0.3
  Uric acid, µmol/L 458±111 436±117 423±121 482±146 488±151 512±138 509±152 501±147
Urine markers
  Urine total protein, mg/g creatinine 343±333 583±742 492±845 664±930 273±168 418±469 503±847 309±214
  Urine albumin, mg/g creatinine 112±153 250±543 233±682 309±812 80.6±93.1 127±177 164±408 86.7±82.2

 Urine α1- microglobulin, mg/g 95.1±91 105±159 55.4±38.6 76.7±75.8 141±258 141±496 31.3±33.6 20.6±15.7
creatinine
  Urine creatinine, mmol/L 5.8±5.3 4.48±3.11 4.6±3.5 6.3±3.0 6.78±4.32 6.45±4.66 6.5±7.3 7.92±6.77
  Urine uric acid, µmol/L 1460±1045 1852±1767 1635±1243 1820±1695 1460±1444 1635±1782 1595±1164 1537±1346
  Urine glucose, mmol/L 7.4±15.6 84.7±76.3 70.1±82.3 13.7±44.5 0.3±0.3 5.5±18.8 7.7±29.4 15.5±30.2

All data are expressed as mean±SD. eGFR indicates estimated glomerular filtration rate.

with incremental diuresis and no evidence for untoward Who Have Been Stabilized)])20 only after clinical stabiliza-
effects on kidney function in acute HF.9 However, early tion. A previous study using 10 mg of empagliflozin ver-
initiation of tolvaptan in patients with HF and reduced sus placebo initiated within 24 hours of hospitalization for
Downloaded from http://ahajournals.org by on July 8, 2023

ejection fraction did not improve outcomes in EVEREST ADHF (EMPA-RESPONSE [Randomized, Double Blind,
(Efficacy of Vasopressin Antagonism in Heart Failure Placebo Controlled, Multicenter Pilot Study on the Effects
Outcome Study With Tolvaptan).7 of Empagliflozin on Clinical Outcomes in Patients with Acute
Because of concerns about AEs, recent acute HF trials Decompensated Heart Failure]) failed to show a difference
have focused on initiation of medication (eg, sacubitril-val- in the primary end point of dyspnea score, diuretic response,
sartan [PIONEER-HF (Comparison of Sacubitril-Valsartan length of stay, or change in NT-proBNP.24 Here, we show for
Versus Enalapril on Effect on NT-proBNP in Patients Sta- the first time in a randomized study the safety and tolerabil-
bilized From an Acute Heart Failure Episode)]23 or empa- ity of an early initiation of the SGLT2 inhibitor empagliflozin
gliflozin [EMPULSE (Empagliflozin Compared to Placebo on top of diuretic regimens in patients with ADHF without
Initiated in Patients Hospitalized for Acute Heart Failure negative effects on kidney function or injury patterns.

A EQ-5D-3L sum score by day B EQ-5D-3L VAS by day


Mixed model analysis Mixed model analysis
Difference in slope (treatment-time interaction): 70 Difference in slope (treatment-time interaction):
0.85 0.003 per day (95% CI -0.002 to 0.008) 0.02 per day (95% CI -0.37 to 0.42)
P=NS P=NS
Index et al 2005

Empagliflozin Empagliflozin
Actual health state (VAS)

0.80 60
acc. Greiner

Placebo Placebo
EQ-5D IndexEQ-5D

0.75
50

0.70

40

0.65
Baseline day 5 Discharge day 30 Baseline day 5 Discharge day 30
Number of patients Number of patients
Empagliflozin 30 29 24 28 Empagliflozin 30 29 23 27
Placebo 28 29 25 25 Placebo 28 29 25 24

Figure 4. Clinical outcomes.


A, Change in EQ-5D. B, Change in health status visual analogue scale. Error bars represent SEM.

296 July 26, 2022 Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038


Schulze et al Empagliflozin in ADHF

Table 4.  Safety Events ment instruments such as the Kansas City Cardiomyopa-
thy Questionnaire. Because of the COVID-19 pandemic,

ORIGINAL RESEARCH
Event Empagliflozin Placebo
Worsening heart failure 1 (3.3) 4 (13.8)
we recruited patients at slower enrollment rates and over
a relatively long period (2019 through 2021). The use of

ARTICLE
Worsening renal function (increase in 3/26 (11.5) 9/28 (32.1)
serum creatinine ≥26.5 µmol/L [0.3
sacubitril-valsartan was low and patients with previous
mg/dL]) SGLT2 inhibition were excluded.
Worsening liver function 0 0 In patients with ADHF, early initiation of the SGLT2
Change in coagulation status at day 5 1/23 (4.3) 2/25 (8)
inhibitor empagliflozin in addition to standard treatment
is safe and increases urine output without affecting kid-
Urinary tract infection 1 (3.3) 4 (13.8)
ney function. Thus, addition of SGLT2 inhibition to stan-
Stroke or transient ischemic attack 1 (11.1) 0
dard diuretic therapy is a promising strategy to improve
30-day mortality 1 (3.3) 2 (6.9) early decongestion in patients with ADHF.
All data are expressed as n/total N (%).

Consistent with data from previous trials in ambulatory ARTICLE INFORMATION


patients with HF, an initial decrease of eGFR was noted Received January 4, 2022; accepted May 25, 2022.
that was lower in the empagliflozin group compared with Affiliations
placebo, followed by higher eGFR in the empagliflozin Department of Internal Medicine I, Division of Cardiology, Angiology and Intensive
group at 30 days.18 This suggests the possible absence Medical Care (P.C.S., J.B., J.W., P.A., F.H., S.G., S.M.-W.), Center for Clinical Stud-
ies (U.S.), and Department of Internal Medicine III, Division of Nephrology (M.B.),
of a reduction in intraglomerular pressure attributable to Friedrich-Schiller-University, University Hospital Jena, Germany. Department of
enhanced tubulo-glomerular feedback19,21,25 occurring dur- Cardiology and Pneumology, Georg-August-University, University Medical Cen-
ing the very early phase of SGLT-2 inhibition in patients with tre Göttingen, Germany (S.v.H.). German Center for Cardiovascular Research
(DZHK), partner site Göttingen, Germany (S.v.H.).
ADHF. An intermittent reduction of glomerular filtration was
typically seen during the first 2 to 4 weeks of treatment Sources of Funding
with SGLT2 inhibitors and subsequently increased within 2 This study was supported through funding by Boehringer Ingelheim to the Uni-
versity Hospital Jena.
to 3 months.19,26,27 This finding led to concerns about the
very early use of SGLT2 inhibitors in patients with acute HF, Disclosures
especially in those with concurrent renal impairment. More- Dr Schulze received honoraria and travel support from Bayer, Astra Zeneca, Dai-
over, this so-called eGFR dip caused by SGLT2 inhibition ichi Sankyo, Novartis, Actelion, Roche, Sanofi Aventis, Pharmacosmos, Medtronic,
Downloaded from http://ahajournals.org by on July 8, 2023

Thoratec, Boehringer Ingelheim, HeartWare, Coronus, Abbott, Edwards Inc,


was more pronounced in patients having additional diuretic Boston Scientific, St Jude Medical, Abiomed, and the German Cardiac Society;
therapy and more advanced kidney disease.27 Despite this research support from the National Institutes of Health, the German Research
background, our study demonstrated that the early use of Foundation, the Else Kröner Fresenius Foundation, the German Heart Founda-
tion, the European Society of Cardiology, Actelion, Medtronic, BMBF, Abiomed,
empagliflozin in ADHF is safe and well-tolerated without Boehringer Ingelheim, and Boston Scientific; and served on advisory boards for
evidence of kidney dysfunction or injury. the German Research Council, Eurotransplant, Novartis, Bayer, Pharmacosmos,
No difference in renal or cardiovascular safety end Astra Zeneca, Boehringer Ingelheim Inc, the German Cardiac Society, and the
European Society of Cardiology. Dr Möbius-Winkler received honoraria and travel
points was detected and no event of ketoacidosis support from Bayer, Daiichi Sankyo, Novartis, Boston Scientific, Abiomed, and
occurred in this study despite reduced eGFR and albu- the German Society of Cardiology; and research support from Boston Scien-
minuria at baseline. Of note, patients in the empagliflozin tific and Abiomed. Dr von Haehling has been a paid consultant for or received
honoraria payments from AstraZeneca, Bayer, Boehringer Ingelheim, BRAHMS,
group had lower levels of uric acid. This observation Chugai, Grünenthal, Helsinn, Hexal, Novartis, Pharmacosmos, Respicardia, Roche,
supports the hypothesis that the use of SGLT2 inhibi- Servier, Sorin, and Vifor; and reports research support from Amgen, AstraZeneca,
tion in patients with ADHF may affect the incidence of Boehringer Ingelheim, IMI, and the German Center for Cardiovascular Research
(DZHK). Dr Busch has been a paid consultant for or received honoraria payments
acute gout attacks, a common complication of intensive or travel support from Novartis, AstraZeneca, Boehringer Ingelheim, Vifor Pharma,
diuretic regiments. OTSUKA, Bristol Myers Squibb, and Pfizer. The other authors report no conflicts.
Our trial has some limitations. The short observation Supplemental Material
period of only 5 days may not be sufficient to observe AEs Executive Committee and Data Safety Monitoring Board
that occur after discharge and during longer use. The study Study Protocol and Statistical Analysis Plan
was not powered or designed for analysis of clinical end Figure S1
points such as cardiovascular mortality and hospitalization. Tables S1–S3
The focus on early decongestion and inclusion of patients
within 12 hours of hospitalization also excluded patients REFERENCES
with a more prolonged course of cardiac decompensation.
1. Tromp J, Bamadhaj S, Cleland JGF, Angermann CE, Dahlstrom U, Ouwerkerk
Our study used New York Heart Association classification, W, Tay WT, Dickstein K, Ertl G, Hassanein M, et al. Post-discharge prognosis
the EQ-5D index, and visual analogue scale health status of patients admitted to hospital for heart failure by world region, and national
for clinical status and quality of life assessment instead of level of income and income disparity (REPORT-HF): a cohort study. Lancet
Glob Health. 2020;8:e411–e422. doi: 10.1016/S2214-109X(20)30004-8
visual assessment scales for assessment of dyspnea and 2. McDonagh TA, Metra M, Adamo M, Gardner RS, Baumbach A, Böhm
well-being and other conventional HF symptom assess- M, Burri H, Butler J, Čelutkienė J, Chioncel O, et al; ESC Scientific

Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038 July 26, 2022 297


Schulze et al Empagliflozin in ADHF

Document Group. 2021 ESC guidelines for the diagnosis and treatment heart failure events in patients with heart failure and preserved ejection
of acute and chronic heart failure. Eur Heart J. 2021;42:3599–3726. doi: fraction: EMPEROR-Preserved Trial. Circulation. 2021;144:1284–1294.
ORIGINAL RESEARCH

10.1093/eurheartj/ehab368 doi: 10.1161/CIRCULATIONAHA.121.056824


3. Mebazaa A, Yilmaz MB, Levy P, Ponikowski P, Peacock WF, Laribi S, Ristic 16. Clark WF, Sontrop JM, Macnab JJ, Suri RS, Moist L, Salvadori M, Garg
AD, Lambrinou E, Masip J, Riley JP, et al. Recommendations on pre-hospital AX. Urine volume and change in estimated GFR in a community-based
ARTICLE

& early hospital management of acute heart failure: a consensus paper cohort study. Clin J Am Soc Nephrol. 2011;6:2634–2641. doi: 10.2215/
from the Heart Failure Association of the European Society of Cardi- CJN.01990211
ology, the European Society of Emergency Medicine and the Society of 17. Greiner W, Weijnen T, Nieuwenhuizen M, Oppe S, Badia X, Busschbach J,
Academic Emergency Medicine. Eur J Heart Fail. 2015;17:544–558. doi: Buxton M, Dolan P, Kind P, Krabbe P, et al. A single European currency for
10.1002/ejhf.289 EQ-5D health states: results from a six-country study. Eur J Health Econ.
4. Oh SW, Han SY. Loop diuretics in clinical practice. Electrolyte Blood Press. 2003;4:222–231. doi: 10.1007/s10198-003-0182-5
2015;13:17–21. doi: 10.5049/EBP.2015.13.1.17 18. Jhund PS, Solomon SD, Docherty KF, Heerspink HJL, Anand IS, Böhm
5. Metra M, Teerlink JR, Cotter G, Davison BA, Felker GM, Filippatos G, M, Chopra V, de Boer RA, Desai AS, Ge J, et al. Efficacy of dapagliflozin
Greenberg BH, Pang PS, Ponikowski P, Voors AA, et al; RELAX-AHF-2 Com- on renal function and outcomes in patients with heart failure with reduced
mittees Investigators. Effects of serelaxin in patients with acute heart failure. ejection fraction: results of DAPA-HF. Circulation. 2021;143:298–309. doi:
N Engl J Med. 2019;381:716–726. doi: 10.1056/NEJMoa1801291 10.1161/CIRCULATIONAHA.120.050391
6. O’Connor CM, Starling RC, Hernandez AF, Armstrong PW, Dickstein K, 19. Petrie MC, Verma S, Docherty KF, Inzucchi SE, Anand I, Belohlávek J, Böhm
Hasselblad V, Heizer GM, Komajda M, Massie BM, McMurray JJ, et al. Ef- M, Chiang CE, Chopra VK, de Boer RA, et al. Effect of dapagliflozin on
fect of nesiritide in patients with acute decompensated heart failure. N Engl worsening heart failure and cardiovascular death in patients with heart
J Med. 2011;365:32–43. doi: 10.1056/NEJMoa1100171 failure with and without diabetes. JAMA. 2020;323:1353–1368. doi:
7. Konstam MA, Gheorghiade M, Burnett JC Jr, Grinfeld L, Maggioni AP, 10.1001/jama.2020.1906
Swedberg K, Udelson JE, Zannad F, Cook T, Ouyang J, et al; Efficacy of 20. Tromp J, Ponikowski P, Salsali A, Angermann CE, Biegus J, Blatchford J,
Vasopressin Antagonism in Heart Failure Outcome Study With Tolvaptan Collins SP, Ferreira JP, Grauer C, Kosiborod M, et al. Sodium-glucose co-
(EVEREST) Investigators. Effects of oral tolvaptan in patients hospital- transporter 2 inhibition in patients hospitalized for acute decompensated
ized for worsening heart failure: the EVEREST Outcome Trial. JAMA. heart failure: rationale for and design of the EMPULSE trial. Eur J Heart Fail.
2007;297:1319–1331. doi: 10.1001/jama.297.12.1319 2021;23:826–834. doi: 10.1002/ejhf.2137
8. Packer M, O’Connor C, McMurray JJV, Wittes J, Abraham WT, Anker SD, 21. Zelniker TA, Braunwald E. Mechanisms of cardiorenal effects of sodium-
Dickstein K, Filippatos G, Holcomb R, Krum H, et al; TRUE-AHF Investiga- glucose cotransporter 2 inhibitors: JACC state-of-the-art review. J Am Coll
tors. Effect of ularitide on cardiovascular mortality in acute heart failure. N Cardiol. 2020;75:422–434. doi: 10.1016/j.jacc.2019.11.031
Engl J Med. 2017;376:1956–1964. doi: 10.1056/NEJMoa1601895 22. Lytvyn Y, Bjornstad P, Udell JA, Lovshin JA, Cherney DZI. Sodium glucose
9. Matsue Y, Suzuki M, Nagahori W, Yoshida K, Onishi Y, Satoh Y, Ono Y, cotransporter-2 inhibition in heart failure: potential mechanisms, clinical ap-
Nishioka T, Noda M, Sugi K, et al. Clinical effectiveness of tolvaptan in pa- plications, and summary of clinical trials. Circulation. 2017;136:1643–1658.
tients with acute decompensated heart failure and renal failure: design and doi: 10.1161/CIRCULATIONAHA.117.030012
rationale of the AQUAMARINE study. Cardiovasc Drugs Ther. 2014;28:73– 23. Velazquez EJ, Morrow DA, DeVore AD, Duffy CI, Ambrosy AP, McCague
77. doi: 10.1007/s10557-013-6491-8 K, Rocha R, Braunwald E; PIONEER-HF Investigators. Angiotensin-
10. Costanzo MR, Negoianu D, Jaski BE, Bart BA, Heywood JT, Anand IS, neprilysin inhibition in acute decompensated heart failure. N Engl J Med.
Smelser JM, Kaneshige AM, Chomsky DB, Adler ED, et al. Aquapheresis 2019;380:539–548. doi: 10.1056/NEJMoa1812851
versus intravenous diuretics and hospitalizations for heart failure. JACC 24. Damman K, Beusekamp JC, Boorsma EM, Swart HP, Smilde TDJ, Elvan
Downloaded from http://ahajournals.org by on July 8, 2023

Heart Fail. 2016;4:95–105. doi: 10.1016/j.jchf.2015.08.005 A, van Eck JWM, Heerspink HJL, Voors AA. Randomized, double-blind,
11. Costanzo MR, Guglin ME, Saltzberg MT, Jessup ML, Bart BA, Teerlink JR, placebo-controlled, multicentre pilot study on the effects of empagliflozin
Jaski BE, Fang JC, Feller ED, Haas GJ, et al; UNLOAD Trial Investigators. on clinical outcomes in patients with acute decompensated heart fail-
Ultrafiltration versus intravenous diuretics for patients hospitalized for acute ure (EMPA-RESPONSE-AHF). Eur J Heart Fail. 2020;22:713–722. doi:
decompensated heart failure. J Am Coll Cardiol. 2007;49:675–683. doi: 10.1002/ejhf.1713
10.1016/j.jacc.2006.07.073 25. van Bommel EJM, Lytvyn Y, Perkins BA, Soleymanlou N, Fagan NM,
12. Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Koitka-Weber A, Joles JA, Cherney DZI, van Raalte DH. Renal he-
Verma S, Tsutsui H, Brueckmann M, et al; EMPEROR-Reduced Trial Investiga- modynamic effects of sodium-glucose cotransporter 2 inhibitors
tors. Cardiovascular and renal outcomes with empagliflozin in heart failure. N in hyperfiltering people with type 1 diabetes and people with type 2
Engl J Med. 2020;383:1413–1424. doi: 10.1056/NEJMoa2022190 diabetes and normal kidney function. Kidney Int. 2020;97:631–635. doi:
13. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez 10.1016/j.kint.2019.12.021
FA, Ponikowski P, Sabatine MS, Anand IS, Bělohlávek J, et al; DAPA-HF Trial 26. Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus
Committees and Investigators. Dapagliflozin in patients with heart failure M, Johansen OE, Woerle HJ, Broedl UC, Zinman B; EMPA-REG OUT-
and reduced ejection fraction. N Engl J Med. 2019;381:1995–2008. doi: COME Investigators. Empagliflozin and progression of kidney dis-
10.1056/NEJMoa1911303 ease in type 2 diabetes. N Engl J Med. 2016;375:323–334. doi:
14. Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, 10.1056/NEJMoa1515920
Brunner-La Rocca HP, Choi DJ, Chopra V, Chuquiure-Valenzuela E, et al; 27. Kraus BJ, Weir MR, Bakris GL, Mattheus M, Cherney DZI, Sattar N,
EMPEROR-Preserved Trial Investigators. Empagliflozin in heart failure with Heerspink HJL, Ritter I, von Eynatten M, Zinman B, et al. Characteriza-
a preserved ejection fraction. N Engl J Med. 2021;385:1451–1461. doi: tion and implications of the initial estimated glomerular filtration rate
10.1056/NEJMoa2107038 ‘dip’ upon sodium-glucose cotransporter-2 inhibition with empagliflozin
15. Packer M, Butler J, Zannad F, Filippatos G, Ferreira JP, Pocock SJ, Carson in the EMPA-REG OUTCOME trial. Kidney Int. 2021;99:750–762. doi:
P, Anand I, Doehner W, Haass M, et al. Effect of empagliflozin on worsening 10.1016/j.kint.2020.10.031

298 July 26, 2022 Circulation. 2022;146:289–298. DOI: 10.1161/CIRCULATIONAHA.122.059038

You might also like