You are on page 1of 17

AJN Patient-Oriented, Translational Research: Research Article

American Journal
of Nephrology Am J Nephrol Received: July 2, 2023
Accepted: September 26, 2023
DOI: 10.1159/000534366 Published online: October 4, 2023

Effects of Mineralocorticoid Receptor

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
Antagonists for Chronic Kidney Disease:
A Systemic Review and Meta-Analysis
Chen-Yi Yuan a, b Yuan-Cheng Gao c Yi Lin d Lin Liu a Xiao-Gang Shen a
Wen-Li Zou a Min-Min Wang a Quan-Quan Shen a Li-Na Shao a
Yue-Ming Liu a Jia-Wei Zhang a Zhi-Hui Pan d Yan Zhu d Jing-Ting Yu d
Xu-Guang Yu e Bin Zhu a
a
Urology and Nephrology Center, Department of Nephrology, Zhejiang Provincial People’s Hospital, Affiliated
People’s Hospital, Hangzhou Medical College, Hangzhou, China; bDepartment of Nephrology, The First
Affiliated Hospital of Ningbo University, Ningbo, China; cDepartment of Nephrology, Zhejiang Hospital of Chinese
Medicine, Affiliated to Zhejiang University of Chinese Medicine, Hangzhou, China; dDepartment of Nephrology,
Hangzhou Hospital of Chinese Medicine (Guangxing Hospital), Affiliated to Zhejiang University of Chinese
Medicine, Hangzhou, China; eDepartment of Nephrology, Yueqing People’s Hospital, Wenzhou, China

Keywords CI, −140.17 to −41.64 mg/g), 24-h urinary protein excretion


Mineralocorticoid receptor antagonists · Diabetic kidney (WMD, −0.20 g, 95% CI, −0.28 to −0.12 g), estimated glomerular
disease · Chronic kidney disease · Meta-analysis filtration rate (eGFR) (WMD, −1.99 mL/min/1.73 m2, 95% CI, −3.28
to −0.70 mL/min/1.73 m2), chronic renal failure events (RR, 0.86,
95% CI, 0.79–0.93), and cardiovascular events (RR, 0.84, 95% CI,
Abstract 0.77–0.92). MRAs increased the incidence of hyperkalemia (RR,
Background: Mineralocorticoid receptor blockade could be 2.04, 95% CI, 1.73–2.40) and hypotension (RR, 1.80, 95% CI,
a potential approach for the inhibition of chronic kidney 1.41–2.31). MRAs reduced the incidence of peripheral edema (RR,
disease (CKD) progression. The benefits and harms of dif- 0.65, 95% CI, 0.56–0.75) but not the risk of acute kidney injury (RR,
ferent mineralocorticoid receptor antagonists (MRAs) in CKD 0.94, 95% CI, 0.79–1.13). Nonsteroidal MRAs (RR, 0.66, 95% CI,
are inconsistent. Objectives: The aim of the study was to 0.57–0.75) but not steroidal MRAs (RR, 0.20, 95% CI, 0.02–1.68)
summarize the benefits and harms of MRAs for CKD patients. significantly reduced the risk of peripheral edema. Steroidal
Methods: We searched MEDLINE, EMBASE, and the Co- MRAs (RR, 5.68, 95% CI, 1.26–25.67) but not nonsteroidal MRAs
chrane databases for trials assessing the effects of MRAs on (RR, 0.52, 95% CI, 0.22–1.22) increased the risk of breast disorders.
non-dialysis-dependent CKD populations. Treatment and Conclusions: In the CKD patients, MRAs, particularly in combi-
adverse effects were summarized using meta-analysis. nation with angiotensin-converting enzyme inhibitor/angiotensin
Results: Fifty-three trials with 6 different MRAs involving receptor blocker, reduced albuminuria/proteinuria, eGFR, and the
22,792 participants were included. Compared with the
control group, MRAs reduced urinary albumin-to-creatinine
ratio (weighted mean difference [WMD], −90.90 mg/g, 95% Chen-Yi Yuan and Yuan-Cheng Gao contributed equally to this work.

karger@karger.com © 2023 S. Karger AG, Basel Correspondence to:


www.karger.com/ajn Bin Zhu, zhubing @ hotmail.com
incidence of chronic renal failure, cardiovascular and peripheral 1966 through November 2022), and the Cochrane Library (Co-
edema events, whereas increasing the incidence of hyperkalemia chrane Central Register of Controlled Trials; no date restriction).
and hypotension, without the augment of acute kidney injury Searches were conducted using relevant text words and medical
subject headings, including all spellings of MRA, aldosterone
events. Nonsteroidal MRAs were superior in the reduction of antagonist, spironolactone, eplerenone, finerenone, esaxerenone,
more albuminuria with fewer peripheral edema events and apararenone, ocedurenone, and CKD for randomized controlled
without the augment of breast disorder events. trials (RCTs) without language restriction. The ClinicalTrials.gov
© 2023 S. Karger AG, Basel website and reference lists from identified trials and review articles
were scanned to identify other relevant trials (shown in online
suppl. Table 1; for all online suppl. material, see https://doi.org/10.
1159/000534366).

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
Introduction
Study Selection
Chronic kidney disease (CKD) is a global health concern The literature search, data extraction, and quality assessment
whose medical burden is substantial and increasing. It was were independently performed by two investigators (C.-Y.Y. and
estimated that CKD had a prevalence of 14% in America from Y.-C.G.). All RCTs assessing the effects of MRAs in CKD (defined
2017 to 2020 [1]. CKD was indicated to be a predominant as the urinary albumin-to-creatinine ratio [UACR] ≥30 mg/g and
cause of mortality worldwide [2]. Diabetes mellitus (DM) has the estimated glomerular filtration rate [eGFR] ≤90 mL/min/
1.73 m2) not receiving renal replacement therapy were eligible for
become the leading cause of CKD [3]. It is estimated that inclusion. We excluded non-RCT trials, trials with participants
diabetic kidney disease (DKD) accounts for 30–50% of all receiving maintenance dialysis or kidney transplantation, or the
global CKD cases and may increase to 69% by 2030 [4]. trials without a control arm.
Renin-angiotensin-aldosterone system inhibitors are the
major treatment for CKD [5, 6]. However, 40% of CKD Outcome Evaluation
patients receiving angiotensin-converting enzyme inhibitors The outcomes assessed included UACR, 24-h urinary protein
(ACEI) or angiotensin receptor blockers (ARB) develop “al- (UPE), eGFR, chronic kidney failure events, systolic blood pressure
dosterone breakthrough,” which is associated with insulin (SBP), diastolic blood pressure (DBP), serum potassium, CVE, and
all-cause mortality, as well as adverse effects of hyperkalemia,
resistance and endothelial dysfunction to aggravate CKD reproductive system disorders, and breast disorders. Chronic
progression [6, 7]. Aldosterone activates the mineralocorticoid kidney failure was defined as end-stage kidney disease (defined as
receptor (MR) in the nonepithelial tissues of the heart and the initiation of long-term dialysis or kidney transplantation, or an
kidney to induce inflammation and fibrosis, leading to left eGFR below 15 mL/min/1.73 m2), or a sustained decrease from
ventricular hypertrophy and CKD progression [8, 9]. An- baseline of at least 40% in eGFR. CVEs were defined as a composite
of cardiovascular death, myocardial infarction, myocardial is-
giotensin blockade is inefficient against the nonclassical chemia, heart failure, ischemic stroke, and cerebrovascular acci-
profibrotic actions of aldosterone [10]. CKD patients remain dent. Reproductive system disorders were defined as a composite
at a high risk of cardiovascular events (CVEs) and kidney of gynecological oncology and menstrual disorders. Breast dis-
disease progression despite receiving ACEI/ARB [11, 12]. orders included breast tenderness, breast hyperplasia, and gy-
Then, MR blockade could be a potential approach for the necomastia. Hyperkalemia, hypotension, and eGFR slope were
inhibition of CKD progression [13, 14]. Steroidal MR an- defined according to the original trials (shown in online suppl.
Table 2). When these definitions were not provided, alternative,
tagonists (MRAs) have been demonstrated to benefit the heart comparable definitions provided by the trial authors were
and kidney [15]. The new-generation nonsteroidal MRAs included.
(finerenone, ocedurenone, esaxerenone, and apararenone)
exhibit better selectivity for MR over the other steroidal re- Data Extraction and Assessment of Study Quality and Risk
ceptors [16–18] and appear to have a better benefit-risk ratio of Bias
[19–22]. However, the effects of different MRAs on CKD are Published reports were obtained for each eligible trial, and
inconsistent. This meta-analysis was performed to clarify the relevant data were extracted for analyses. In the multi-dose trial,
benefits and harms of MRAs for CKD patients. we combined subgroups of different MRA doses into a group using
Review Manager 5.4 (Cochrane Collaboration, Oxford, UK) [24].
We contacted the authors when the required data were not
provided in the published articles.
Materials and Methods The Cochrane quality criteria were used to assess study quality
and the risk of bias based on random sequence generation, al-
Data Sources, Search Strategy, and Selection Criteria location concealment, blinding, reporting bias, attrition bias, and
We performed this systematic review following the PRISMA any other potential sources of bias. The trials were categorized as
guidelines for meta-analyses [23] and registered in PROSPERO low, medium, or high risk of bias. A graphical view of bias status
(CRD42023393389). Relevant trials were identified by searching was generated with Review Manager 5.4. A third investigator (B.Z.)
MEDLINE (from 1950 through November 2022), EMBASE (from adjudicated any disagreements.

2 Am J Nephrol Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
DOI: 10.1159/000534366 Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
Data Synthesis and Analyses were compared with ACEi/ARB in 3 trials [30–32] and
In calculating risk ratios, the total number of patients in each dapagliflozin in one trial [33]. The comparators of di-
group was utilized as the denominator. Individual trial weighted
mean difference (WMD), relative risk (RR), and 95% confidence
uretics (6 trials) comprised hydrochlorothiazide (3 trials)
interval (CI) were calculated before data pooling. [34–36], trichlormethiazide and furosemide (3 trials) [37,
A random-effects model was adopted as proposed by DerSi- 38, 39].
monian and Laird [25]; the percentage of variability across trials
attributable to heterogeneity beyond chance was estimated using I2 Risk of Bias
statistics. Publication bias for the efficacy outcomes was assessed Of the included trials, one trial [52] was evaluated as
using the funnel plot and Egger’s test. Sensitivity analyses were
conducted to assess the robustness of the results. Subgroup an- having a low risk of bias, twenty-four trials [19, 21, 29, 31,

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
alyses were performed to explore possible sources of heterogeneity. 34, 41, 42, 47, 49, 51, 54, 58–62, 64, 65, 68, 69, 71, 73, 75,
Univariate meta-regression analysis was performed using the 76] were identified as having a medium risk of bias, and
residual maximum likelihood estimation. Study quality was an- twenty-eight trials [26–28, 30, 32, 33, 35–40, 43–46, 48,
alyzed based on the Cochrane quality criteria. A two-sided p < 0.05 50, 52, 53, 55–57, 63, 66, 70, 72, 74] as having a high risk
was considered statistically significant. All statistical analyses were
performed with STATA, version 11 (StataCorp LP). of bias. All trials were reported to be randomized; 14 trials
(26%) had clear allocation concealment methods; 33 trials
(62%) described blinding of participants and personnel.
38 trials (72%) completely reported the outcomes. Se-
Results lective reporting was judged to be at low risk of bias in 29
trials (55%), and high risk in 16 (30%). Other biases in 23
Literature Search trials (43%) were judged to be low risk. However, de-
The literature search yielded 1,447 articles, and 53 tection bias was unclear in 32 trials (60%) (shown in
trials with 22,792 patients were finally included (shown in online suppl. Fig. 1, 2).
Fig. 1).

Trial Characteristics Outcomes


The characteristics of the included trials [19, 21,
26–76] were summarized in Table 1. Of them, 39 trials Effects on UACR/UPE
were RCTs [19, 21, 28–30, 32, 33, 35, 38, 39, 42–44, 46, 47, Data regarding the effects of MRAs compared to
50–53, 55–62, 64–75], and 14 were randomized crossover placebo on UACR were available from 10 trials with
trials [27, 31, 34, 36, 37, 40, 41, 45, 48, 49, 54, 63, 76, 77], 14,033 participants. Overall, MRAs decreased UACR by
followed up from 2 to 42 months. The baseline charac- 90.90 mg/g (10 trials, 95% CI, −140.17 to −41.64 mg/g)
teristics of the intervention and control groups were compared with the control. All MRAs showed consistent
similar within all trials. Thirty trials enrolled 16,102 evidence of UACR reduction, while finerenone reduced
participants with DKD; nine trials recruited 869 partic- UACR more significantly (finerenone: WMD, −256.86 mg/g,
ipants with non-diabetes CKD; and the other fourteen 95% CI, −425.02 to −88.17 mg/g; esaxerenone: WMD,
trials recruited 5,821 participants with unspecified CKD. −56.40 mg/g, 95% CI, −103.39 to −9.41 mg/g; eplerenone:
The mean age reported for participants ranged from 34 to WMD, −26.10 mg/g, 95% CI, −48.98 to −3.22 mg/g; spi-
74 years, and the proportion of males ranged from 19% to ronolactone: WMD, −74.08 mg/g, 95% CI, −188.18
100%. Trials sample size varied from 12 to 7,352, and the to −40.02 mg/g, p for interaction = 0.004) (shown in Fig. 2).
publication time ranged from 2005 to 2022. These trials Consistently, MRAs decreased UPE by 0.20 g (8 trials, 95%
involved 6 MRA agents including 4 nonsteroidal MRAs of CI, −0.28 to −0.12 g) compared with the control group
finerenone (5 trials) [19, 21, 55, 60, 65], esaxerenone (2 (shown in online suppl. Fig. 3).
trials) [26, 27], apararenone (1 trial) [28], ocedurenone (1
trial) [29], and 2 steroidal MRAs of eplerenone (12 trials) Effects on Kidney Function
[30, 34, 40, 45, 49–51, 58, 68, 70, 72, 76], and spi- The effects of MRAs on eGFR were reported in 36 trials
ronolactone (33 trials) [26–29, 31–33, 35–39, 41–44, with 18,936 patients. Overall, compared with the control
46–48, 52–57, 59, 61–64, 66, 67, 73]. Ten trials were group, MRAs significantly decreased eGFR by 1.99 mL/
placebo-controlled only [31, 44, 50, 55, 67, 68, 72–74, 77]. min/1.73 m2 (36 trials, 95% CI, −3.28 to −0.70 mL/min/
MRAs were compared with placebo, with background of 1.73 m2) with significant heterogeneity among various
ACEi/ARB in thirty-four trials [19, 21, 27–30, 33, 36–39, MRAs (apararenone: 1 trial, WMD, −7.97 mL/min/
41, 42, 45–49, 51, 52, 54, 56–61, 64–66, 69–71, 75]. MRAs 1.73 m2, 95% CI, −10.76 to −5.18 mL/min/1.73 m2;

Mineralocorticoid Receptor Antagonists for Am J Nephrol 3


Chronic Kidney Disease DOI: 10.1159/000534366
Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
Fig. 1. Diagram of the identification process for eligible studies. RCT, randomized controlled trial.

finerenone: 3 trials, WMD, −3.80 mL/min/1.73 m2, 95% from baseline (3 trials, RR, 0.85, 95% CI, 0.78–0.92).
CI, −5.67 to −1.92 mL/min/1.73 m2; esaxerenone: 1 trials, MRAs did not significantly reduce the risk of devel-
WMD, −6.01 mL/min/1.73 m2, 95% CI, −12.08 to 0.06 oping ESRD (3 trials, RR, 0.87, 95% CI, 0.74–1.01)
mL/min/1.73 m2; eplerenone: 10 trials, WMD, −1.08 mL/ (shown in online suppl. Fig. 5).
min/1.73 m2, 95% CI, −2.40 to 0.23 mL/min/1.73 m2;
spironolactone: 21 trials, WMD, −0.52 mL/min/1.73 m2, Effects on Blood Pressure
95% CI, −3.52 to 2.48 mL/min/1.73 m2, p for interac- MRAs significantly reduced SBP by 3.88 mm Hg (33
tion <0.001) (shown in Fig. 3). trials, 95% CI, −5.11 to −2.65 mm Hg) and DBP by
1.27 mm Hg (31 trials, 95% CI, −1.99 to −0.56 mm Hg),
Effects on Outcome of Chronic Kidney Failure respectively, without significant heterogeneity among
Four trials involving 14,397 patients recorded 1,914 the different MRAs (p for interaction for SBP = 0.06; p
chronic kidney failure events. Overall, MRAs signifi- for interaction for DBP = 0.1) (shown in online suppl.
cantly reduced the risk of developing chronic kidney Fig. 6).
failure (4 trials, RR, 0.86, 95% CI, 0.79–0.93) that was
mainly driven by finerenone (2 trials, RR, 0.86, 95% CI, Effects on Serum Potassium
0.79–0.93), without significant heterogeneity among MRAs increased serum potassium by 0.18 mmol/L (32
different MRAs (p for interaction = 0.93) (shown in trials, 95% CI, 0.14–0.22 mmol/L) without significant
online suppl. Fig. 4). Separately, MRAs significantly heterogeneity among various MRAs (p for interaction =
reduced the risk of outcome of eGFR decline by ≥40% 0.3) (shown in online suppl. Fig. 7).

4 Am J Nephrol Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
DOI: 10.1159/000534366 Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
Table 1. Characteristics of included trials

Study Disease Patients, Age, Malea Intervention Follow-up UACR, mg/g eGFR, mL/min/1.73 m2 SBP, mm Hg DBP, mm Hg
n years duration,
months
experimental control group background therapy experimental control group experimental control experimental control experimental control
group group group group group group group group

FIGARO-DKD DKD 7,352 64 69 Finerenone, Usual care ACEI/ARB 41 586.40±764.60 586.90±752.70 67.60±21.60 68.00±21.70 135.80±14.00 135.70±14.10 76.70±9.50 76.80±9.50
[21] (2021) 10–20 mg/d
FIDELIO-DKD DKD 5,674 66 70 Finerenone, Usual care ACEI/ARB 31 1,185.90±1,036.90 1,210.30±1,052.30 44.40±12.50 44.30±12.60 138.10±14.30 138.00±14.40 75.80±9.70 75.80±9.70
[19] (2020) 10–20 mg/d
ARTS-DN [40] DKD 48 64 88 Finerenone, Usual care ACEI/ARB 3 194.87±278.32 256.80±166.31 65.13±12.85 60.88±16.53 137.25±16.35 135.72±16.90 77.71±10.11 69.25±11.63

Chronic Kidney Disease


(2017) 10–20 mg/d
ARTS-DN [41] DKD 437 64 76 Finerenone, Usual care ACEI/ARB 3 211.94±179.57 188.40±169.80 66.83±22.31 72.20±20.40 137.77±14.32 139.90±14.30 76.63±9.98 78.20±9.70
(2015) 10–20 mg/d
Pitt et al. [42] CKD 132 73 83 Finerenone, Usual care General therapy 4 23.70±5.20 22.20±5.60 47.00±10.90 46.90±8.20 / / / /
(2013) 10 mg/d
Ito et al. [27] DKD 449 66 77 Esaxerenone, Usual care ACEI/ARB 13 / / 69.00±18.00 69.00±18.00 140.00±10.00 140.00±10.00 82.00±8.00 84.00±8.00
(2020) 1.25–2.5 mg/d
Ito et al. [26] DKD 215 65 78 Esaxerenone, Usual care ACEI/ARB 3 126.00±63.60 123.00±58.00 66.99±18.46 69.00±19.00 138.01±11.58 138.00±11.00 / /
(2019) 1.25–2.5 mg/d
Wada et al. [28] DKD 292 62 76 Apararenone, Placebo ACEI/ARB 24 / / 74.09±20.25 77.50±20.50 / / / /
(2021) 2.5–10 mg/d

Mineralocorticoid Receptor Antagonists for


Bakris et al. [29] CKD 162 65 55 Ocedurenone, Placebo General therapy 3 / / / / 155.02±14.85 155.80±10.84 88.69±12.58 85.90±11.46
(2021) 0.25–0.5 mg/d
Provenzano CKD 46 70 76 Eplerenone, Dapagliflozin, 10 mg/d ACEI/ARB 6 / / 57.20±18.00 56.80±18.00 135.00±14.00 135.00±12.00
et al. [33] (2022) 50 mg/d
Provenzano CKD 46 70 76 Eplerenone, Usual care Dapagliflozin, 10 mg/ 6 / / 58.40±19.00 56.80±18.00 133.00±11.00 135.00±12.00 / /
et al. [33] (2022) 50 mg/d d, ACEI/ARB
El Mokadem DKD 50 49 56 Eplerenone, Usual care ACEI/ARB 6 165.40±62.94 159.96±66.22 72.84±11.93 71.80±8.46 146.36±5.39 145.00±5.00 90.91±3.98 90.65±3.79
et al. [32] (2020) 50 mg/d
El Mokadem DKD 50 50 56 Eplerenone, Ramipril 10, mg/d General therapy 6 154.33±67.15 159.96±66.22 72.28±8.85 71.80±8.46 143.50±5.80 145.00±5.00 90.83±3.81 90.65±3.79
et al. [32] (2020) 50 mg/d
Minakuchi et al. CKD 48 67 / Eplerenone, Usual care General therapy 36 / / 65.20±16.84 63.90±19.02 131.90±10.83 132.10±10.40 76.60±6.85 76.30±5.46
[43] (2020) 25 mg/d
Ferreira et al. CKD 1,152 71 77 Eplerenone, Usual care General therapy 21 / / 63.00±20.00 62.00±20.00 123.00±18.00 122.00±17.00 / /
[44] (2019) 50 mg/d
Ando et al. [45] CKD 342 59 63 Eplerenone, Usual care ACEI/ARB 13 163.10±148.00 156.80±133.60 67.70±14.30 68.60±13.60 138.60±11.10 138.80±12.60 82.40±10.20 81.90±10.00
(2014) 50 mg/d
Boesby et al. [46] CKD 54 55 19 Eplerenone, Usual care General therapy 6 / / 36.00±10.00 35.00±13.00 126.00±10.00 127.00±17.00 73.00±8.00 75.00±10.00
(2013) 25–50 mg/d
EMPHASIS-HF CKD 2737 69 78 Eplerenone, Usual care ACEI/ARB 42 / / 71.20±21.90 70.40±21.70 124.00±17.00 124.00±17.00 / /
[47] (2013) 25–50 mg/d
Tylicki et al. [48] CKD 18 39 78 Eplerenone, Usual care ACEI/ARB 10 / / 94.00±34.37 94.00±34.37 116.80±10.18 116.80±10.18 79.90±7.64 79.90±7.64
(2012) 50 mg/d
Boesby et al. [49] CKD 40 / 68 Eplerenone, Usual care ACEI/ARB 2 / / 82.00±39.09 82.00±39.09 125.00±13.00 125.00±13.00 86.00±10.00 86.00±10.00

Am J Nephrol
(2011) 25 mg/d
Morales et al. CKD 12 57 58 Eplerenone, Lisinopril, 20 mg/d General therapy 11 / / 57.30±29.10 56.20±30.00 139.00±16.50 139.00±16.50 77.67±8.50 77.67±8.50
[31] (2009) 25 mg/d
Joffe et al. [34] DKD 16 / 82 Eplerenone, Hydrochlorothiazide, ACEI/ARB 4 / / 110.00±23.00 110.00±23.00 134.00±18.00 134.00±18.00 76.00±8.00 76.00±8.00
(2007) 50 mg/d 12.5 mg/d, potassium
chloride 10mEq/d

DOI: 10.1159/000534366
Epstein et al. [50] DKD 182 / 60 Eplerenone, Usual care ACEI/ARB 3 / / / / / / / /
(2006) 50 mg/d
Tofte et al. [51] DKD 209 63 70 Spironolactone, Usual care General therapy 30 / / 81.00±15.00 82.00±19.00 135.00±12.00 134.00±12.00 79.00±9.00 79.00±9.00
(2020) 25 mg/d
Beldhuis et al. CKD 944 74 / Spironolactone, Usual care General therapy 40 / / 45.68±8.85 45.88±8.83 126.56±15.44 126.57±15.43 / /
[52] (2019) 30 mg/d
Chen et al. [53] DKD 105 68 51 Spironolactone, Usual care Irbesartan 18 / / 79.30±13.90 79.10±11.25 / / / /
(2018) 20 mg/d
Chen et al. [53] DKD 101 67 53 Spironolactone, Usual care Irbesartan 18 / / 80.00±13.50 78.70±12.70 / / / /
(2018) 20 mg/d
Srivastava et al. DKD 54 51 54 Spironolactone, Usual care ACEI/ARB 12 1,094.00±1,088.28 917.00±922.58 / / 135.00±11.00 138.00±15.00 71.00±9.00 75.00±9.00
[54] (2016) 25 mg/d
Kato et al. [55] DKD 52 60 71 Spironolactone, Usual care ACEI/ARB 2 702.10±728.40 511.30±450.10 / / 137.30±16.30 131.30±13.00 76.80±12.20 77.60±8.80
(2015) 25 mg/d
Momeni et al. DKD 40 56 / Spironolactone, Hydrochlorothiazide, General therapy 3 / / 90.70±24.60 87.38±22.00 132.80±12.80 133.50±13.20 84.30±5.40 84.00±7.40
[35] (2015) 50 mg/d 25 mg/d
Momeni et al. DKD 40 57 / Spironolactone, Usual care Hydrochlorothiazide 3 / / 86.20±25.00 87.38±22.00 133.50±14.50 133.50±13.20 84.00±9.40 84.00±7.40
[35] (2015) 50 mg/d
Moras CKD 42 56 67 Spironolactone, Hydrochlorothiazide, ACEI/ARB 6 / / 66.00±26.00 66.00±25.00 130.00±18.00 129.00±18.00 76.00±13.00 75.00±12.00
(2015) [56] 25 mg/d 50 mg/d
Moras et al. [56] CKD 42 56 67 Spironolactone, Hydrochlorothiazide, ACEI/ARB 6 / / 66.00±26.00 64.00±24.00 130.00±18.00 128.00±20.00 76.00±13.00 74.00±12.00
(2015) 25 mg/d 50 mg/d, Amiloride,
5 mg/d
Van Buren et al. DKD 53 52 49 Spironolactone, Losartan, 100 mg/d ACEI/ARB 12 1,094.00±1,076.35 897.00±934.51 51.40±34.07 64.80±34.73 135.00±11.00 143.00±15.00 71.00±9.00 75.00±9.00
[57] (2014) 25 mg/d

5
Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
6
Table 1 (continued)

Study Disease Patients, Age, Malea Intervention Follow-up UACR, mg/g eGFR, mL/min/1.73 m2 SBP, mm Hg DBP, mm Hg
n years duration,
months
experimental control group background therapy experimental control group experimental control experimental control experimental control
group group group group group group group group

Van Buren et al. DKD 54 51 46 Spironolactone, Usual care ACEI/ARB 12 1,094.00±1,056.23 917.00±905.34 51.40±33.43 73.00±38.76 135.00±11.00 138.00±15.00 71.00±9.00 75.00±9.00
[57] (2014) 25 mg/d
Esteghamati DKD 136 58 67 Spironolactone, Enalapri, 30–40 mg/d ACEI/ARB 18 / / 73.75±16.78 69.13±19.69 134.27±18.12 140.08±18.67 82.26±8.54 83.17±10.29
et al. [58] (2013) 25 mg/d
Wang et al. [59] CKD 208 34 57 Spironolactone, Usual care ACEI/ARB 4 / / 65.77±22.21 66.45±24.34 119.30±13.50 121.10±13.70 72.20±11.60 68.20±11.10
(2013) 20 mg/d
Nielsen et al. [60] DKD 69 53 77 Spironolactone, Usual care ACEI/ARB 4 / / / / / / / /
(2013) 25 mg
Ziaee et al. [61] DKD 60 53 62 Spironolactone, Usual care ACEI/ARB 3 126.30±69.36 119.00±66.86 79.84±18.05 82.55±19.18 127.24±12.50 124.19±12.85 79.31±8.42 75.96±8.60
(2013) 25 mg
Hase et al. [39] DKD 36 64 67 Spironolactone, Trichlormethiazide, ACEI/ARB 6 605.60±703.82 582.60±587.96 61.60±23.90 63.40±17.00 149.00±20.00 146.00±10.00 79.00±14.00 79.00±11.00
(2013) 25–50 mg 2 mg/d

Am J Nephrol
Pitt et al. [42] CKD 132 73 83 Spironolactone, Usual care General therapy 4 24.10±4.60 22.20±5.60 46.80±10.30 46.90±8.20 / / / /
(2013) 25–50 mg/d
Nielsen et al. [62] DKD 21 58 67 Spironolactone, Usual care ACEI/ARB 2 / / / / / / / /
(2012) 25 mg/d
Edwards et al. CKD 112 54 58 Spironolactone, Usual care ACEI/ARB 10 107.47±394.80 92.76±547.51 49.00±12.00 53.00±11.00 130.00±16.00 130.00±19.00 77.00±10.00 77.00±10.00

DOI: 10.1159/000534366
[63] (2012) 25 mg/d
Abolghasmi and CKD 41 50 54 Spironolactone, Usual care General therapy 3
Taziki [64] (2011) 25–50 mg/d
Zheng et al. [65] DKD 40 58 55 Spironolactone, Usual care ACEI/ARB 3 / / / / / / / /
(2011) 20 mg/d
Hammer et al. CKD 112 54 58 Spironolactone, Usual care ACEI/ARB 9 156.64±427.68 72.16±425.92 49.00±12.00 53.00±12.30 124.00±11.00 125.00±12.00 76.00±8.00 77.00±9.00
[66] (2010) 25 mg/d
Guney et al. [67] CKD 30 43 57 Spironolactone, Usual care ACEI/ARB 6 / / 63.02±22.71 56.28±35.59 118.80±15.70 115.80±10.60 72.50±9.70 72.90±7.50
(2009) 25 mg/d
Mehdi et al. [68] DKD 54 51 46 Spironolactone, Usual care ACEI/ARB 13 1,094.00±1,088.28 917.00±922.58 51.40±32.48 73.00±37.67 135.00±11.00 138.00±15.00 71.00±9.00 75.00±9.00
(2009) 25 mg/d
Preston et al. CKD 18 53 78 Spironolactone, Usual care ACEI/ARB 3 / / 45.15±14.44 112.10±13.04 141.44±16.20 117.22±9.17 81.55±10.17 74.16±7.13
[69] (2009) 25 mg/d
Furumatsu et al. CKD 32 52 59 Spironolactone, Trichlormethiazide, ACEI/ARB 12 / / 91.80±11.80 68.90±7.80 126.40±11.20 129.60±10.00 79.10±8.80 79.40±8.80
[37] (2008) 25 mg/d 1 mg/d or furosemide,
10 mg/d
Saklayen et al. DKD 24 / 100 Spironolactone, Usual care ACEI/ARB 7 / / 61.91±23.40 54.44±20.09 153.64±25.95 154.52±21.17 79.56±12.23 79.74±12.85
[70] (2008) 25 mg/d
Tylicki et al. [71] CKD 18 42 61 Spironolactone, Usual care ACEI and ARB 2 / / 107.80±51.84 107.80±51.84 115.28±11.84 115.28±11.84 73.83±7.67 73.83±7.67
(2008) 25 mg/d
van den DKD 59 / 56 Spironolactone, Usual care ACEI/ARB 3 / / / / 144.00±17.00 148.00±13.00 80.00±10.00 82.00±8.00
Meiracker et al. 25–50 mg/d
[72] (2006)
Bianchi et al. [73] CKD 165 55 64 Spironolactone, Usual care ACEI/ARB 12 / / 62.40±21.87 62.20±19.02 132.90±7.29 131.60±5.43 78.50±4.56 78.10±3.62
(2006) 25 mg/d
Schjoedt et al. DKD 20 / 85 Spironolactone, Usual care General therapy 2 / / / / / / / /
[74] (2006) 25 mg/d
Ogawa et al. [38] DKD 30 63 / Spironolactone, Furosemide 20 mg/d ACEI/ARB 12 287.00±37.00 269.00±69.00 70.50±14.50 69.80±11.70 154.60±10.10 156.80±9.80 83.10±6.80 86.80±7.90
(2006) 25 mg/d
Chrysostomou CKD 20 63 70 Spironolactone, Usual care ACEI and ARB 3 / / 59.40±19.44 81.60±55.10 141.00±12.90 133.00±19.50 76.00±10.50 79.50±8.60
et al. [75] (2006) 25 mg/d
Chrysostomou CKD 21 56 67 Spironolactone, Usual care ACEI and ARB 3 / / 57.60±27.58 59.70±28.26 130.00±10.20 132.00±11.40 75.50±6.10 79.50±9.00
et al. [75] (2006) 25 mg/d
Takebayashi DKD 37 / / Spironolactone, Amiloride, 2.5 mg/d General therapy 3 / / / / 135.30±8.10 137.60±8.90 71.70±5.70 74.20±8.90
et al. [30] (2006) 50 mg/d
Schjoedt et al. DKD 22 45 75 Spironolactone, Usual care ACEI/ARB 2 / / / / / / / /
[76] (2005) 25 mg/d
Rossing et al. DKD 21 58 85 Spironolactone, Usual care General therapy 4 / / / / / / / /
[77] (2005) 25 mg/d

Continuous variables are expressed as mean ± standard deviation. UACR, urinary albumin-to-creatinine ratio; eGFR, estimated glomerular filtration rate; SBP, systolic blood pressure; DBP, diastolic blood pressure; DKD, diabetic kidney disease; CKD, chronic kidney disease; ACEI, angiotensin
converting enzyme inhibitor; ARB, angiotensin receptor blockers. aPresented as percentage.

Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
Fig. 2. Effects of MRAs on UACR in CKD patients by different MRAs. UACR, urinary albumin-to-creatinine
ratio (expressed in mg/g); WMD, weighted mean difference; MRAs, mineralocorticoid receptor antagonists;
CKD, chronic kidney disease; DKD, diabetic kidney disease.

Effects on Cardiovascular Events Adverse Effects


Eight trials assessed 16,014 patients with 2,124 CVEs. Overall, MRAs significantly increased the incidence
MRAs significantly reduced the CVE risk (8 trials, RR, of hyperkalemia (31 trials, RR, 2.04, 95% CI,
0.84, 95% CI, 0.77–0.92) without significant heterogeneity 1.73–2.40) without significant heterogeneity among
among various MRAs (p for interaction = 0.06) (shown in different MRAs (p for interaction = 0.34). MRAs
online suppl. Fig. 8). significantly increased the incidence of hypotension (7
trials; RR, 1.80, 95% CI, 1.41–2.31), decreased the
Effects on All-Cause Mortality incidence of peripheral edema (3 trials, RR, 0.65, 95%
Five trials included 14,341 participants reported 1,256 CI, 0.56–0.75), whereas they did not significantly in-
death events. MRAs had a trend to decrease all-cause crease the incidence of acute kidney injury (AKI) (4
mortality compared with the control group (5 trials, RR, trials, RR, 0.94, 95% CI, 0.79–1.13), reproductive
0.90, 95% CI, 0.81–1.00). There was a tendency of re- system disorders (4 trials, RR, 1.23, 95% CI,
duction in all-cause mortality by finerenone therapy (2 0.45–3.37), and breast disorders (10 trials, RR, 1.39,
trials, RR, 0.90, 95% CI, 0.80–1.00) (shown in online 95% CI, 0.43–4.48) (shown in Table 2). The incidence
suppl. Fig. 9). of breast disorders was not significantly different

Mineralocorticoid Receptor Antagonists for Am J Nephrol 7


Chronic Kidney Disease DOI: 10.1159/000534366
Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023

(For legend see next page.)

Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
DOI: 10.1159/000534366
Am J Nephrol
8
3
between the male and female participants (gyneco- Compared with the control group, MRAs significantly
mastia: 5 trials, RR, 0.88, 95% CI, 0.42–1.85; breast decreased UACR in DKD patients (6 trials,
disorders in females: 2 trials, RR, 0.17, 95% CI, WMD, −114.41 mg/g, 95% CI, −178.40 to −50.41 mg/g)
0.02–1.40; p for interaction = 0.16). and UPE in non-diabetes CKD patients (1 trials,
WMD, −0.70 g, 95% CI, −1.14 to −0.26 g), whereas they
Publication Bias did not significantly change UACR in non-diabetes CKD
There was significant evidence of publication bias for patients (4 trials, WMD, −21.26 mg/g, 95% CI, −42.66 to
chronic kidney failure events (Egger’s test, p = 0.04), 0.15 mg/g). MRAs significantly decreased eGFR in DKD
whereas there was no significant publication bias in the patients (20 trials, WMD, −3.32 mL/min/1.73 m2, 95%

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
other outcomes (Egger’s test, p > 0.05) (shown in online CI, −4.63 to −2.01 mL/min/1.73 m2), whereas they did
suppl. Fig. 10). not significantly change GFR in non-diabetes CKD
(8 trials, WMD, 1.11 mL/min/1.73 m2 , 95%
Subgroup Analyses CI, −6.38–8.60 mL/min/1.73 m2 ) and unspecified
Subgroup analyses were performed to clarify the effects CKD patients (8 trials, WMD, −0.82 mL/min/1.73 m2 ,
of MRAs in the relationship with ACEI/ARB, in different 95% CI, −2.35 to 0.71 mL/min/1.73 m2) (p for
CKD diseases, and to compare the effects between ste- interaction = 0.04).
roidal and nonsteroidal MRA agents by the stratum of age MRAs more significantly reduced both UACR and
(cutoff point at 60 years old), eGFR (cutoff point at eGFR in the elder patients (UACR: 4 trials,
60 mL/min/1.73 m2), UACR (cutoff point at 300 mg/g), WMD, −189.38 mg/g, 95% CI, −305.83 to −72.93 mg/g;
sample size (cutoff point at 30 participants), and risk of eGFR: 11 trials, WMD, −3.42 mL/min/1.73 m2, 95%
bias (high vs. low/medium risk of bias) (shown in Fig. 4). CI, −4.91 to −1.93 mL/min/1.73 m2) compared with the
Overall, MRAs significantly reduced eGFR compared young patients (UACR: 6 trials, WMD, −24.20 mg/g, 95%
with placebo (32 trials, WMD, −3.04 mL/min/1.73 m2, CI, −39.60 to −8.80 mg/g; eGFR: 22 trials,
95% CI, −4.12 to −1.96 mL/min/1.73 m2). MRAs sig- WMD, −0.20 mL/min/1.73 m2, 95% CI, −2.93 to 2.52 mL/
nificantly reduced UACR (9 trials, WMD, −90.32 mg/g, min/1.73 m2) (p for interaction for UACR = 0.006, p for
95% CI, −149.44 to −31.20 mg/g) and eGFR (27 trials, interaction for eGFR = 0.04).
WMD, −3.35 mL/min/1.73 m2, 95% CI, −4.52 MRAs significantly reduced more UACR in the pa-
to −2.18 mL/min/1.73 m2) compared with placebo, with a tients with high baseline UACR levels (3 trials,
background of ACEi/ARB. MRAs did not significantly WMD, −256.86 mg/g, 95% CI, −425.02 to −88.71 mg/g)
change eGFR compared with placebo, without the compared with those with low baseline UACR levels (7
background of ACEi/ARB (5 trials, WMD, −0.72 mL/ trials, WMD, −43.63 mg/g, 95% CI, −70.91 to −16.35 mg/g)
min/1.73 m2, 95% CI, −3.60 to 2.17 mL/min/1.73 m2). (p for interaction = 0.01). There was no significant difference
MRAs alone did not significantly change UACR (1 trial, in the change of eGFR between the patients with high
WMD, −9.26 mg/g, 95% CI, −31.99 to 13.47 mg/g) and baseline UACR levels and those with low baseline UACR
eGFR (2 trials, WMD, 0.72 mL/min/1.73 m2, 95% levels (p for interaction = 0.37).
CI, −3.63–5.07 mL/min/1.73 m2) compared with ACEI/ There was no significant difference in the change of
ARB alone. UACR between the trials with high risk of bias and those
Nonsteroidal MRAs more significantly reduced UACR with low/medium risk of bias (p for interaction for
(nonsteroidal MRAs, 3 trials, WMD, −189.79, 95% UACR = 0.38). MRAs reduced eGFR more in the trials
CI, −344.42 to −35.16 mg/g; steroidal MRAs, 7 trials, with low/medium risk of bias (17 trials, WMD, −3.67 mL/
WMD, −41.06, 95% CI, −71.06 to −11.06 mg/g, p for min/1.73 m2, 95% CI, −4.85 to −2.49 mL/min/1.73 m2)
interaction = 0.06) and eGFR (nonsteroidal MRAs, 5 compared with those with high risk of bias (19 trials,
trials, WMD, −4.82 mL/min/1.73 m2, 95% CI, −6.79 WMD, 1.45 mL/min/1.73 m2, 95% CI, −1.41–4.31 mL/
to −2.84 mL/min/1.73 m2; steroidal MRAs, 31 trials, min/1.73 m2) (p for interaction = 0.001).
WMD, −0.49 mL/min/1.73 m2, 95% CI, −2.17 to 1.19 mL/ There was no significant difference in the change of
min/1.73 m2; p for interaction = 0.001) compared with UACR and eGFR by MRAs between the patients with
steroidal MRAs. high eGFR and those with low eGFR, and between the

Fig. 3. Effects of MRAs on eGFR in CKD patients by different MRAs. eGFR, estimated glomerular filtration rate
(expressed in mL/min/1.73 m2); WMD, weighted mean difference; MRAs, mineralocorticoid receptor antag-
onists; CKD, chronic kidney disease; DKD, diabetic kidney disease.

Mineralocorticoid Receptor Antagonists for Am J Nephrol 9


Chronic Kidney Disease DOI: 10.1159/000534366
trials with a large sample size and those with a small
sample size (shown in Fig. 4). There was no significant
interaction

heterogeneity among the nonsteroidal MRAs and ste-


p for

0.91

0.34

0.80

0.28

0.99

0.41

0.67
roidal MRAs in the incidence of hyperkalemia (p for

<0.001

<0.001

<0.001
value

0.52

0.69

0.58

0.67
interaction = 0.65) and AKI (p for interaction = 0.91).
p

(95% CI)

Nonsteroidal MRAs (2 trials, RR, 0.66, 95% CI, 0.57–0.75)

23.17)

60.78)

25.67)
(1.83,

(1.12,

(0.10,

(1.26,

(0.35,
3.87)

4.04)
2.66

5.10

2.52

5.68

1.18
RR

but not steroidal MRAs (1 trial, RR, 0.20, 95% CI,


36/1,193
control

0/215

0/118

0/328

4/179
0.02–1.68) significantly reduced the incidence of pe-
spironolactone

ripheral edema. Steroidal MRAs (8 trials, RR, 5.68, 95%


experimental

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
148/1,204

CI, 1.26–25.67) but not nonsteroidal MRAs (2 trials, RR,


11/325
9/212

1/130

5/174
0.52, 95% CI, 0.22–1.22) increased the incidence of breast
(95% CI)

disorders (shown in online suppl. Fig. 11).


71.78)

40.89)

72.69)
(0.13,

(1.10,

(0.55,

(0.02,

(0.12,
2.33)

1.68)
3.00

1.60

4.74

0.20

3.00
RR

/
139/2119
control

Meta-Regression Analyses
0/46

0/92

5/91

0/91

0/91

Univariate meta-regression analyses suggested that for


experimental
eplerenone

254/2,505

every 1 mg/g increase in the baseline UACR, UACR


1/46

4/92

1/91

1/91

0/91

reduction was improved by 0.3 mg/g by the MRAs


therapy (95% CI, −0.4 to −0.2 mg/g; p < 0.001) (shown in
(0.48,
(95%

3.51)
1.30
CI)
RR

Table 3). For every 1 mL/min/1.73 m2 increase in the


control

0/57

5/57

baseline eGFR, UACR reduction was decreased by


ocedurenone

experimental

6.9 mg/d by the MRAs therapy (95% CI, 1.4–12.4 mg/g;


12/105

p = 0.02). With every year increase in the baseline age, the


0/105

effects of MRAs on UACR decreased by 11.8 mg/g (95%


(95% CI)

64.92)

CI, −22.3 to −1.2 mg/g; p = 0.03). For every 1 mm Hg


(0.20,
3.63
RR

increase in the baseline SBP, UACR reduction was im-


control

0/72

proved by 15.1 mg/g (95% CI, −28.6 to −1.52 mg/g; p =


experimental
apararenone

0.04) by the spironolactone therapy (shown in online


5/220

suppl. Table 3). There was no significant association


among the changes of different parameters in the meta-
(1.79,
(95%

8.88)
3.98
CI)
RR

regression analyses for the intervention by eplerenone/


finerenone (shown in online suppl. Tables 4–5).
control

7/302
Table 2. Adverse effects of MRAs as compared with the control

Values for events are given as n/N. MRAs, mineralocorticoid receptor antagonists; RR, relative risk; CI, confidence interval.
experimental
esaxerenone

Sensitivity Analyses
35/369

The effects of MRAs on the outcomes (shown in online


suppl. Fig. 12–18) and the results of subgroup analyses
(0.78,

(1.83,

(1.34,

(0.57,

(0.32,

(0.22,

(0.29,

(0.02,
(95%

1.12)

2.26)

2.23)

0.75)

3.10)

1.22)

1.89)

1.40)
0.94

2.03

1.73

0.66

1.00

0.52

0.74

0.17
CI)
RR

(shown in online suppl. Fig. 19) did not change signifi-


234/6,489

449/6,660

584/6,489
90/3,723

16/6,489

11/6,489

cantly after excluding the five trials employing multiple


5/6,491
control

6/2831

MRA dose groups [60, 65, 69, 74, 28].


experimental
finerenone

220/6,510

919/6,955

157/3,750

384/6,510

8/6,510

8/6,510

0/6,513
6/2827

Discussion
(0.79,

(1.73,

(1.41,

(0.56,

(0.45,

(0.43,

(0.42,

(0.02,
(95%

1.13)

2.40)

2.31)

0.75)

3.37)

4.48)

1.85)

1.40)
0.94

2.04

1.80

0.65

1.23

1.39

0.88

0.17
CI)
RR

641/10,460
234/6,592

589/6,580
90/4,030

16/6,908

15/6,668

The present study consolidated data from extensive


6/3,040

5/6,491
control

trials of MRAs for CKD patients, indicating that in the


experimental

1,385/11,463

CKD patients, MRAs, particularly in combination with


221/6,661

170/4,054

385/6,601

19/6,926

13/6,684
8/3,048

0/6,513
overall
Events

ACEI/ARB, reduced albuminuria/proteinuria, eGFR,


and the incidence of chronic renal failure, cardiovas-
Studies,

cular, and peripheral edema events, whereas increasing


31

10
n

the incidence of hyperkalemia and hypotension


system disorders

Breast disorders

Breast disorders
Adverse events

Gynecomastia
Hyperkalemia
Acute kidney

Reproductive
Hypotension

without the augment of acute kidney injury events.


in females
Peripheral
edema
injury

Nonsteroidal MRAs were superior in the reduction of

10 Am J Nephrol Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
DOI: 10.1159/000534366 Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
a b

Fig. 4. Subgroup analyses for UACR and eGFR. a Subgroup an- medium risk of bias). UACR, urinary albumin-to-creatinine ratio
alyses of MRAs for UACR. b Subgroup analyses of MRAs for (expressed in mg/g); eGFR, estimated glomerular filtration rate
eGFR. Subgroup analyses were performed in the relationship with (expressed in mL/min/1.73 m2); WMD, weighted mean difference;
ACEi/ARB, in different CKD diseases, and to compare the effects MRAs, mineralocorticoid receptor antagonists; RAASi, renin-
between steroidal and nonsteroidal MRA agents by the stratum of angiotensin-aldosterone system inhibitors; ACEI, angiotensin-
age (cutoff point at 60 years old), eGFR (cutoff point at 60 mL/ converting enzyme inhibitor; ARB, angiotensin receptor
min/1.73 m2), UACR (cutoff point at 300 mg/g), sample size blocker; CKD, chronic kidney disease; DKD, diabetic kidney
(cutoff point at 30 participants), and risk of bias (high vs. low/ disease.

more albuminuria with fewer peripheral edema events in the elder patients [81]. The meta-regression analyses
and without the augment of breast disorder events. suggested the UACR reduction by MRAs was reduced
The present study showed that MRAs significantly with the increase of eGFR, which may be due to the more
reduced UACR compared with placebo with background aldosterone excretion in the patients with higher eGFR
of ACEI/ARB, which may be related to the hemodynamic levels [82]. Spironolactone has a more pronounced effect
effects, the preservation of the glycocalyx and modulation on the UACR reduction in the higher baseline SBP pa-
of matrix metalloproteinase (MMP) activity by MRAs, tients, which might be due to its effects in reduction of
and the protective effects for podocyte via the inhibition aldosterone with its attendant renal effects on sodium and
of Rac1 activity of MRAs [78, 79]. Nonsteroidal MRAs, fluid retention in hypertensive patients [83].
with a high selectivity for MR, were superior to steroidal The present study indicated that MRAs decreased
MRAs in the reduction of UACR. UACR was decreased eGFR in CKD patients and reduced chronic kidney
more significantly in the DKD patients compared with failure events. It has been demonstrated that MRAs in-
the non-diabetes CKD patients, which may be due to the tervention may cause an initial acute reduction in eGFR
usage of predominantly nonsteroidal MRAs in the in- and then stabilization of eGFR in the long run [27, 43, 51,
cluded DKD trials [80]. There was an inconsistency that 55] as the effects by ACEI/ARB intervention [84–86].
MRAs significantly reduced UPE but not UACR in the MRAs reduce intraglomerular pressure via impaired
non-diabetes CKD patients; this may be explained by the tubuloglomerular feedback to inhibit proteinuria and
fact that 2 of 4 trials for the assessment of UACR in the protect the kidney in the long term [87]. This hemo-
non-diabetes CKD patients adopted eplerenone (the dynamic effect was demonstrated by the recovery of
other two used spironolactone), whose effect was weaker eGFR after discontinuation of MRAs [26, 33]. In addition,
than that of spironolactone. The effects of MRAs on MRAs may also alleviate renal injury by anti-
UACR were increased by the increase in baseline age, inflammatory, antifibrotic, and antioxidative effects
which might be related to lower plasma aldosterone levels [88]. The reduction of eGFR by MRAs in the present

Mineralocorticoid Receptor Antagonists for Am J Nephrol 11


Chronic Kidney Disease DOI: 10.1159/000534366
Table 3. Metaregression analyses in
the association of eGFR, UACR, SBP, Parameters and covariates Coefficient (95% CI) p value
DBP, serum potassium with the other
eGFR, mL/min/1.73 m2
parameters
Baseline UACR, mg/g 0.0001 (−0.0004 to 0.0006) 0.66
Baseline eGFR, mL/min/1.73 m2 0.07 (−0.09 to 0.23) 0.40
Baseline SBP, mm Hg 0.01 (−0.08 to 0.10) 0.86
Baseline DBP, mm Hg 0.03 (−0.13 to 0.19) 0.70
Age, years −0.12 (−0.31 to 0.06) 0.17
Background therapy
ACEi/ARB 0.6 (−4.2 to 5.4) 0.81

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
UACR, mg/g
Baseline UACR, mg/g −0.3 (−0.4 to −0.2) <0.001
Baseline eGFR, mL/min/1.73 m2 6.9 (1.4–12.4) 0.02
Baseline SBP, mm Hg −3.8 (−15.6 to 8.0) 0.48
Baseline DBP, mm Hg 9.22 (−5.44 to 23.89) 0.19
Age, years −11.8 (−22.3 to −1.2) 0.03
Background therapy
ACEi/ARB −189 (−464 to 86) 0.44
SBP, mm Hg
Baseline UACR, mg/g −0.001 (−0.005 to 0.003) 0.58
Baseline eGFR, mL/min/1.73 m2 0.07 (0.01–0.14) 0.06
Baseline SBP, mm Hg −0.05 (−0.13 to 0.04) 0.26
Baseline DBP, mm Hg −0.03 (−0.18 to 0.11) 0.65
Age, years 0.03 (−0.21 to 0.15) 0.73
Background therapy
ACEi/ARB 3.92 (−1.02 to 8.87) 0.12
DBP, mm Hg
Baseline UACR, mg/g −0.001 (−0.004 to 0.001) 0.34
Baseline eGFR, mL/min/1.73 m2 −0.04 (−0.02 to 0.10) 0.16
Baseline SBP, mm Hg −0.01 (−0.05 to 0.03) 0.51
Baseline DBP, mm Hg −0.01 (−0.07 to 0.06) 0.86
Age, years −0.05 (−0.14 to 0.03) 0.20
Background therapy
ACEi/ARB 1.6 (−1.0 to 4.1) 0.22
Serum potassium, mmol/L
Baseline UACR, mg/g 0.0001 (−0.0001 to 0.0002) 0.36
Baseline eGFR, mL/min/1.73 m2 −0.003 (−0.006 to 0.001) 0.15
Baseline SBP, mm Hg −0.002 (−0.008 to 0.003) 0.41
Baseline DBP, mm Hg −0.001 (−0.002 to 0.001) 0.44
Age, years 0.005 (−0.001 to 0.010) 0.11
Background therapy
ACEi/ARB 0.07 (−0.07 to 0.20) 0.34

There were too few studies of esaxerenone and apararenone to conduct meta-
regression analyses. eGFR, estimated glomerular filtration rate; UACR, urinary albumin-
to-creatinine ratio; SBP, systolic blood pressure; DBP, diastolic blood pressure; ACEi,
angiotensin-converting enzyme inhibitors; ARB, angiotensin II receptor blocker; CI,
confidence interval.

study may be associated with the short period of follow- greater impact on the computation of total slope when
up (≥3 years in only 4 of 36 trials) in the included the total slope is computed over shorter time intervals
trials, reflecting the initial reduction in eGFR by [89]. Here, MRAs significantly reduced chronic renal
MRAs. It has been demonstrated that 3-year total failure events by pooling the four trials with a relative
eGFR slope as the surrogate endpoint can predict long-term follow-up, suggesting the protective effects
kidney benefit, but 2-year total eGFR did not, which of MRAs for CKD patients in the long term. Non-
related to the fact that acute eGFR reduction has a steroidal MRAs, with a higher selectivity for MR, can

12 Am J Nephrol Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
DOI: 10.1159/000534366 Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
reduce eGFR more compared with steroidal MRAs to accumulation of plasma aldosterone with GFR decline
reduce intraglomerular pressure more for glomerular [96, 97]. The risk of hypotension induced by finer-
protection. Finerenone, as the nonsteroidal MRA, can enone therapy was far lower than that by the inter-
more significantly reduce chronic kidney failure vention of eplerenone or spironolactone. Nonsteroidal
events. Subgroup analyses indicated that the elder MRAs, particularly finerenone, did not increase the
patients receiving mainly nonsteroidal MRAs of the risk of breast diseases. These may be associated with
included trial presented with more reduction of eGFR. the high potency and selectivity of finerenone for MR
The funnel plot of chronic kidney failure events and the low half-life of finerenone [20, 98].
exhibited noticeable asymmetry, suggesting publica- The strengths of this work included its comprehensive

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
tion bias that some negative results were not pub- and inclusive approach, robust methodology, and en-
lished. It might also be associated with the limited rolling many trials with large populations. The limitations
number of included trials with the outcome of chronic are the poor quality of some of the included trials.
kidney failure.
The dual blockade of ACEI and ARB in CKD patients
could lower blood pressure and albuminuria more sub- Conclusions
stantially, whereas reduce the glomerular perfusion to
cause AKI [90, 91]. The present study indicated that In the CKD patients, MRAs, particularly in com-
MRAs in combination with ACEI/ARB significantly re- bination with ACEI/ARB, reduced albuminuria/
duced UACR and high glomerular hyperfiltration status proteinuria, eGFR, and the incidence of chronic re-
without augment of AKI events. This might be related to nal failure, and cardiovascular and peripheral edema
the inhibition of “aldosterone breakthrough” and Rac1- events, whereas increasing the incidence of hyper-
induced oxidative stress in smooth muscle cells by adding kalemia and hypotension, without the augment of
MRAs [92], showing a superiority of the combination of acute kidney injury events. Nonsteroidal MRAs were
MRAs and ACEI/ARB over the combination of ACEI and superior in the reduction of more albuminuria with
ARB. MRAs could significantly reduce the incidence of fewer peripheral edema events and without the aug-
CVE and marginally reduce all-cause mortality, which ment of breast disorder events.
may be associated with their anti-inflammatory and
antifibrotic effects [13, 93].
The present study suggested that MRAs increase Acknowledgments
blood potassium levels and the risk of hyperkalemia.
Here, no significant difference existed between ste- We thank Dr. Pitt Bertram and Bakris George L. for providing
us with additional data not shown in the published paper.
roidal MRAs and nonsteroidal MRAs in the incidence
of hyperkalemia, which may be explained by including
the trials of steroidal MRAs with a high MR selectivity
Statement of Ethics
of eplerenone with up to 40-fold less potency than
spironolactone for MR activation with a low risk of An ethics statement is not applicable because this study is based
hyperkalemia [20, 94], as well as the relative high risk exclusively on published literature.
of hyperkalemia by nonsteroidal MRAs of esaxer-
enone. Our meta-analysis found that apararenone and
ocedurenone did not significantly increase the risk of Conflict of Interest Statement
hyperkalemia, and the risk of hyperkalemia of finer-
enone was lower than that of spironolactone. Routine The authors have no conflicts of interest to declare.
potassium monitoring and hyperkalemia management
strategies might minimize the impact of hyperkalemia,
thus providing a basis for the clinical use of nonste- Funding Sources
roidal MRAs [95]. Embracing these innovative agents
not only offered the promise of safeguarding against This research was funded by grants from the State Adminis-
hyperkalemia but also retained the profound thera- tration of Traditional Chinese Medicine and Zhejiang Province
Co-construction Project (No. 2023017961); the project of Zhejiang
peutic advantages inherently associated with MRAs. Traditional Chinese Medicine Administration (No. 2022ZA122);
The effects of MRAs on the reduction of SBP decreased and the project of Basic Public Welfare Research Program of
with the increase of eGFR, which may be due to the Zhejiang Province (No. LGF18H050005).

Mineralocorticoid Receptor Antagonists for Am J Nephrol 13


Chronic Kidney Disease DOI: 10.1159/000534366
Author Contributions Ting Yu, and Xu-Guang Yu. Each author contributed important
intellectual content during manuscript drafting or revision and
Study concept and design: Bin Zhu, Chen-Yi Yuan, and Yuan- gave final approval of the version to be submitted.
Cheng Gao; literature retrieval and data extraction: Chen-Yi Yuan,
Yuan-Cheng Gao, and Bin Zhu; statistical analysis: Chen-Yi Yuan,
Yuan-Cheng Gao, Bin Zhu, Yi Lin, and Lin Liu; interpretation of Data Availability Statement
data: Chen-Yi Yuan, Yuan-cheng Gao, Bin Zhu, Xiao-Gang Shen,
Wen-Li Zou, and Yue-Ming Liu; and quality assessment: Chen-Yi All data generated or analyzed during this study are included in
Yuan, Yuan-Cheng Gao, Bin Zhu, Min-Min Wang, Quan-Quan this article and its online supplementary material. Further in-
Shen, Li-Na Shao, Jia-Wei Zhang, Zhi-Hui Pan, Yan Zhu, Jing- quiries can be directed to the corresponding author.

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
References

1 Johansen KL, Chertow GM, Gilbertson DT, kidney and cardiovascular outcomes: a post hoc steroidal mineralocorticoid receptor antago-
Ishani A, Israni A, Ku E, et al. US renal data analysis from the CREDENCE trial. J Am Soc nists in cardiorenal medicine. Eur Heart J.
system 2022 annual data report: epidemiology Nephrol. 2020 Dec;31(12):2925–36. 2020;42(2):152–61.
of kidney disease in the United States. Am 13 Kolkhof P, Jaisser F, Kim SY, Filippatos G, 21 Pitt B, Filippatos G, Agarwal R, Anker SD, Bakris
J Kidney Dis. 2023 Mar;81(3 Suppl 1):A8–11. Nowack C, Pitt B. Steroidal and novel non- GL, Rossing P, et al. Cardiovascular events with
2 Kovesdy CP. Epidemiology of chronic kidney steroidal mineralocorticoid receptor antago- finerenone in kidney disease and type 2 diabetes.
disease: an update 2022. Kidney Int Suppl. nists in heart failure and cardiorenal diseases: N Engl J Med. 2021 Dec 9;385(24):2252–63.
2022 Apr;12(1):7–11. comparison at bench and bedside. Handb 22 Jiang X, Zhang Z, Li C, Zhang S, Su Q, Yang S,
3 Li H, Lu W, Wang A, Jiang H, Lyu J. Exp Pharmacol. 2017;243:271–305. et al. Efficacy and safety of non-steroidal
Changing epidemiology of chronic kidney 14 Epstein M. Aldosterone and mineralocorti- mineralocorticoid receptor antagonists in
disease as a result of type 2 diabetes mellitus coid receptor signaling as determinants of patients with chronic kidney disease and type
from 1990 to 2017: estimates from Global cardiovascular and renal injury: an extraor- 2 diabetes: a systematic review incorporating
Burden of Disease 2017. J Diabetes Investig. dinary paradigm shift. Am J Nephrol. 2021; an indirect comparisons meta-analysis. Front
2021 Mar;12(3):346–56. 52(3):209–16. Pharmacol. 2022;13:896947.
4 Kearney PM, Whelton M, Reynolds K, 15 Alexandrou ME, Papagianni A, Tsapas A, 23 Vrabel M. Preferred reporting items for sys-
Muntner P, Whelton PK, He J. Global burden Loutradis C, Boutou A, Piperidou A, et al. tematic reviews and meta-analyses: the PRIS-
of hypertension: analysis of worldwide data. Effects of mineralocorticoid receptor antag- MA statement. Revista Espaola De Nutrición
Lancet. 2005;365(9455):217–23. onists in proteinuric kidney disease: a sys- Humana Y Dietética. 2009;18(3):e123.
5 Umanath K, Lewis JB, LewisJulia B. Update tematic review and meta-analysis of ran- 24 Higgins JPTTJ, Chandler J, Cumpston M, Li T,
on diabetic nephropathy: core curriculum domized controlled trials. J Hypertens. 2019 Page MJ, Welch VA. Cochrane handbook for
2018. Am J Kidney Dis. 2018;71(6):884–95. Dec;37(12):2307–24. systematic reviews of interventionsversion 6.3.
6 Kidney Disease Improving Global Outcomes 16 Takahashi M, Ubukata O, Homma T, Asoh Cochrane; 2022 [updated February 2022].
KDIGO Diabetes Work Group. KDIGO 2020 Y, Honzumi M, Hayashi N, et al. Crystal 25 DerSimonian R, Laird N. Meta-analysis in
clinical practice guideline for diabetes man- structure of the mineralocorticoid receptor clinical trials revisited. Contemp Clin Trials.
agement in chronic kidney disease. Kidney ligand-binding domain in complex with a 2015 Nov;45(Pt A):139–45.
Int. 2020;98(4S):S1–115. potent and selective nonsteroidal blocker, 26 Ito S, Shikata K, Nangaku M, Okuda Y, Sa-
7 American Diabetes Association. 11. Micro- esaxerenone (CS-3150). FEBS Lett. 2020 May; wanobori T. Efficacy and safety of esaxer-
vascular complications and foot care: stan- 594(10):1615–23. enone (CS-3150) for the treatment of type 2
dards of medical care in diabetes-2020. Di- 17 Barrera-Chimal J, Kolkhof P, Lima-Posada I, diabetes with microalbuminuria: a random-
abetes Care. 2020 Jan;43(Suppl 1):S135–51. Joachim A, Rossignol P, Jaisser F. Differen- ized, double-blind, placebo-controlled, phase
8 Joffe HV, Adler GK. Effect of aldosterone and tiation between emerging non-steroidal and II trial. Clin J Am Soc Nephrol. 2019 Aug 7;
mineralocorticoid receptor blockade on vascular established steroidal mineralocorticoid re- 14(8):1161–72.
inflammation. Heart Fail Rev. 2005 Jan; ceptor antagonists: head-to-head compari- 27 Ito S, Kashihara N, Shikata K, Nangaku M,
10(1):31–7. sons of pharmacological and clinical char- Wada T, Okuda Y, et al. Esaxerenone (CS-3150)
9 Patel V, Joharapurkar A, Jain M. Role of acteristics. Expert Opin Investig Drugs. 2021 in patients with type 2 diabetes and micro-
mineralocorticoid receptor antagonists in Nov;30(11):1141–57. albuminuria (ESAX-DN): phase 3 randomized
kidney diseases. Drug Dev Res. 2021 May; 18 Nakamura T, Kawaguchi A. Phase 1 studies controlled clinical trial. Clin J Am Soc Nephrol.
82(3):341–63. to define the safety, tolerability, and phar- 2020 Dec 7;15(12):1715–27.
10 Schwenk MH, Hirsch JS, Bomback AS. Al- macokinetic and pharmacodynamic profiles 28 Wada T, Inagaki M, Yoshinari T, Terata R,
dosterone blockade in CKD: emphasis on of the nonsteroidal mineralocorticoid re- Totsuka N, Gotou M, et al. Apararenone in
pharmacology. Adv Chronic Kidney Dis. ceptor antagonist apararenone in healthy patients with diabetic nephropathy: results of
2015 Mar;22(2):123–32. volunteers. Clin Pharmacol Drug Dev. 2021 a randomized, double-blind, placebo-
11 Perkovic V, Jardine MJ, Neal B, Bompoint S, Apr;10(4):353–65. controlled phase 2 dose-response study and
Heerspink HJL, Charytan DM, et al. Cana- 19 Bakris GL, Agarwal R, Anker SD, Pitt B, open-label extension study. Clin Exp Neph-
gliflozin and renal outcomes in type 2 dia- Ruilope LM, Rossing P, et al. Effect of fi- rol. 2021 Feb;25(2):120–30.
betes and nephropathy. N Engl J Med. 2019 nerenone on chronic kidney disease out- 29 Bakris G, Pergola PE, Delgado B, Genov D,
Jun 13;380(24):2295–306. comes in type 2 diabetes. N Engl J Med. 2020 Doliashvili T, Vo N, et al. Effect of KBP-5074
12 Oshima M, Neuen BL, Li J, Perkovic V, Dec 3;383(23):2219–29. on blood pressure in advanced chronic kid-
Charytan DM, de Zeeuw D, et al. Early change 20 Agarwal R, Kolkhof P, Bakris G, Bauersachs J, ney disease: results of the BLOCK-CKD
in albuminuria with canagliflozin predicts Haller H, Wada T, et al. Steroidal and non- study. Hypertension. 2021 Jul;78(1):74–81.

14 Am J Nephrol Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
DOI: 10.1159/000534366 Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
30 Takebayashi K, Matsumoto S, Aso Y, Inukai 40 Katayama S, Yamada D, Nakayama M, Yamada lective aldosterone blockade with eplerenone
T. Aldosterone blockade attenuates urinary T, Myoishi M, Kato M, et al. A randomized reduces albuminuria in patients with type 2
monocyte chemoattractant protein-1 and controlled study of finerenone versus placebo in diabetes. Clin J Am Soc Nephrol. 2006 Sep;
oxidative stress in patients with type 2 dia- Japanese patients with type 2 diabetes mellitus 1(5):940–51.
betes complicated by diabetic nephropathy. and diabetic nephropathy. J Diabetes Compli- 51 Tofte N, Lindhardt M, Adamova K, Bakker
J Clin Endocrinol Metab. 2006 Jun;91(6): cations. 2017 Apr;31(4):758–65. SJL, Beige J, Beulens JWJ, et al. Early detec-
2214–7. 41 Bakris GL, Agarwal R, Chan JC, Cooper ME, tion of diabetic kidney disease by urinary
31 Morales E, Huerta A, Gutiérrez E, Gutiérrez Gansevoort RT, Haller H, et al. Effect of fi- proteomics and subsequent intervention with
Solís E, Segura J, Praga M. The anti- nerenone on albuminuria in patients with spironolactone to delay progression (PRI-
proteinuric effect of the blockage of the renin- diabetic nephropathy: a randomized clinical ORITY): a prospective observational study
angiotensin-aldosterone system (RAAS) in trial. JAMA. 2015 Sep 1;314(9):884–94. and embedded randomised placebo-
obese patients. Which treatment option is the 42 Pitt B, Kober L, Ponikowski P, Gheorghiade controlled trial. Lancet Diabetes Endo-

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
most effective?. Nefrologia. 2009;29(5): M, Filippatos G, Krum H, et al. Safety and crinol. 2020 Apr;8(4):301–12.
421–9. tolerability of the novel non-steroidal min- 52 Beldhuis IE, Myhre PL, Claggett B, Damman
32 El Mokadem M, Abd El Hady Y, Aziz A. A eralocorticoid receptor antagonist BAY 94- K, Fang JC, Lewis EF, et al. Efficacy and safety
prospective single-blind randomized trial of 8862 in patients with chronic heart failure of spironolactone in patients with HFpEF and
ramipril, eplerenone and their combination and mild or moderate chronic kidney disease: chronic kidney disease. JACC Heart Fail.
in type 2 diabetic nephropathy. Cardiorenal a randomized, double-blind trial. Eur Heart J. 2019 Jan;7(1):25–32.
Med. 2020;10(6):392–401. 2013 Aug;34(31):2453–63. 53 Chen Y, Liu P, Chen X, Li Y, Zhang F, Wang
33 Provenzano M, Puchades MJ, Garofalo C, 43 Minakuchi H, Wakino S, Urai H, Kur- Y. Effects of different doses of irbesartan
Jongs N, D’Marco L, Andreucci M, et al. okochi A, Hasegawa K, Kanda T, et al. The combined with spironolactone on urinary
Albuminuria-lowering effect of dapagliflozin, effect of aldosterone and aldosterone albumin excretion rate in elderly patients
eplerenone, and their combination in patients blockade on the progression of chronic with early type 2 diabetic nephropathy. Am
with chronic kidney disease: a randomized kidney disease: a randomized placebo- J Med Sci. 2018 May;355(5):418–24.
crossover clinical trial. J Am Soc Nephrol. controlled clinical trial. Sci Rep. 2020 54 Srivastava A, Adams-Huet B, Vega GL, Toto
2022 Aug;33(8):1569–80. Oct 6;10(1):16626. RD. Effect of losartan and spironolactone on
34 Joffe HV, Kwong RY, Gerhard-Herman MD, 44 Ferreira JP, Abreu P, McMurray JJV, van triglyceride-rich lipoproteins in diabetic ne-
Rice C, Feldman K, Adler GK. Beneficial Veldhuisen DJ, Swedberg K, Pocock SJ, et al. phropathy. J Investig Med. 2016 Aug;64(6):
effects of eplerenone versus hydrochlorothi- Renal function stratified dose comparisons of 1102–8.
azide on coronary circulatory function in eplerenone versus placebo in the EMPHA- 55 Kato S, Maruyama S, Makino H, Wada J,
patients with diabetes mellitus. J Clin En- SIS-HF trial. Eur J Heart Fail. 2019 Mar;21(3): Ogawa D, Uzu T, et al. Anti-albuminuric
docrinol Metab. 2007 Jul;92(7):2552–8. 345–51. effects of spironolactone in patients with
35 Momeni A, Behradmanesh MS, Kheiri S, 45 Ando K, Ohtsu H, Uchida S, Kaname S, type 2 diabetic nephropathy: a multicenter,
Karami Horestani M. Evaluation of spi- Arakawa Y, Fujita T, et al. Anti-albuminuric randomized clinical trial. Clin Exp Nephrol.
ronolactone plus hydrochlorothiazide in re- effect of the aldosterone blocker eplerenone 2015 Dec;19(6):1098–106.
ducing proteinuria in type 2 diabetic ne- in non-diabetic hypertensive patients with 56 Moras N, Caro J, Urrz D, Svano A, Auñón P,
phropathy. J Renin Angiotensin Aldosterone albuminuria: a double-blind, randomised, Frnandz C, et al. Diverse diuretics regimens
Syst. 2015 Mar;16(1):113–8. placebo-controlled trial. Lancet Diabetes differentially enhance the antialbuminuric
36 Morales E, Caro J, Gutierrez E, Sevillano A, Endocrinol. 2014 Dec;2(12):944–53. effect of renin–angiotensin blockers in pa-
Auñón P, Fernandez C, et al. Diverse di- 46 Boesby L, Elung-Jensen T, Strandgaard S, tients with chronic kidney disease. Kidney
uretics regimens differentially enhance the Kamper AL. Eplerenone attenuates pulse Int. 2015;88(6):1434–41.
antialbuminuric effect of renin- wave reflection in chronic kidney disease 57 Van Buren PN, Adams-Huet B, Nguyen M,
angiotensin blockers in patients with stage 3-4: a randomized controlled study. Molina C, Toto RD. Potassium handling with
chronic kidney disease. Kidney Int. 2015 PLoS One. 2013;8(5):e64549. dual renin-angiotensin system inhibition in
Dec;88(6):1434–41. 47 Eschalier R, McMurray JJ, Swedberg K, van diabetic nephropathy. Clin J Am Soc Neph-
37 Furumatsu Y, Nagasawa Y, Tomida K, Mi- Veldhuisen DJ, Krum H, Pocock SJ, et al. rol. 2014 Feb;9(2):295–301.
kami S, Kaneko T, Okada N, et al. Effect of Safety and efficacy of eplerenone in patients 58 Esteghamati A, Noshad S, Jarrah S,
renin-angiotensin-aldosterone system triple at high risk for hyperkalemia and/or wors- Mousavizadeh M, Khoee SH, Nakhjavani M.
blockade on non-diabetic renal disease: ad- ening renal function: analyses of the EM- Long-term effects of addition of mineralo-
dition of an aldosterone blocker, spi- PHASIS-HF study subgroups (Eplerenone in corticoid receptor antagonist to angiotensin
ronolactone, to combination treatment with mild patients hospitalization and Survival II receptor blocker in patients with diabetic
an angiotensin-converting enzyme inhibitor Study in Heart Failure). J Am Coll Cardiol. nephropathy: a randomized clinical trial.
and angiotensin II receptor blocker. Hyper- 2013 Oct 22;62(17):1585–93. Nephrol Dial Transplant. 2013 Nov;28(11):
tens Res. 2008 Jan;31(1):59–67. 48 Tylicki L, Lizakowski S, Rutkowski P, Renke 2823–33.
38 Ogawa S, Takeuchi K, Mori T, Nako K, Ito S. M, Sulikowska B, Heleniak Z, et al. The en- 59 Wang W, Li L, Zhou Z, Gao J, Sun Y. Effect of
Spironolactone further reduces urinary al- hanced renin-angiotensin-aldosteron system spironolactone combined with angiotensin-
bumin excretion and plasma B-type natri- pharmacological blockade: which is the best? converting enzyme inhibitors and/or angioten-
uretic peptide levels in hypertensive type II Kidney Blood Press Res. 2012;36(1):335–43. sin II receptor blockers on chronic glomerular
diabetes treated with angiotensin-converting 49 Boesby L, Elung-Jensen T, Klausen TW, disease. Exp Ther Med. 2013 Dec;6(6):1527–31.
enzyme inhibitor. Clin Exp Pharmacol Strandgaard S, Kamper AL. Moderate anti- 60 Nielsen SE, Schjoedt KJ, Rossing K, Persson
Physiol. 2006;33(5–6):477–9. proteinuric effect of add-on aldosterone F, Schalkwijk CG, Stehouwer CD, et al. Levels
39 Hase M, Babazono T, Ujihara N, Uchigata Y. blockade with eplerenone in non-diabetic of NT-proBNP, markers of low-grade in-
Comparison of spironolactone and tri- chronic kidney disease. A randomized flammation, and endothelial dysfunction
chlormethiazide as add-on therapy to renin- cross-over study. PLoS One. 2011;6(11): during spironolactone treatment in patients
angiotensin blockade for reduction of albu- e26904. with diabetic kidney disease. J Renin An-
minuria in diabetic patients. J Diabetes In- 50 Epstein M, Williams GH, Weinberger M, giotensin Aldosterone Syst. 2013 Jun;14(2):
vestig. 2013 May 6;4(3):316–9. Lewin A, Krause S, Mukherjee R, et al. Se- 161–6.

Mineralocorticoid Receptor Antagonists for Am J Nephrol 15


Chronic Kidney Disease DOI: 10.1159/000534366
61 Ziaee A, Abbas Vaezi A, Oveisi S, Javadi A, and renal function. J Hypertens. 2006 Nov; induced fall in glomerular filtration rate
Hashemipour S, Kazemifar AM. Effects of 24(11):2285–92. predicts long-term stability of renal function.
additive therapy with spironolactone on al- 73 Bianchi S, Bigazzi R, Campese VM. Long- Kidney Int. 1997 Mar;51(3):793–7.
buminuria in diabetes mellitus: a pilot ran- term effects of spironolactone on proteinuria 85 Holtkamp FA, de Zeeuw D, Thomas MC,
domized clinical trial. Caspian J Intern Med. and kidney function in patients with chronic Cooper ME, de Graeff PA, Hillege HJ, et al. An
2013 Spring;4(2):648–53. kidney disease. Kidney Int. 2006 Dec;70(12): acute fall in estimated glomerular filtration rate
62 Nielsen SE, Persson F, Frandsen E, Sugaya T, 2116–23. during treatment with losartan predicts a slower
Hess G, Zdunek D, et al. Spironolactone di- 74 Schjoedt KJ, Rossing K, Juhl TR, Boomsma F, decrease in long-term renal function. Kidney
minishes urinary albumin excretion in patients Tarnow L, Rossing P, et al. Beneficial impact Int. 2011 Aug;80(3):282–7.
with type 1 diabetes and microalbuminuria: a of spironolactone on nephrotic range albu- 86 Bakris GL, Weir MR. Initial drops in glo-
randomized placebo-controlled crossover study. minuria in diabetic nephropathy. Kidney Int. merular filtration rate with certain drug
Diabet Med. 2012 Aug;29(8):e184–90. 2006 Aug;70(3):536–42. classes retard kidney disease progression. Am

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023
63 Edwards NC, Steeds RP, Chue CD, Stewart 75 Chrysostomou A, Pedagogos E, MacGregor J Nephrol. 2022;53(7):513–5.
PM, Ferro CJ, Townend JN. The safety and L, Becker GJ. Double-Blind, placebo- 87 Fu Y, Hall JE, Lu D, Lin L, Manning RD Jr,
tolerability of spironolactone in patients with controlled study on the effect of the aldo- Cheng L, et al. Aldosterone blunts tubulo-
mild to moderate chronic kidney disease. Br sterone receptor antagonist spironolactone in glomerular feedback by activating macula
J Clin Pharmacol. 2012 Mar;73(3):447–54. patients who have persistent proteinuria and densa mineralocorticoid receptors. Hyper-
64 Abolghasmi R, Taziki O. Efficacy of low dose are on long-term angiotensin-converting tension. 2012 Mar;59(3):599–606.
spironolactone in chronic kidney disease with enzyme inhibitor therapy, with or without 88 Barrera-Chimal J, Girerd S, Jaisser F. Min-
resistant hypertension. Saudi J Kidney Dis an angiotensin II receptor blocker. Clin J Am eralocorticoid receptor antagonists and kid-
Transpl. 2011 Jan;22(1):75–8. Soc Nephrol. 2006;1(2):256–62. ney diseases: pathophysiological basis. Kid-
65 Zheng H, Qiu H, Liu X. Clinical investigation 76 Schjoedt KJ, Rossing K, Juhl TR, Boomsma ney Int. 2019 Aug;96(2):302–19.
of combined therapy with benazepril and F, Rossing P, Tarnow L, et al. Beneficial 89 Inker LA, Collier W, Greene T, Miao S,
spironolactone in diabetic nephropathy. impact of spironolactone in diabetic ne- Chaudhari J, Appel GB, et al. A meta-analysis
Pharma Care Res. 2011;11(4):266–8. phropathy. Kidney Int. the2005;68(6): of GFR slope as a surrogate endpoint for
66 Hammer F, Edwards NC, Hughes BA, Steeds 2829–36. kidney failure. Nat Med. 2023 Jul;29(7):
RP, Ferro CJ, Townend JN, et al. The effect of 77 Rossing K, Schjoedt KJ, Smidt UM, Boomsma 1867–76.
spironolactone upon corticosteroid hormone F, Parving HH. Beneficial effects of adding 90 Yusuf S, Teo KK, Pogue J, Dyal L, Copland I,
metabolism in patients with early stage spironolactone to recommended antihyper- Schumacher H, et al. Telmisartan, ramipril,
chronic kidney disease. Clin Endocrinol. 2010 tensive treatment in diabetic nephropathy: a or both in patients at high risk for vascular
Nov;73(5):566–72. randomized, double-masked, cross-over events. N Engl J Med. 2008 Apr 10;358(15):
67 Guney I, Selcuk NY, Altintepe L, Atalay H, study. Diabetes Care. 2005 Sep;28(9): 1547–59.
Başarali MK, Büyükbaş S. Antifibrotic effects 2106–12. 91 Rutkowski B, Tylicki L. Nephroprotective
of aldosterone receptor blocker (spi- 78 Shibata S, Nagase M, Yoshida S, Kawarazaki action of renin-angiotensin-aldosterone sys-
ronolactone) in patients with chronic kidney W, Kurihara H, Tanaka H, et al. Modification tem blockade in chronic kidney disease pa-
disease. Ren Fail. 2009;31(9):779–84. of mineralocorticoid receptor function by tients: the landscape after ALTITUDE and
68 Mehdi UF, Adams-Huet B, Raskin P, Vega Rac1 GTPase: implication in proteinuric VA NEPHRON-D trails. J Ren Nutr. 2015;
GL, Toto RD. Addition of angiotensin re- kidney disease. Nat Med. 2008 Dec;14(12): 25(2):194–200.
ceptor blockade or mineralocorticoid antag- 1370–6. 92 Barrera-Chimal J, André-Grégoire G,
onism to maximal angiotensin-converting 79 Crompton M, Ferguson JK, Ramnath RD, Nguyen Dinh Cat A, Lechner SM, Cau J,
enzyme inhibition in diabetic nephropathy. Onions KL, Ogier AS, Gamez M, et al. Prince S, et al. Benefit of mineralocorticoid
J Am Soc Nephrol. 2009 Dec;20(12):2641–50. Mineralocorticoid receptor antagonism in receptor antagonism in AKI: role of vascular
69 Preston RA, Afshartous D, Garg D, Medrano diabetes reduces albuminuria by preserving smooth muscle Rac1. J Am Soc Nephrol. 2017
S, Alonso AB, Rodriguez R. Mechanisms of the glomerular endothelial glycocalyx. JCI Apr;28(4):1216–26.
impaired potassium handling with dual Insight. 2023 Mar 8;8(5):e154164. 93 Barrera-Chimal J, Estrela GR, Lechner SM,
renin-angiotensin-aldosterone blockade in 80 Ferreira NS, Bruder-Nascimento T, Pereira Giraud S, El Moghrabi S, Kaaki S, et al. The
chronic kidney disease. Hypertension. 2009 CA, Zanotto CZ, Prado DS, Silva JF, et al. myeloid mineralocorticoid receptor controls
May;53(5):754–60. NLRP3 inflammasome and mineralocorti- inflammatory and fibrotic responses after
70 Saklayen MG, Gyebi LK, Tasosa J, Yap J. coid receptors are associated with vascular renal injury via macrophage interleukin-4
Effects of additive therapy with spi- dysfunction in type 2 diabetes mellitus. Cells. receptor signaling. Kidney Int. 2018 Jun;
ronolactone on proteinuria in diabetic pa- 2019 Dec 8;8(12):1595. 93(6):1344–55.
tients already on ACE inhibitor or ARB 81 Nanba K, Vaidya A, Rainey WE. Aging and 94 Garthwaite SM, McMahon EG. The evolution
therapy: results of a randomized, placebo- adrenal aldosterone production. Hyperten- of aldosterone antagonists. Mol Cell Endo-
controlled, double-blind, crossover trial. sion. 2018 Feb;71(2):218–23. crinol. 2004 Mar 31;217(1–2):27–31.
J Investig Med. 2008 Apr;56(4):714–9. 82 Roldán J, Morillas P, Castillo J, Andrade H, 95 Agarwal R, Joseph A, Anker SD, Filippatos G,
71 Tylicki L, Rutkowski P, Renke M, Larczyński Guillén S, Núñez D, et al. Plasma aldosterone Rossing P, Ruilope LM, et al. Hyperkalemia
W, Aleksandrowicz E, Lysiak-Szydlowska W, and glomerular filtration in hypertensive risk with finerenone: results from the FI-
et al. Triple pharmacological blockade of the patients with preserved renal function. Rev DELIO-DKD trial. J Am Soc Nephrol. 2022
renin-angiotensin-aldosterone system in Esp Cardiol. 2010 Jan;63(1):103–6. Jan;33(1):225–37.
nondiabetic CKD: an open-label crossover 83 Chen C, Zhu XY, Li D, Lin Q, Zhou K. 96 Bramlage P, Swift SL, Thoenes M, Minguet J,
randomized controlled trial. Am J Kidney Clinical efficacy and safety of spironolactone Ferrero C, Schmieder RE. Non-steroidal
Dis. 2008 Sep;52(3):486–93. in patients with resistant hypertension: a mineralocorticoid receptor antagonism for
72 van den Meiracker AH, Baggen RG, Pauli S, systematic review and meta-analysis. Medi- the treatment of cardiovascular and renal
Lindemans A, Vulto AG, Poldermans D, et al. cine. 2020 Aug 21;99(34):e21694. disease (vol 18, pg 28, 2016). Eur J Heart Fail:
Spironolactone in type 2 diabetic nephrop- 84 Apperloo AJ, de Zeeuw D, de Jong PE. A J Working Group Heart Fail Eur Soc Cardiol.
athy: effects on proteinuria, blood pressure short-term antihypertensive treatment- 2017(6):19.

16 Am J Nephrol Yuan/Gao/Lin/Liu/Shen/Zou/Wang/Shen/
DOI: 10.1159/000534366 Shao/Liu/Zhang/Pan/Zhu/Yu/Yu/Zhu
97 Epstein M, Kovesdy CP, Clase CM, Sood 98 Lerma E, White WB, Bakris G. Effec- 99 Rossing K, Schjoedt KJ, Smidt UM, Boomsma F,
MM, Pecoits-Filho R. Aldosterone, min- tiveness of nonsteroidal mineralocorti- Parving HH. Beneficial effects of adding spi-
eralocorticoid receptor activation, and coid receptor antagonists in patients ronolactone to recommended antihypertensive
CKD: a review of evolving treatment par- with diabetic kidney disease. treatment in diabetic nephropathy: a random-
adigms. Am J Kidney Dis. 2022 Nov;80(5): Postgrad Med. 2022 Apr 20;135(3): ized, double-masked, cross-over study. Diabetes
658–66. 224–33. Care. 2005 Sep;28(9):2106–12.

Downloaded from http://karger.com/ajn/article-pdf/doi/10.1159/000534366/4048477/000534366.pdf by The Library of Nanjing Medical University user on 19 November 2023

Mineralocorticoid Receptor Antagonists for Am J Nephrol 17


Chronic Kidney Disease DOI: 10.1159/000534366

You might also like