Next Article in Journal
Ren.Nu, a Dietary Program for Individuals with Autosomal-Dominant Polycystic Kidney Disease Implementing a Sustainable, Plant-Focused, Kidney-Safe, Ketogenic Approach with Avoidance of Renal Stressors
Next Article in Special Issue
The Pathophysiological Basis of Diabetic Kidney Protection by Inhibition of SGLT2 and SGLT1
Previous Article in Journal
Future Directions for Dialysis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease

by
Maria-Eleni Alexandrou
,
Marieta P. Theodorakopoulou
and
Pantelis A. Sarafidis
*
Department of Nephrology, Hippokration Hospital, Aristotle University of Thessaloniki, GR54642 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Kidney Dial. 2022, 2(2), 163-182; https://doi.org/10.3390/kidneydial2020019
Submission received: 22 February 2022 / Revised: 30 March 2022 / Accepted: 1 April 2022 / Published: 8 April 2022
(This article belongs to the Special Issue Diabetic Kidney Disease)

Abstract

:
Diabetic kidney disease (DKD) represents a major public health issue, currently posing an important burden on healthcare systems. Renin–angiotensin system (RAS) blockers are considered the cornerstone of treatment of albuminuric DKD. However, a high residual risk of progression to more advanced CKD stages under RAS blockade still remains, while relevant studies did not show significant declines in cardiovascular events with these agents in patients with DKD. Among several other pharmacological classes, mineralocorticoid receptor antagonists (MRAs) have received increasing interest, due to a growing body of high-quality evidence showing that spironolactone and eplerenone can significantly lower blood pressure and albuminuria in patients with CKD. Furthermore, finerenone, a novel nonsteroidal MRA with unique physicochemical properties, was shown to effectively reduce cardiovascular events and death, as well as the incidence of end-stage kidney disease in patients with type 2 diabetes. This review discusses previous and recent clinical evidence on the issue of nephroprotection and cardioprotection in DKD offered by mineralocorticoid receptor antagonism, aiming to aid clinicians in their treatment decisions for diabetic patients.

1. Introduction

Diabetes represents a worldwide silent epidemic, affecting in 2019 9.3% of the adult population and accounting for 4 million deaths in 2017 [1]. The increase in diabetes prevalence has led to an increase in patients diagnosed with diabetic kidney disease (DKD), a complication occurring in 20–40% of diabetic patients [2]. DKD is the leading cause of end-stage kidney disease (ESKD) [3], with renal replacement therapy posing a major economic challenge in high-income countries (3–5% of national healthcare budgets in Europe) [4]. In patients with diabetes and chronic kidney disease (CKD), 10-year cumulative mortality is nearly three-fold higher compared to people with diabetes without CKD (31.1% vs. 11.5%) [4]. Thus, comprehension of the pathophysiology of DKD and identification of new directions for treatment strategies is crucial for health systems.
Over the last decades, the use of renin–angiotensin system (RAS) blockers has been the mainstay for retarding progression of DKD [5,6], along with lifestyle modifications and blood pressure (BP) and glycemic control [7]. Despite the indisputable nephroprotective effects of RAS blockers, accumulated evidence suggests persistence of a high residual risk for CKD and cardiovascular (CVD) progression in these patients, underlining the need for further research to establish novel treatment approaches. Double RAS blockade, initially associated with greater reductions in albuminuria compared to single blockade [8], was terminally abandoned in the form of a combination of an angiotensin converting enzyme inhibitor (ACEi) with an angiotensin receptor blocker [ARB], or of a renin inhibitor with an ACEi/ARB in diabetics and all other populations, in view of safety concerns raised after publication of two major cardiovascular and renal outcome trials [9,10]. In contrast, sodium–glucose cotransporter-2 inhibitors (SGLT2i) have shown to offer kidney protection and are now included in recent KDIGO recommendations as preferred first line treatment in patients with type 2 diabetes mellitus (DM) and estimated glomerular-filtration rate (eGFR) ≥ 30 mL/min/1.73 m2 [7].
Steroidal mineralocorticoid receptor antagonists (MRAs) have proven to be effective in the management of primary aldosteronism due to bilateral adrenal hyperplasia or aldosterone-producing adrenal adenomas [11], in the treatment of resistant hypertension [12,13], as well as in the reduction of albuminuria in patients with diabetic and non-diabetic nephropathy (alone or on top of an RAS blocker) [14]. Moreover, the accumulated clinical trial evidence supporting a significant reduction in mortality and CV events in patients with heart failure (HF) with reduced ejection fraction with MRAs changed the landscape of HF treatment [15,16,17], offering these drug agents a Class 1A recommendation [18]. Subgroup and post hoc analyses [15,19] of the latter studies showed improved clinical outcomes irrespective of baseline kidney function; however, patients with more advanced CKD stages (i.e., eGFR < 30 mL/min/1.73 m2) had been excluded.
The risk of hyperkalemia remains a major determinant of MRA use, especially for the older first- and second-generation steroidal agents spironolactone and eplerenone [20]; an acute, usually reversible, decline in glomerular filtration observed among patients with low eGFR was another problem, particularly when these drugs were used in conjunction with an ACEi or an ARB [21]. In recent years, novel nonsteroidal agents have been developed, i.e., finerenone, esaxerenone and apararenone, with the former having been more extensively studied in diabetic populations. Finerenone belongs to third-generation MRAs and has demonstrated a more potent and selective antagonism for mineralocorticoid receptors (MR) compared to spironolactone and eplerenone, with a balanced distribution between heart and kidney tissue and different agonistic effects with regards to coregulatory molecules, rendering it a more potent anti-inflammatory and anti-fibrotic agent [22]. Large-scale clinical trials have recently provided evidence of improved renal and cardiovascular outcomes with addition of finerenone to the standard of care in patients with type 2 DM and moderately or severely increased albuminuria [23,24]. The aim of this review is to summarize available evidence on the effects of MRAs on BP, albuminuria and hard outcomes in patients with DKD, in whom background treatment has failed to delay CKD progression and novel drug regimens are being sought to address the residual CV risk.

2. Pleiotropic Effects of Aldosterone in Diabetic Kidney Disease

2.1. Physiological Role of Aldosterone and Mechanisms of Ligand-Specific Activation of Mineralocorticoid Receptors

Aldosterone is a steroid hormone that is produced by the adrenal cortex and acts, together with other members of the steroid hormone family (cortisol and corticosterone), as a ligand of MRs [25]. MRs are nuclear receptors, structurally similar to glucocorticoid receptors (GRs), that are expressed in epithelial and non-epithelial tissues, serving as transcription factors of target genes that regulate cellular processes [25] (Figure 1). In epithelial cells of the distal nephron, where both MR and GR are expressed, aldosterone exerts its classical actions with regards to volume depletion and hyperkalemia by regulating sodium, chloride and potassium handling, through transcription of the epithelial sodium channel (ENaC), Cl/HCO3 exchangers and ROMK channels [26,27]. Activation of MR in non-epithelial tissues, including cardiomyocytes, smooth muscle cells, fibroblasts and macrophages in heart tissue [28,29], and glomerular podocytes [30], monocytes [31] and mesangial cells [32] in the kidney tissue, targets expression of genes that are involved in tissue repair and results in excess inflammation and fibrosis [22,33]. Activation of GRs modulates transcription of responsive genes related to energy homeostasis, response to stress and control of inflammation [25]. While only cortisol binds to the GR, the MR has multiple ligands with high affinity, including both cortisol and aldosterone [22,25]. Despite the fact that cortisol reaches up to 1000-fold higher concentrations in several tissues, aldosterone is the primary physiological MR ligand in humans [34]. Pre-receptor conversion of cortisol by 11-beta-hydroxysteroid-dehydrogenase-2 (11βHSD2) to inactive cortisone is a key mechanism for maintaining MR selectivity in target tissues and regulating distinct MR functions in the heart and kidneys [33,35]. Differentiations in the concentrations of 11βHSD2 in each tissue represent the major modulator of this process, with the enzyme being abundantly expressed in distal tubular epithelial cells but not in cardiomyocytes, podocytes and macrophages, where cortisol is the primary physiological ligand of MRs [33].
During the previous decade, the previously called nongenomic effects of aldosterone were elucidated. The genomic response is the process that includes all the classical steps of cell membrane diffusion of aldosterone: binding to the MR in the cytoplasm, translocation to the nucleus and activation of gene transcription [36,37]. This process results in an increase of ENaC concentration in epithelial cells (i.e., distal tubule, colon) and can be evidenced 30–60 min post aldosterone release/administration [38]. In addition to genomic actions, rapid effects of aldosterone have been described that cannot be explained by the traditional pathway, nor be blocked by inhibition of the transcriptional process molecules such as actinomycin D and aldosterone blockers, now considered to be mediated by MRs [39]. These rapid effects are associated with enhanced activity of the Na+-K+-2Cl cotransporter and Na+-K+-ATPase in the heart, and of the Na+-H+ antiporter, the ENaC and Na+-K+-ATPase in the kidney, and are connected to subcellular trafficking [39].
In addition to the above, over the past years the crucial role of coregulatory molecules in mediating the genomic response to nuclear receptor activation has emerged [36]. Accumulated evidence suggests that upon ligand binding, the transcription of effector proteins is modulated via recruitment of coactivator or corepressor proteins according to distinct MR conformations induced by binding of different agonist ligands [25,36]. Ligand-selective peptides acting as potent antagonists of the MR-mediated transcription process were identified about a decade ago [36].

2.2. Aldosterone Breakthrough and Potential Mechanisms of Aldosterone-Induced Diabetic Kidney Disease and Cardiac Damage

In 10–53% of patients that initiate an ACEi/ARB, plasma aldosterone levels tend to rise again 6–12 months later, leaving them exposed to the deleterious proinflammatory and profibrotic effects of aldosterone in the kidneys, heart and vessels [26,40,41]. It has been speculated that this phenomenon of “aldosterone breakthrough” may represent a major cause of accelerated GFR decline in patients with type 1 diabetes and DKD and a poorer antiproteinuric response despite use of a single RAS blockade [33]. Inappropriate activation of MRs by aldosterone in podocytes, monocytes and mesangial cells in the kidney induces monocyte and macrophage infiltration [42] and promotion of glomerulosclerosis and interstitial fibrosis [43]. In the heart, overactivation of MRs promotes cardiac fibrosis, increased collagen synthesis and cardiac remodeling and hypertrophy [38,44,45]. An additional negative inotropic effect of aldosterone, counteracting the positive inotropic effect of angiotensin-II, has been also described [46].
Moreover, in the experimental model of streptozotocin-induced diabetes, a classical model of type 1 DM, overexpression of mRNA of MR, NADPH oxidase and collagen I/IV was described, resulting in collagen deposition in glomerular and tubulointerstitial areas in the kidney [47]. Local production of aldosterone in mesangial cells induced by angiotensin-II, high glucose and LDL has been also associated with the pathogenesis of DKD through MR activation [48]. Administration of spironolactone was shown to block MR overexpression [47] and reduce collagen deposition in streptozotocin-induced diabetic rats [49]. Similarly, administration of spironolactone, eplerenone and finerenone in classical experimental models of DKD due to type 2 DM led to amelioration of glomerulosclerosis and macrophage infiltration [50], prevention of podocyte injury [51] and reduction of proteinuria and NGAL expression [52].
The crucial role of GR in progression of DKD has recently emerged. In an experimental model of streptozotocin-induced diabetes in podocyte GR knockout mice, the loss of podocyte GR resulted in enhanced Wnt signaling, higher expression of transforming growth factor-β and β-catenin and disturbed fatty acid metabolism, accompanied by histological evidence of worsened fibrosis, increased collagen deposition, as well as mesenchymal transition of the glomerular endothelium and glomerulomegaly [53]. Similarly, the loss of the endothelial GR has been shown to induce upregulation of Wnt/β-catenin signaling and to promote angiogenesis and mesenchymal transition in tubular epithelial cells in diabetic experimental models [54,55]. The above findings suggest the presence of a podocyte–endothelial cell crosstalk that is regulated by GR; this probably represents an additional mechanism in the development of diabetic nephropathy, on which more light should be shed in the future.

3. Clinical Studies of Mineralocorticoid Receptor Antagonism in Diabetic Kidney Disease

After considerable preclinical data yielding information on the pivotal role of MR overactivation in the progression of kidney and cardiac injury were available, several clinical studies embarked to investigate the potential benefits from MR blockade in patients with diabetic and non-diabetic kidney disease.
During the previous two decades, the non-selective MRA spironolactone and the selective eplerenone were extensively studied mainly for their anti-albuminuric effects. These older classical steroidal MRAs function as active competitors to aldosterone for binding to the MR ligand-binding pocket, destabilizing the active conformation of the receptor [56]. They prevent recruitment of transcriptional coactivators in the presence of aldosterone, but in the absence of aldosterone they exhibit partially agonistic coactivator recruitment properties [22]. They both lack tissue and ligand specificity, while spironolactone additionally lacks receptor specificity [56]. Canrenone, an active metabolite of spironolactone, was expected to have fewer side effects than spironolactone by preventing the formation of intermediate products with antiandrogenic and progestational actions [57]. It was approved for clinical use in Europe, however, its use was limited by significant hyperkalemia associated with lack of receptor-specific selectivity, similar to a first-generation MRA [58].
Finerenone is a third-generation nonsteroidal MRA that in vitro inhibits binding of coregulatory molecules independent of the presence or absence of aldosterone; it is characterized by tissue and ligand specificity and equal distribution between heart and kidney [26]. Equinatriuretic to eplerenone doses of finerenone demonstrate stronger anti-inflammatory and anti-fibrotic effects [59]. Esaxerenone is an equally potent and selective nonsteroidal MRA also exhibiting ligand-specific binding properties [60] and a dose-dependent BP-lowering effect that is at least equivalent to eplerenone, according to the Esaxerenone and Eplerenone in Patients With Essential Hypertension (ESAX-HTN Study) [61]. Apararenone is another compound belonging in the nonsteroidal family, also exhibiting highly selective MRA activity [62].

3.1. Effect of Mineralocorticoid Receptor Antagonists in Albuminuria/Proteinuria and Kidney-Related Outcomes

3.1.1. Spironolactone

In 2003, Sato et al. [63] were the first to study the clinical effect of aldosterone breakthrough in diabetic populations (Table 1). They followed 45 patients with type 2 DM, UACR 30–300 mg/g and creatinine clearance >60 mL/min for 40 weeks and observed a rise in aldosterone levels in 40% of them, despite treatment with trandolapril. In 15 of these 18 patients, the antiproteinuric effect of ACEi had declined to baseline; among them 13 patients were assigned to additionally receive spironolactone 25 mg for 24 weeks, resulting in a significant decrease in albuminuria and left ventricular mass index. In 2006, Chrysostomou et al. [64] evaluated 41 patients with overt proteinuria > 1.5 g/day and showed that the addition of spironolactone alone to a background treatment with ramipril produced a higher proteinuria reduction than addition of irbesartan and an equal reduction to the addition of both spironolactone and irbesartan.
Subsequently, Rossing et al. randomly assigned 21 type 2 DM patients with urinary albumin excretion (UAE) > 300 mg/24 h to receive spironolactone 25 mg or placebo added to a maximally up-titrated ACEi or ARB for 8 weeks in a cross-over design study [65]. The authors observed a significant reduction in albuminuria by −33% for spironolactone, as well as higher reductions in ambulatory 24 h SBP and DBP (−6 mmHg and −4 mmHg) compared to placebo (p < 0.001 for both comparisons). A non-significant and reversible eGFR decline by −3 mL/min/1.73 m2 was also observed. In subsequent trials from the same center, similar effects on UAE, 24 h BP and eGFR were reported with addition of spironolactone in 20 type 1 DM patients with UAE > 300 mg/24 h [66], as well as in 20 type 1 and type 2 DM patients with UAE > 2500 mg/24 h [67]. In a study by van der Meirecker et al. [68], 59 type 2 DM patients with UAE > 300 mg/day or UACR > 20 mg/g were evaluated for 1 year, to examine effects of spironolactone (25–50 mg) compared to placebo on top of an ACEi/ARB on proteinuria, BP and renal function. A significant decrease in albuminuria by −41% and in BP by −7/−3 mmHg was noted with spironolactone but not with placebo. In the spironolactone group, the reduction in eGFR was largest during the first 3 months after initiation of treatment but subsided during further follow-up. The slope of eGFR decline was similar between the spironolactone and placebo groups 3 months after allocation to study medication.
In perhaps one of the most detailed studies with UACR as the primary endpoint, Mehdi et al. [70] randomly allocated 81 patients with type 1 or type 2 DM and UACR > 300 mg/g, already under maximum dose of lisinopril (80 mg/day) to receive placebo, losartan (100 mg daily) or spironolactone (25 mg daily) for 48 weeks. They observed a decrease in UACR by −34.0% for spironolactone and by −16.8% for losartan. No significant between-group differences were noted in changes of creatinine clearance (−13.1% for spironolactone, −16.8% for losartan and −16% for placebo, p = 0.8). Several subsequent studies confirmed the additional anti-albuminuric effect of spironolactone when administered on top of an ACEi/ARB in diabetic patients with moderately or severely increased albuminuria [69,71,72,74,75,76] (Table 1).

3.1.2. Eplerenone

In a large study by Epstein et al. [77] in 2006, 268 patients with type 2 DM and UACR ≥ 50 mg/g were randomly assigned to receive placebo, eplerenone 50 mg or eplerenone 100 mg on top of enalapril 20 mg for 12 weeks (Table 2). A significant reduction of UACR by −41% and −48.4% was noted in the two eplerenone groups, respectively, but only by −7.4% in the placebo group (p < 0.001 vs. placebo for both comparisons). Systolic and diastolic BP decreased significantly in all three treatment groups, without between-group significant differences being observed. Overall, these results suggested that the anti-albuminuric effect of MR blockade was independent of BP reduction. The incidence of sustained (>5.5 mmol/L on two consecutive measurements) and severe (≥6.0 mmol/L at any timepoint) hyperkalemia did not significantly differ among the three treatment arms. A significant reduction in UACR by −70% was shown with an eplerenone/ramipril combination compared to ramipril monotherapy (−37%) in a recent smaller single-blind study in 75 type 2 DM patients with UACR 30–300 mg/g (p < 0.0001) [78]. In patients with eGFR < 60 mL/min/1.73 m2, a higher incidence of sustained hyperkalemia (> 5.5 mmol/L on 2 consecutive occasions) was noted in the eplerenone/ramipril combination group.
In a pre-specified secondary analysis of the Mineralocorticoid Receptor Antagonist in Type 2 Diabetes (MIRAD) trial by Brandt-Jacobsen et al. [79], 140 patients with type 2 DM and high risk of CVD (baseline UACR 17 mg/g, 12% of patients had an eGFR < 60 mL/min/1.73 m2), that were randomly allocated to either a high dose of eplerenone (100–200 mg) or placebo on top of background antihypertensive treatment for 26 weeks, were evaluated for UACR reduction, incidence of hyperkalemia and kidney-related adverse events. A decrease of UACR by −34% was observed for eplerenone compared to placebo (p = 0.005), an effect that was consistent across subgroups defined by the presence of UACR ≥ 30 mg/g (p-interaction = 0.441) and treatment with a RAS blocker. No significant differences were noted in the incidence of mild hyperkalemia (≥5.5 mmol/L) (p = 0.276); initiation of eplerenone, however, was associated with an increase in serum potassium by 0.26 mmol/L and a decrease in eGFR of −3.5 mL/min/1.73 m2.

3.1.3. Finerenone

The Mineralocorticoid Receptor Antagonist Tolerability Study–Diabetic Nephropathy (ARTS-DN) Study, published in 2015 by Bakris et al. [80], was the first placebo-controlled randomized clinical trial to investigate the safety and efficacy of different oral doses of finerenone compared to placebo, in 821 patients with type 2 DM and high or very high albuminuria on top of an RAS blocker (Table 3). After 90 days of treatment, dose-dependent reductions in placebo-corrected mean ratios of UACR relative to baseline were evidenced for finerenone (0.79, 0.76, 0.67 and 0.62 for the finerenone 7.5, 10, 15, 20 mg/day dose groups respectively); the risk of hyperkalemia leading to study discontinuation was 2.1%, 0%, 3.2% and 1.7%, respectively. In ARTS-DN Japan [81], in a subsequent similar study in 96 patients with type 2 DM and DKD, Katayama et al. [54] reported a numerical reduction of UACR at day 90 relative to baseline for each finerenone treatment group compared with placebo. Small mean increases were observed in serum potassium; no patient experienced an increase to levels ≥ 5.6 mmol/L or > 6 mmol/L throughout the study.
The clinical trials discussed above were focused on changes of albuminuria and were often not adequately powered to detect changes in the slope of eGFR or progression of CKD and CVD. Despite the fact that changes in albuminuria have proven to be strongly associated with a lower hazard for renal clinical endpoints (ESKD, fall of eGFR < 15 mL/min/1.73 m2) and lower risk of death [82,83], these represent an intermediate marker of kidney disease. Thus, from an efficacy point of view, the only currently available data on the impact of MRAs on long-term cardiorenal protection originate from the two recently published large-scale phase 3 randomized clinical trials of finerenone, the FIDELIO-DKD (Finerenone in reducing kidney failure and disease progression in Diabetic Kidney Disease) [23] and the FIGARO-DKD (Finerenone in reducing cardiovascular mortality and morbidity in Diabetic Kidney Disease) [24].
In the FIDELIO-DKD study, 5734 patients with type 2 DM and (a) UACR 300–5000 mg/g and eGFR 25 to < 75 mL/min/1.73 m2 or (b) UACR 30 to < 300 mg/g, eGFR 25 to < 60 mL/min/1.73 m2 and diabetic retinopathy, were randomly assigned to finerenone 10–20 mg or placebo on top of a maximum tolerated dose of RAS blocker [23]. During a median of 2.6 years, the primary outcome event, a composite of kidney failure (ESKD or eGFR < 15 mL/min/1.73 m2), sustained (≥4 weeks) eGFR decrease of ≥40% from baseline or death from renal causes, occurred in 17.8% of patients in the finerenone and 21.1% of patients in the placebo group (HR 0.82; 95%CI 0.73–0.93). Notably, finerenone was associated with an even larger reduction in the hazard of the main secondary renal outcome, a composite of kidney failure, sustained eGFR decrease of ≥57% from baseline or renal death (HR 0.76; 95% CI 0.65–0.90). Mean increases of serum potassium of about 0.23 mmol/L were observed with finerenone, while slightly higher rates of hyperkalemia episodes leading to drug discontinuation were reported with finerenone compared to placebo (2.3% vs. 0.9%). In addition to the above, lower rates of the key secondary cardiovascular outcome, a composite of time to first occurrence of cardiovascular death, nonfatal MI, nonfatal stroke or hospitalization for heart failure (HHF), were observed for patients receiving finerenone (13.0%) than in those receiving placebo (14.8%) (HR 0.86; 95%CI 0.75–0.99) [10].
In the FIGARO-DKD study, 7437 patients with type 2 DM and (a) UACR ≥ 30 to < 300 mg/g and eGFR ≥ 25–90 mL/min/1.73 m2 or (b) UACR ≥ 300 to ≤ 5000 mg/g and eGFR ≥ 60 mL/min/1.73 m2, were randomly assigned to receive finerenone 10–20 mg or placebo on top of a maximum tolerated dose of RAS blocker [24]. During a median follow-up of 3.4 years, the primary cardiovascular outcome event, a composite of cardiovascular death, nonfatal MI, nonfatal stroke or HHF, occurred in 12.4% of patients in the finerenone and 14.2% of patients in the placebo group (HR 0.87; 95%CI 0.76–0.98). Of importance, among the individual components of the primary outcome the greatest benefit was observed in reduction of risk for HHF (HR 0.71; 95%CI 0.56–0.90). Finerenone was also associated with a marginally but not statistically significant lower risk for the key secondary renal outcome, a composite of kidney failure, sustained eGFR decrease of ≥40% from baseline or renal death (HR 0.87; 95%CI 0.76–1.01). With regards to different components of the secondary outcome, significantly lower rates of progression to ESKD were observed with finerenone (0.9%) than with placebo (1.3%) (HR 0.64; 95%CI 0.41–0.995). The frequency of hyperkalemia episodes leading to drug discontinuation was even lower than in the FIDELIO-DKD study (1.2% vs. 0.4%, respectively).
The FIDELITY analysis, published at the end of 2021, was a pre-specified pooled efficacy and safety analysis of both the FIDELIO-DKD and FIGARO-DKD studies that together enrolled 13,171 patients, with a mean eGFR of 57.6 mL/min/1.73 m2 and a UACR of 515 mg/g [84]. According to the full analysis set, finerenone was associated with a significant reduction of 14% in the risk for the composite cardiovascular outcome of time to cardiovascular death, nonfatal MI, nonfatal stroke or HHF (HR 0.86; 95%CI 0.78–0.95), and a significant reduction of 23% in the risk for the composite kidney outcome of time to first onset of kidney failure, sustained eGFR decrease ≥ 57% from baseline or renal death (HR 0.77; 95%CI 0.67–0.88). The cardiovascular benefit was primarily driven by a lower incidence of HHF (HR 0.78; 95%CI 0.66–0.92). The renal benefit was attributed to a 30% reduction in the risk of sustained eGFR decrease ≥ 57% (HR 0.70; 95%CI 0.60–0.83), as well as to a 20% reduction in the risk for ESKD (HR 0.80; 95%CI 0.64–0.99). It should be further emphasized that these results represent the first available evidence on beneficial effects of an MRA agent on hard renal and cardiovascular outcomes. Hyperkalemia leading to drug discontinuation was more frequently reported in patients receiving finerenone (1.7%) than in placebo (0.6%). No fatal hyperkalemia-related adverse event was reported.
The cardiovascular benefit obtained by finerenone administration in patients with DKD seems to extend beyond decreases in HHF. According to a secondary analysis of the FIDELIO-DKD study by Filippatos et al. [85], a significant reduction of 29% was evidenced in the risk of new-onset atrial fibrillation (AF) (HR 0.71; 95%CI 0.53–0.94), that was partly attributed by the authors to inhibition of aldosterone-associated atrial structural and electrical remodeling, to which patients with CKD and type 2 DM are highly predisposed. The importance of this finding is underlined by the results of a stratified Cox proportional hazards model demonstrating an increase in the risk of fatal or nonfatal stroke after a new-onset episode of AF (HR 7.13; 95%CI 4.01–12.70). On the contrary, in pre-specified exploratory analyses of individual components of the composite cardiovascular outcome of the FIDELIO-DKD study, no significant effect on the incidence of nonfatal stroke was observed for finerenone (HR 1.03; 95%CI 0.76–1.38) [86].
The impact of the use of SGLT2is, agents with proven renoprotective effects in patients with CKD [87,88], on the beneficial effects of finerenone observed in the FIDELIO-DKD was recently explored in a pre-specified exploratory sub-analysis of the trial [89]. Overall, 4.4% of patients in the finerenone group and 4.8% in the placebo group were treated with a SGLT2i at baseline. The effect of finerenone compared with placebo on the primary composite renal outcome seemed to be consistent in the subgroups of patients receiving (HR 1.38; 95%CI 0.61–3.10) and those not receiving a SGLT2i (HR 0.82, 95%CI 0.72–0.92) (p-interaction = 0.21). Subgroup analysis for the key secondary renal and cardiovascular outcomes yielded similar results (p-interaction = 0.54 and 0.46, respectively). In addition, numerically fewer episodes of hyperkalemia-related adverse events were observed for patients receiving a SGLT2i at baseline (8.1%) compared to those who did not (18.7%), a phenomenon that was attributed to enhanced potassium excretion due to natriuresis and osmotic diuresis. Overall, these results suggest that MRAs and SGLT2is have complementary actions and that a combined treatment with both agents may provide additional effects above and beyond those associated with each individual agent.

3.1.4. Canrenone

To date there are only two studies comparing the effect of canrenone to an active comparator (hydrochlorothiazide) on albuminuria- and kidney-related outcomes in diabetic patients (Table 4). In the study by Fogari et al. [57], among 120 patients with type 2 DM and UACR 60–300 mg/g, UAE decreased by −45.3% in the canrenone group and by −20.3% in the hydrochlorothiazide group (p < 0.01). No changes in serum levels of creatinine and potassium were noted. In the second study, from the same group [90], authors reported a slight decrease in eGFR with hydrochlorothiazide but not with canrenone, along with a neutral effect on potassium levels.

3.1.5. Esaxerenone

Currently, there are results from three studies evaluating the effects of esaxerenone in patients with DKD (Table 4). In the first one, a phase 2b study in 365 patients with type 2 DM with UACR ≥ 45 to < 300 mg/g and eGFR ≥ 30 mL/min/1.73 m2, esaxerenone showed a significant reduction of albuminuria that was dose-dependent (38% for 1.25 mg, 50% for 2.5 mg and 56% for 5 mg/day) compared with placebo (7%) after 12 weeks of treatment (all p < 0.001) [91] (Table 4). In the phase 3 Esaxerenone in Patients with Type 2 Diabetes and Microalbuminuria (ESAX-DN) study, 455 patients with type 2 DM and moderately increased albuminuria were randomized to receive esaxerenone 1.25–2.5 mg/day or placebo on top of an RAS blocker for 52 weeks [92]. Remission of albuminuria was achieved in 22% of patients in the esaxerenone group compared to 4% in the placebo group (p < 0.001), with a mean reduction of UACR by −58% for esaxerenone vs. +8% for placebo at the end of treatment (p < 0.001). A significantly higher proportion of patients in the esaxerenone group had a serum potassium of either ≥6.0 mmol/L or ≥5.5 mmol/L at two consecutive occasions compared to placebo (9% vs. 2%, p = 0.002), while hyperkalemia led to treatment discontinuation in 4% vs. 1%, respectively. A significantly higher eGFR decline from baseline was noted for esaxerenone than in placebo (−11% vs. −1%). In an open-label 28-week phase 3 study in 56 patients with type 2 DM, UACR ≥ 300 mg/g and eGFR ≥ 30 mL/min/1.73 m2, administration of esaxerenone led to a UACR reduction of −54.6% from baseline and a mean eGFR decline of −8.3 mL/min/1.73 m2; the proportion of patients with serum potassium of ≥6.0 mmol/L or ≥5.5 mmol/L at two occasions was 5.4% [93].

3.1.6. Apararenone

The only available data on apararenone originate from a placebo-controlled phase 2 dose-response study with an open-label extension period in 293 Japanese patients with type 2 DM and UACR 50–300 mg/g [62] (Table 4). After 24 weeks of treatment, a significant reduction in UACR ranging between 46.5–62.9% was observed with apararenone, but not with placebo (p < 0.001 for all comparisons vs. placebo). The anti-albuminuric effect persisted in the 52-week extension period. A mean increase in serum potassium by 0.2–0.3 mmol/L was evidenced with apararenone. The main pitfall of this study is that only ~65% of participants in each treatment group received an RAS blocker at randomization.
Table 4. Main studies on the effects of canrenone, esaxerenone and apararenone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
Table 4. Main studies on the effects of canrenone, esaxerenone and apararenone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
StudyStudy
Design
Follow
UP
Nr of
Patients
Patient
Characteristics
Active
Treatment
ComparatorBack-
Ground Treatment
Main Results
Fogari
et al.,
2014 [57]
Open-label,
parallel-
group RCT
24 weeks120T2DM and UACR 60–300 mg/gCanrenoneHydrochlorothiazideValsartan↓ UACR −45.3% for canrenone; −20.3% for hydrochlorothiazide (p < 0.01)
Derosa
et al., 2018 [90]
Double-blind, parallel-group RCT12 months182T2DM and hypertensionCanrenoneHydrochlorothiazideARBSignificant ↓ K only for hydrochlorothiazide (p < 0.05); neutral effect for canrenone
Significant ↓ eGFR for hydrochlorothiazide (p < 0.01)
Significant ↑ eGFR for canrenone (p < 0.05)
Ito et al., 2019 [91]Double-blind, parallel-group RCT12 weeks365T2DM, UACR 45–300 mg/g, eGFR ≥ 30 mL/min/1.73 m2EsaxerenonePlaceboACEi or ARB↓ UACR −38% for esaxerenone 1.25 mg;
−50% for esaxerenone 2.5 mg; −56% for esaxerenone 5 mg; −7% for placebo (p < 0.001)
Remission of albuminuria: 21% for esaxerenone groups 2.5 and 5.0 mg; 3% for placebo (p < 0.05 for both comparisons)
HyperK leading to drug discontinuation: 3% for esaxerenone 1.25 and 2.5 mg; 10% esaxerenone 10 mg; 1% for placebo
Ito et al., 2020 (ESAX-DN) [92]Double-blind, parallel-group RCT52 weeks455T2DM, UACR 45–300 mg/g, eGFR ≥ 30 mL/min/1.73 m2EsaxerenonePlaceboACEi or ARB↓ UACR −58% for esaxerenone; +8% for placebo (p < 0.001); eGFR −11% for esaxerenone; −1% for placebo
Remission of albuminuria: 22% in esaxerenone; 4% in placebo (p < 0.001)
Time to 1st transition to overt proteinuria:
HR 0.23; 95%CI 0.11–0.48 for esaxerenone
HyperK episodes (>6.0 mmol/L or ≥5.5 mmol/L at two consecutive occasions): 9% esaxerenone; 2% placebo (p = 0.002)
HyperK leading to drug discontinuation: 4% for esaxerenone; 1% for placebo
Ito et al., 2021 [93]Open-label, single-arm study28 weeks56T2DM, UACR ≥ 300 mg/g, eGFR ≥ 30 mL/min/1.73 m2Esaxerenone-ACEi or ARB↓ UACR −54.6% (p < 0.001); eGFR −8.3 mL/min /1.73 m2 for esaxerenone
HyperK episodes (>6.0 mmol/L or ≥5.5 mmol/L at two consecutive occasions): 5.4% for esaxerenone
Wada et al., 2021 [62]Double-blind, parallel-group RCT
with open-label extension
24 weeks and
28 weeks
293T2DM,
UACR
 50–300 mg/g
ApararenonePlaceboACEi or ARB↓ UACR at 23 weeks −62.9% apararenone 2.5 mg; −50.8% apararenone 5 mg; −46.5% apararenone 10 mg; +113.7% placebo (p < 0.001 vs. placebo for all comparisons)
% change in eGFR at 52 weeks: −5.3% (−22.0, +10.5) apararenone 2.5 mg; −10.2% (−34.5, +14.6) apararenone 5 mg; −10.80% (−36.8, +19.1) apararenone 10 mg
↑ K at 52 weeks: +0.14 mmol/L (0.006–0.22) apararenone 2.5 mg; +0.18 mmol/L (0.1–0.26) apararenone 5 mg; +0.25 mmol/L (0.16–0.33) apararenone 10 mg
↓, decrease; ↑, increase; ACEi, angiotensin converting enzyme inhibitor; ARB, angiotensin receptor blocker; CI, confidence interval; eGFR, estimated glomerular filtration rate; hyperK, hyperkalemia; HR, hazard ratio; K, potassium; RCT, randomized controlled trial; T2DM, type 2 diabetes mellitus; UACR, urinary albumin creatinine ratio.

3.2. Meta-Analyses of Randomized Clinical Studies with Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease

In a meta-analysis pooling available results from studies published up to 2019, including also the first two finerenone studies (31 studies, 2767 participants) [14], the addition of an MRA (spironolactone, eplerenone, canrenone, finerenone) alone or on top of an RAS blocker led to a significant reduction of UACR by −24.55%, of 24 h UAE by −32.47% and of urine-protein–creatinine ratio by −53.93%. An eGFR decrease of −2.82 mL/min/1.73 m2 in parallel to an increase in serum potassium by 0.19 mmol/L was noted compared to placebo.
In a recently published meta-analysis of four randomized clinical trials evaluating the efficacy and safety of finerenone in 13,945 patients with DKD [94], the addition of finerenone was associated with a decrease in UACR by −0.30 (95%CI −0.33 to −0.27) and a lower risk of experiencing an eGFR reduction ≥40% (RR 0.85, 95%CI 0.78 to 0.93) compared to placebo. The incidence of hyperkalemia was higher in the finerenone than in the placebo group (RR 2.03, 95%CI 1.83 to 2.26).

4. Future Directions and Perspectives

Overall, the results of the aforementioned randomized controlled provide strong evidence of the beneficial effects of MRAs towards reduction of albuminuria and protection against CKD and CVD progression in patients with DKD. The landscape of treatment of this disease is expected to change in the near future; the use of finerenone will probably be incorporated in forthcoming guidelines and consensus statements of major organizations as first-line treatment for DKD along with SGLT2is to maximize cardio- and nephroprotection. Further controlled trials with cardiovascular and renal outcomes are necessary to shed more light on the presence or absence of similar beneficial effects of the other two nonsteroidal agents, esaxerenone and apararenone.

5. Conclusions

Although the use of ACEis/ARBs has been recommended for decades in patients with DKD, and novel, nephroprotective and cardioprotective treatment options such as SGLT2is have been available for some years, important residual risk remains with regards to the rate of CKD and CVD progression. Aldosterone breakthrough represents the main caveat of monotherapy with an ACEi/ARB, leaving the patient exposed to the deleterious genomic and non-genomic effects of aldosterone, including heart and kidney tissue inflammation and fibrosis by MR overactivation. MRA treatment has demonstrated beneficial effects in BP control and reduction of cardiovascular risk in various patient populations. In patients with DKD, first- and second-generation MRAs have been shown to reduce UAE, a strong surrogate marker of renal function decline and progression to ESKD. Despite these promising effects, the absence of hard-outcome data and fear of hyperkalemia has not enabled their extensive use in patients with CKD. Novel third-generation nonsteroidal MRAs, such as finerenone, exhibit greater selectivity than spironolactone and better affinity than eplerenone for the MR, as well as a more balanced distribution between the myocardium and renal tissue in preclinical studies. A pre-specified analysis of a combined population of >13,000 patients with DKD from two major randomized clinical trials of finerenone with hard cardiorenal outcomes demonstrated a significant reduction of −20% in the risk for ESKD and by −22% in the risk for HHF [84]. Overall, nonsteroidal MRAs exhibit an improved benefit–risk ratio than steroidal MRAs, exceeding the risks of combined treatment with an ACEI/ARB. Therefore, the addition of finerenone on top of the standard of care in patients remaining at high risk of progression of their renal and CV disease is expected to confer an additional cardiorenal benefit. Finerenone should receive a clear recommendation in forthcoming guidelines by delaying progression to ESKD and mitigating CV morbidity in patients with DKD.

Author Contributions

Conceptualization, P.A.S.; bibliography research, M.-E.A. and M.P.T.; writing—original draft preparation, M.-E.A.; writing—review and editing, M.-E.A., M.P.T. and P.A.S.; supervision, P.A.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Saeedi, P.; Petersohn, I.; Salpea, P.; Malanda, B.; Karuranga, S.; Unwin, N.; Colagiuri, S.; Guariguata, L.; Motala, A.A.; Ogurtsova, K.; et al. Global and Regional Diabetes Prevalence Estimates for 2019 and Projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th Edition. Diabetes Res. Clin. Pract. 2019, 157, 107843. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Tuttle, K.R.; Bakris, G.L.; Bilous, R.W.; Chiang, J.L.; de Boer, I.H.; Goldstein-Fuchs, J.; Hirsch, I.B.; Kalantar-Zadeh, K.; Narva, A.S.; Navaneethan, S.D.; et al. Diabetic Kidney Disease: A Report from an ADA Consensus Conference. Diabetes Care 2014, 37, 2864–2883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. United States Renal Data System. 2021 USRDS Annual Data Report: Epidemiology of Kidney Disease in the United States; National Institutes of Health; National Institute of Diabetes and Digestive and Kidney Diseases: Bethesda, MD, USA, 2021. [Google Scholar]
  4. Afkarian, M.; Sachs, M.C.; Kestenbaum, B.; Hirsch, I.B.; Tuttle, K.R.; Himmelfarb, J.; de Boer, I.H. Kidney Disease and Increased Mortality Risk in Type 2 Diabetes. J. Am. Soc. Nephrol. 2013, 24, 302–308. [Google Scholar] [CrossRef] [PubMed]
  5. Brenner, B.M.; Cooper, M.E.; de Zeeuw, D.; Keane, W.F.; Mitch, W.E.; Parving, H.H.; Remuzzi, G.; Snapinn, S.M.; Zhang, Z.; Shahinfar, S. Effects of Losartan on Renal and Cardiovascular Outcomes in Patients with Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2001, 345, 861–869. [Google Scholar] [CrossRef] [Green Version]
  6. Lewis, E.J.; Hunsicker, L.G.; Clarke, W.R.; Berl, T.; Pohl, M.A.; Lewis, J.B.; Ritz, E.; Atkins, R.C.; Rohde, R.; Raz, I. Renoprotective Effect of the Angiotensin-Receptor Antagonist Irbesartan in Patients with Nephropathy Due to Type 2 Diabetes. N. Engl. J. Med. 2001, 345, 851–860. [Google Scholar] [CrossRef] [Green Version]
  7. Kidney Disease: Improving Global Outcomes (KDIGO) Diabetes Work Group. KDIGO 2020 Clinical Practice Guideline for Diabetes Management in Chronic Kidney Disease. Kidney Int. 2020, 98, S1–S115. [Google Scholar] [CrossRef]
  8. Sarafidis, P.A.; Alexandrou, M.E.; Ruilope, L.M. A Review of Chemical Therapies for Treating Diabetic Hypertension. Expert Opin. Pharmacother. 2017, 18, 909–923. [Google Scholar] [CrossRef]
  9. Parving, H.H.; Brenner, B.M.; McMurray, J.J.; de Zeeuw, D.; Haffner, S.M.; Solomon, S.D.; Chaturvedi, N.; Persson, F.; Desai, A.S.; Nicolaides, M.; et al. Cardiorenal End Points in a Trial of Aliskiren for Type 2 Diabetes. N. Engl. J. Med. 2012, 367, 2204–2213. [Google Scholar] [CrossRef] [Green Version]
  10. Fried, L.F.; Emanuele, N.; Zhang, J.H.; Brophy, M.; Conner, T.A.; Duckworth, W.; Leehey, D.J.; McCullough, P.A.; O’Connor, T.; Palevsky, P.M.; et al. Combined Angiotensin Inhibition for the Treatment of Diabetic Nephropathy. N. Engl. J. Med. 2013, 369, 1892–1903. [Google Scholar] [CrossRef] [Green Version]
  11. Lim, P.O.; Young, W.F.; MacDonald, T.M. A Review of the Medical Treatment of Primary Aldosteronism. J. Hypertens. 2001, 19, 353–361. [Google Scholar] [CrossRef]
  12. Williams, B.; MacDonald, T.M.; Morant, S.; Webb, D.J.; Sever, P.; McInnes, G.; Ford, I.; Cruickshank, J.K.; Caulfield, M.J.; Salsbury, J.; et al. Spironolactone versus Placebo, Bisoprolol, and Doxazosin to Determine the Optimal Treatment for Drug-Resistant Hypertension (PATHWAY-2): A Randomised, Double-Blind, Crossover Trial. Lancet 2015, 386, 2059–2068. [Google Scholar] [CrossRef] [Green Version]
  13. Oxlund, C.S.; Henriksen, J.E.; Tarnow, L.; Schousboe, K.; Gram, J.; Jacobsen, I.A. Low Dose Spironolactone Reduces Blood Pressure in Patients with Resistant Hypertension and Type 2 Diabetes Mellitus: A Double Blind Randomized Clinical Trial. J. Hypertens. 2013, 31, 2094–2102. [Google Scholar] [CrossRef] [PubMed]
  14. Alexandrou, M.-E.; Papagianni, A.; Tsapas, A.; Loutradis, C.; Boutou, A.; Piperidou, A.; Papadopoulou, D.; Ruilope, L.; Bakris, G.; Sarafidis, P. Effects of Mineralocorticoid Receptor Antagonists in Proteinuric Kidney Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Hypertens. 2019, 37, 2307–2324. [Google Scholar] [CrossRef] [PubMed]
  15. Pitt, B.; Zannad, F.; Remme, W.J.; Cody, R.; Castaigne, A.; Perez, A.; Palensky, J.; Wittes, J. The Effect of Spironolactone on Morbidity and Mortality in Patients with Severe Heart Failure. Randomized Aldactone Evaluation Study Investigators. N. Engl. J. Med. 1999, 341, 709–717. [Google Scholar] [CrossRef] [Green Version]
  16. Pitt, B.; Remme, W.; Zannad, F.; Neaton, J.; Martinez, F.; Roniker, B.; Bittman, R.; Hurley, S.; Kleiman, J.; Gatlin, M. Eplerenone, a Selective Aldosterone Blocker, in Patients with Left Ventricular Dysfunction after Myocardial Infarction. N. Engl. J. Med. 2003, 348, 1309–1321. [Google Scholar] [CrossRef]
  17. Zannad, F.; McMurray, J.J.V.; Krum, H.; van Veldhuisen, D.J.; Swedberg, K.; Shi, H.; Vincent, J.; Pocock, S.J.; Pitt, B. EMPHASIS-HF Study Group Eplerenone in Patients with Systolic Heart Failure and Mild Symptoms. N. Engl. J. Med. 2011, 364, 11–21. [Google Scholar] [CrossRef] [Green Version]
  18. Ponikowski, P.; Voors, A.A.; Anker, S.D.; Bueno, H.; Cleland, J.G.; Coats, A.J.; Falk, V.; Gonzalez-Juanatey, J.R.; Harjola, V.P.; Jankowska, E.A.; et al. 2016 ESC Guidelines for the Diagnosis and Treatment of Acute and Chronic Heart Failure: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure of the European Society of Cardiology (ESC). Developed with the Special Contribution of the Heart Failure Association (HFA) of the ESC. Eur. J. Heart Fail. 2016, 18, 891–975. [Google Scholar]
  19. Ferreira, J.P.; Abreu, P.; McMurray, J.J.V.; van Veldhuisen, D.J.; Swedberg, K.; Pocock, S.J.; Vincent, J.; Lins, K.; Rossignol, P.; Pitt, B.; et al. Renal Function Stratified Dose Comparisons of Eplerenone versus Placebo in the EMPHASIS-HF Trial. Eur. J. Heart Fail. 2019, 21, 345–351. [Google Scholar] [CrossRef] [Green Version]
  20. Trevisan, M.; de Deco, P.; Xu, H.; Evans, M.; Lindholm, B.; Bellocco, R.; Barany, P.; Jernberg, T.; Lund, L.H.; Carrero, J.J. Incidence, Predictors and Clinical Management of Hyperkalaemia in New Users of Mineralocorticoid Receptor Antagonists. Eur. J. Heart Fail. 2018, 20, 1217–1226. [Google Scholar] [CrossRef] [Green Version]
  21. Mullens, W.; Damman, K.; Testani, J.M.; Martens, P.; Mueller, C.; Lassus, J.; Tang, W.H.W.; Skouri, H.; Verbrugge, F.H.; Orso, F.; et al. Evaluation of Kidney Function throughout the Heart Failure Trajectory—A Position Statement from the Heart Failure Association of the European Society of Cardiology. Eur. J. Heart Fail. 2020, 22, 584–603. [Google Scholar] [CrossRef]
  22. Agarwal, R.; Kolkhof, P.; Bakris, G.; Bauersachs, J.; Haller, H.; Wada, T.; Zannad, F. Steroidal and Non-Steroidal Mineralocorticoid Receptor Antagonists in Cardiorenal Medicine. Eur. Heart J. 2021, 42, 152–161. [Google Scholar] [CrossRef] [PubMed]
  23. Bakris, G.L.; Agarwal, R.; Anker, S.D.; Pitt, B.; Ruilope, L.M.; Rossing, P.; Kolkhof, P.; Nowack, C.; Schloemer, P.; Joseph, A.; et al. Effect of Finerenone on Chronic Kidney Disease Outcomes in Type 2 Diabetes. N. Engl. J. Med. 2020, 383, 2219–2229. [Google Scholar] [CrossRef] [PubMed]
  24. Pitt, B.; Filippatos, G.; Agarwal, R.; Anker, S.D.; Bakris, G.L.; Rossing, P.; Joseph, A.; Kolkhof, P.; Nowack, C.; Schloemer, P.; et al. Cardiovascular Events with Finerenone in Kidney Disease and Type 2 Diabetes. N. Engl. J. Med. 2021, 385, 2252–2263. [Google Scholar] [CrossRef] [PubMed]
  25. Gomez-Sanchez, E.; Gomez-Sanchez, C.E. The Multifaceted Mineralocorticoid Receptor. Compr. Physiol. 2014, 4, 965–994. [Google Scholar] [PubMed] [Green Version]
  26. Sarafidis, P.A.; Memmos, E.; Alexandrou, M.-E.; Papagianni, A. Mineralocorticoid Receptor Antagonists for Nephroprotection: Current Evidence and Future Perspectives. Curr. Pharm. Des. 2018, 24, 5528–5536. [Google Scholar] [CrossRef]
  27. Shibata, S.; Rinehart, J.; Zhang, J.; Moeckel, G.; Castañeda-Bueno, M.; Stiegler, A.L.; Boggon, T.J.; Gamba, G.; Lifton, R.P. Mineralocorticoid Receptor Phosphorylation Regulates Ligand Binding and Renal Response to Volume Depletion and Hyperkalemia. Cell Metab. 2013, 18, 660–671. [Google Scholar] [CrossRef] [Green Version]
  28. Brown, N.J. Contribution of Aldosterone to Cardiovascular and Renal Inflammation and Fibrosis. Nat. Rev. Nephrol. 2013, 9, 459–469. [Google Scholar] [CrossRef]
  29. Lavall, D.; Selzer, C.; Schuster, P.; Lenski, M.; Adam, O.; Schäfers, H.-J.; Böhm, M.; Laufs, U. The Mineralocorticoid Receptor Promotes Fibrotic Remodeling in Atrial Fibrillation. J. Biol. Chem. 2014, 289, 6656–6668. [Google Scholar] [CrossRef] [Green Version]
  30. Shibata, S.; Nagase, M.; Yoshida, S.; Kawachi, H.; Fujita, T. Podocyte as the Target for Aldosterone: Roles of Oxidative Stress and Sgk1. Hypertension 2007, 49, 355–364. [Google Scholar] [CrossRef] [Green Version]
  31. Calò, L.A.; Zaghetto, F.; Pagnin, E.; Davis, P.A.; De Mozzi, P.; Sartorato, P.; Martire, G.; Fiore, C.; Armanini, D. Effect of Aldosterone and Glycyrrhetinic Acid on the Protein Expression of PAI-1 and P22(Phox) in Human Mononuclear Leukocytes. J. Clin. Endocrinol. Metab. 2004, 89, 1973–1976. [Google Scholar] [CrossRef]
  32. Terada, Y.; Kobayashi, T.; Kuwana, H.; Tanaka, H.; Inoshita, S.; Kuwahara, M.; Sasaki, S. Aldosterone Stimulates Proliferation of Mesangial Cells by Activating Mitogen-Activated Protein Kinase 1/2, Cyclin D1, and Cyclin A. J. Am. Soc. Nephrol. 2005, 16, 2296–2305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Barrera-Chimal, J.; Girerd, S.; Jaisser, F. Mineralocorticoid Receptor Antagonists and Kidney Diseases: Pathophysiological Basis. Kidney Int. 2019, 96, 302–319. [Google Scholar] [CrossRef] [PubMed]
  34. Bledsoe, R.K.; Madauss, K.P.; Holt, J.A.; Apolito, C.J.; Lambert, M.H.; Pearce, K.H.; Stanley, T.B.; Stewart, E.L.; Trump, R.P.; Willson, T.M.; et al. A Ligand-Mediated Hydrogen Bond Network Required for the Activation of the Mineralocorticoid Receptor. J. Biol. Chem. 2005, 280, 31283–31293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Seckl, J.R.; Walker, B.R. Minireview: 11beta-Hydroxysteroid Dehydrogenase Type 1- a Tissue-Specific Amplifier of Glucocorticoid Action. Endocrinology 2001, 142, 1371–1376. [Google Scholar] [CrossRef] [PubMed]
  36. Yang, J.; Chang, C.; Safi, R.; Morgan, J.; McDonnell, D.P.; Fuller, P.J.; Clyne, C.D.; Young, M.J. Identification of Ligand-Selective Peptide Antagonists of the Mineralocorticoid Receptor Using Phage Display. Mol. Endocrinol. 2011, 25, 32–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Viengchareun, S.; Le Menuet, D.; Martinerie, L.; Munier, M.; Pascual-Le Tallec, L.; Lombès, M. The Mineralocorticoid Receptor: Insights into Its Molecular and (Patho)Physiological Biology. Nucl. Recept. Signal. 2007, 5, e012. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Hermidorff, M.M.; de Assis, L.V.M.; Isoldi, M.C. Genomic and Rapid Effects of Aldosterone: What We Know and Do Not Know Thus Far. Heart Fail. Rev. 2017, 22, 65–89. [Google Scholar] [CrossRef]
  39. Ruhs, S.; Nolze, A.; Hübschmann, R.; Grossmann, C. 30 Years of the Mineralocorticoid Receptor: Nongenomic Effects via the Mineralocorticoid Receptor. J. Endocrinol. 2017, 234, T107–T124. [Google Scholar] [CrossRef] [Green Version]
  40. Bomback, A.S.; Klemmer, P.J. The Incidence and Implications of Aldosterone Breakthrough. Nat. Clin. Pract. Nephrol. 2007, 3, 486–492. [Google Scholar] [CrossRef]
  41. Sarafidis, P.A.; Ruilope, L.M. Cardiorenal Disease Development under Chronic Renin-Angiotensin-Aldosterone System Suppression. J. Renin Angiotensin Aldosterone Syst. 2012, 13, 217–219. [Google Scholar] [CrossRef]
  42. Rocha, R.; Rudolph, A.E.; Frierdich, G.E.; Nachowiak, D.A.; Kekec, B.K.; Blomme, E.A.G.; McMahon, E.G.; Delyani, J.A. Aldosterone Induces a Vascular Inflammatory Phenotype in the Rat Heart. Am. J. Physiol. Heart Circ. Physiol. 2002, 283, H1802–H1810. [Google Scholar] [CrossRef]
  43. Greene, E.L.; Kren, S.; Hostetter, T.H. Role of Aldosterone in the Remnant Kidney Model in the Rat. J. Clin. Investig. 1996, 98, 1063–1068. [Google Scholar] [CrossRef] [PubMed]
  44. Nagase, M.; Ayuzawa, N.; Kawarazaki, W.; Ishizawa, K.; Ueda, K.; Yoshida, S.; Fujita, T. Oxidative Stress Causes Mineralocorticoid Receptor Activation in Rat Cardiomyocytes: Role of Small GTPase Rac1. Hypertension 2012, 59, 500–506. [Google Scholar] [CrossRef] [Green Version]
  45. Köhler, E.; Bertschin, S.; Woodtli, T.; Resink, T.; Erne, P. Does Aldosterone-Induced Cardiac Fibrosis Involve Direct Effects on Cardiac Fibroblasts? J. Vasc. Res. 1996, 33, 315–326. [Google Scholar] [PubMed]
  46. Chai, W.; Garrelds, I.M.; de Vries, R.; Batenburg, W.W.; van Kats, J.P.; Danser, A.H.J. Nongenomic Effects of Aldosterone in the Human Heart: Interaction with Angiotensin II. Hypertension 2005, 46, 701–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Taira, M.; Toba, H.; Murakami, M.; Iga, I.; Serizawa, R.; Murata, S.; Kobara, M.; Nakata, T. Spironolactone Exhibits Direct Renoprotective Effects and Inhibits Renal Renin-Angiotensin-Aldosterone System in Diabetic Rats. Eur. J. Pharmacol. 2008, 589, 264–271. [Google Scholar] [CrossRef]
  48. Nishikawa, T.; Matsuzawa, Y.; Suematsu, S.; Saito, J.; Omura, M.; Kino, T. Effect of Atorvastatin on Aldosterone Production Induced by Glucose, LDL or Angiotensin II in Human Renal Mesangial Cells. Arzneimittelforschung 2010, 60, 445–451. [Google Scholar] [CrossRef]
  49. Fujisawa, G.; Okada, K.; Muto, S.; Fujita, N.; Itabashi, N.; Kusano, E.; Ishibashi, S. Spironolactone Prevents Early Renal Injury in Streptozotocin-Induced Diabetic Rats. Kidney Int. 2004, 66, 1493–1502. [Google Scholar] [CrossRef] [Green Version]
  50. Han, S.-Y.; Kim, C.-H.; Kim, H.-S.; Jee, Y.-H.; Song, H.-K.; Lee, M.-H.; Han, K.-H.; Kim, H.-K.; Kang, Y.-S.; Han, J.-Y.; et al. Spironolactone Prevents Diabetic Nephropathy through an Anti-Inflammatory Mechanism in Type 2 Diabetic Rats. J. Am. Soc. Nephrol. 2006, 17, 1362–1372. [Google Scholar] [CrossRef] [Green Version]
  51. Kang, Y.S.; Ko, G.J.; Lee, M.H.; Song, H.K.; Han, S.Y.; Han, K.H.; Kim, H.K.; Han, J.Y.; Cha, D.R. Effect of Eplerenone, Enalapril and Their Combination Treatment on Diabetic Nephropathy in Type II Diabetic Rats. Nephrol. Dial. Transplant. 2009, 24, 73–84. [Google Scholar] [CrossRef] [Green Version]
  52. Lachaux, M.; Barrera-Chimal, J.; Nicol, L.; Rémy-Jouet, I.; Renet, S.; Dumesnil, A.; Wecker, D.; Richard, V.; Kolkhof, P.; Jaisser, F.; et al. Short- and Long-Term Administration of the Non-Steroidal Mineralocorticoid Receptor Antagonist Finerenone Opposes Metabolic Syndrome-Related Cardio-Renal Dysfunction. Diabetes Obes. Metab. 2018, 20, 2399–2407. [Google Scholar] [CrossRef] [PubMed]
  53. Srivastava, S.P.; Zhou, H.; Setia, O.; Dardik, A.; Fernandez-Hernando, C.; Goodwin, J. Podocyte Glucocorticoid Receptors Are Essential for Glomerular Endothelial Cell Homeostasis in Diabetes Mellitus. J. Am. Heart Assoc. 2021, 10, e019437. [Google Scholar] [CrossRef] [PubMed]
  54. Srivastava, S.P.; Zhou, H.; Setia, O.; Liu, B.; Kanasaki, K.; Koya, D.; Dardik, A.; Fernandez-Hernando, C.; Goodwin, J. Loss of Endothelial Glucocorticoid Receptor Accelerates Diabetic Nephropathy. Nat. Commun. 2021, 12, 2368. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, B.; Zhou, H.; Zhang, T.; Gao, X.; Tao, B.; Xing, H.; Zhuang, Z.; Dardik, A.; Kyriakides, T.R.; Goodwin, J.E. Loss of Endothelial Glucocorticoid Receptor Promotes Angiogenesis via Upregulation of Wnt/β-Catenin Pathway. Angiogenesis 2021, 24, 631–645. [Google Scholar] [CrossRef] [PubMed]
  56. Rogerson, F.M.; Yao, Y.; Smith, B.J.; Fuller, P.J. Differences in the Determinants of Eplerenone, Spironolactone and Aldosterone Binding to the Mineralocorticoid Receptor. Clin. Exp. Pharmacol. Physiol. 2004, 31, 704–709. [Google Scholar] [CrossRef]
  57. Fogari, R.; Derosa, G.; Zoppi, A.; Lazzari, P.; D’Angelo, A.; Mugellini, A. Comparative Effect of Canrenone or Hydrochlorothiazide Addition to Valsartan/Amlodipine Combination on Urinary Albumin Excretion in Well-Controlled Type 2 Diabetic Hypertensive Patients with Microalbuminuria. Expert Opin. Pharmacother. 2014, 15, 453–459. [Google Scholar] [CrossRef]
  58. Gomez-Sanchez, E.P. Third-Generation Mineralocorticoid Receptor Antagonists: Why Do We Need a Fourth? J. Cardiovasc. Pharmacol. 2016, 67, 26–38. [Google Scholar] [CrossRef] [Green Version]
  59. Kolkhof, P.; Delbeck, M.; Kretschmer, A.; Steinke, W.; Hartmann, E.; Bärfacker, L.; Eitner, F.; Albrecht-Küpper, B.; Schäfer, S. Finerenone, a Novel Selective Nonsteroidal Mineralocorticoid Receptor Antagonist Protects from Rat Cardiorenal Injury. J. Cardiovasc. Pharmacol. 2014, 64, 69–78. [Google Scholar] [CrossRef] [PubMed]
  60. Takahashi, M.; Ubukata, O.; Homma, T.; Asoh, Y.; Honzumi, M.; Hayashi, N.; Saito, K.; Tsuruoka, H.; Aoki, K.; Hanzawa, H. Crystal Structure of the Mineralocorticoid Receptor Ligand-Binding Domain in Complex with a Potent and Selective Nonsteroidal Blocker, Esaxerenone (CS-3150). FEBS Lett. 2020, 594, 1615–1623. [Google Scholar] [CrossRef]
  61. Ito, S.; Itoh, H.; Rakugi, H.; Okuda, Y.; Yoshimura, M.; Yamakawa, S. Double-Blind Randomized Phase 3 Study Comparing Esaxerenone (CS-3150) and Eplerenone in Patients With Essential Hypertension (ESAX-HTN Study). Hypertension 2020, 75, 51–58. [Google Scholar] [CrossRef]
  62. Wada, T.; Inagaki, M.; Yoshinari, T.; Terata, R.; Totsuka, N.; Gotou, M.; Hashimoto, G. Apararenone in Patients with Diabetic Nephropathy: Results of a Randomized, Double-Blind, Placebo-Controlled Phase 2 Dose-Response Study and Open-Label Extension Study. Clin. Exp. Nephrol. 2021, 25, 120–130. [Google Scholar] [CrossRef]
  63. Sato, A.; Hayashi, K.; Naruse, M.; Saruta, T. Effectiveness of Aldosterone Blockade in Patients with Diabetic Nephropathy. Hypertension 2003, 41, 64–68. [Google Scholar] [CrossRef] [Green Version]
  64. Chrysostomou, A.; Pedagogos, E.; MacGregor, L.; Becker, G.J. Double-Blind, Placebo-Controlled Study on the Effect of the Aldosterone Receptor Antagonist Spironolactone in Patients Who Have Persistent Proteinuria and Are on Long-Term Angiotensin-Converting Enzyme Inhibitor Therapy, with or without an Angiotensin II Receptor Blocker. Clin. J. Am. Soc. Nephrol. 2006, 1, 256–262. [Google Scholar]
  65. Rossing, K.; Schjoedt, K.J.; Smidt, U.M.; Boomsma, F.; Parving, H.H. Beneficial Effects of Adding Spironolactone to Recommended Antihypertensive Treatment in Diabetic Nephropathy: A Randomized, Double-Masked, Cross-over Study. Diabetes Care 2005, 28, 2106–2112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Schjoedt, K.J.; Rossing, K.; Juhl, T.R.; Boomsma, F.; Rossing, P.; Tarnow, L.; Parving, H.H. Beneficial Impact of Spironolactone in Diabetic Nephropathy. Kidney Int. 2005, 68, 2829–2836. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Schjoedt, K.J.; Rossing, K.; Juhl, T.R.; Boomsma, F.; Tarnow, L.; Rossing, P.; Parving, H.H. Beneficial Impact of Spironolactone on Nephrotic Range Albuminuria in Diabetic Nephropathy. Kidney Int. 2006, 70, 536–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. van den Meiracker, A.H.; Baggen, R.G.; Pauli, S.; Lindemans, A.; Vulto, A.G.; Poldermans, D.; Boomsma, F. Spironolactone in Type 2 Diabetic Nephropathy: Effects on Proteinuria, Blood Pressure and Renal Function. J. Hypertens. 2006, 24, 2285–2292. [Google Scholar] [CrossRef]
  69. Saklayen, M.G.; Gyebi, L.K.; Tasosa, J.; Yap, J. Effects of Additive Therapy with Spironolactone on Proteinuria in Diabetic Patients Already on ACE Inhibitor or ARB Therapy: Results of a Randomized, Placebo-Controlled, Double-Blind, Crossover Trial. J. Investig. Med. 2008, 56, 714–719. [Google Scholar] [CrossRef] [PubMed]
  70. Mehdi, U.F.; Adams-Huet, B.; Raskin, P.; Vega, G.L.; Toto, R.D. Addition of Angiotensin Receptor Blockade or Mineralocorticoid Antagonism to Maximal Angiotensin-Converting Enzyme Inhibition in Diabetic Nephropathy. J. Am. Soc. Nephrol. 2009, 20, 2641–2650. [Google Scholar] [CrossRef] [Green Version]
  71. Nielsen, S.E.; Persson, F.; Frandsen, E.; Sugaya, T.; Hess, G.; Zdunek, D.; Shjoedt, K.J.; Parving, H.H.; Rossing, P. Spironolactone Diminishes Urinary Albumin Excretion in Patients with Type 1 Diabetes and Microalbuminuria: A Randomized Placebo-Controlled Crossover Study. Diabet. Med. 2012, 29, e184–e190. [Google Scholar] [CrossRef]
  72. Ziaee, A.; Abbas Vaezi, A.; Oveisi, S.; Javadi, A.; Hashemipour, S.; Kazemifar, A.M. Effects of Additive Therapy with Spironolactone on Albuminuria in Diabetes Mellitus: A Pilot Randomized Clinical Trial. Casp. J. Intern. Med. 2013, 4, 648–653. [Google Scholar]
  73. Esteghamati, A.; Noshad, S.; Jarrah, S.; Mousavizadeh, M.; Khoee, S.H.; Nakhjavani, M. Long-Term Effects of Addition of Mineralocorticoid Receptor Antagonist to Angiotensin II Receptor Blocker in Patients with Diabetic Nephropathy: A Randomized Clinical Trial. Nephrol. Dial. Transpl. 2013, 28, 2823–2833. [Google Scholar] [CrossRef] [Green Version]
  74. Kato, S.; Maruyama, S.; Makino, H.; Wada, J.; Ogawa, D.; Uzu, T.; Araki, H.; Koya, D.; Kanasaki, K.; Oiso, Y.; et al. Anti-Albuminuric Effects of Spironolactone in Patients with Type 2 Diabetic Nephropathy: A Multicenter, Randomized Clinical Trial. Clin. Exp. Nephrol. 2015, 19, 1098–1106. [Google Scholar] [CrossRef] [Green Version]
  75. Chen, Y.; Liu, P.; Chen, X.; Li, Y.; Zhang, F.; Wang, Y. Effects of Different Doses of Irbesartan Combined With Spironolactone on Urinary Albumin Excretion Rate in Elderly Patients With Early Type 2 Diabetic Nephropathy. Am. J. Med. Sci. 2018, 355, 418–424. [Google Scholar] [CrossRef] [PubMed]
  76. Nielsen, S.E.; Schjoedt, K.J.; Rossing, K.; Persson, F.; Schalkwijk, C.G.; Stehouwer, C.D.A.; Parving, H.-H.; Rossing, P. Levels of NT-ProBNP, Markers of Low-Grade Inflammation, and Endothelial Dysfunction during Spironolactone Treatment in Patients with Diabetic Kidney Disease. J. Renin Angiotensin Aldosterone Syst. 2013, 14, 161–166. [Google Scholar] [CrossRef] [Green Version]
  77. Epstein, M.; Williams, G.H.; Weinberger, M.; Lewin, A.; Krause, S.; Mukherjee, R.; Patni, R.; Beckerman, B. Selective Aldosterone Blockade with Eplerenone Reduces Albuminuria in Patients with Type 2 Diabetes. Clin. J. Am. Soc. Nephrol. 2006, 1, 940–951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. El Mokadem, M.; Abd El Hady, Y.; Aziz, A. A Prospective Single-Blind Randomized Trial of Ramipril, Eplerenone and Their Combination in Type 2 Diabetic Nephropathy. Cardiorenal Med. 2020, 10, 392–401. [Google Scholar] [CrossRef] [PubMed]
  79. Brandt-Jacobsen, N.H.; Johansen, M.L.; Rasmussen, J.; Forman, J.L.; Holm, M.R.; Faber, J.; Rossignol, P.; Schou, M.; Kistorp, C. Effect of High-Dose Mineralocorticoid Receptor Antagonist Eplerenone on Urinary Albumin Excretion in Patients with Type 2 Diabetes and High Cardiovascular Risk: Data from the MIRAD Trial. Diabetes Metab. 2021, 47, 101190. [Google Scholar] [CrossRef]
  80. Bakris, G.L.; Agarwal, R.; Chan, J.C.; Cooper, M.E.; Gansevoort, R.T.; Haller, H.; Remuzzi, G.; Rossing, P.; Schmieder, R.E.; Nowack, C.; et al. Effect of Finerenone on Albuminuria in Patients With Diabetic Nephropathy: A Randomized Clinical Trial. JAMA 2015, 314, 884–894. [Google Scholar] [CrossRef]
  81. Katayama, S.; Yamada, D.; Nakayama, M.; Yamada, T.; Myoishi, M.; Kato, M.; Nowack, C.; Kolkhof, P.; Yamasaki, Y. A Randomized Controlled Study of Finerenone versus Placebo in Japanese Patients with Type 2 Diabetes Mellitus and Diabetic Nephropathy. J. Diabetes Complicat. 2017, 31, 758–765. [Google Scholar] [CrossRef] [PubMed]
  82. Carrero, J.J.; Grams, M.E.; Sang, Y.; Ärnlöv, J.; Gasparini, A.; Matsushita, K.; Qureshi, A.R.; Evans, M.; Barany, P.; Lindholm, B.; et al. Albuminuria Changes Are Associated with Subsequent Risk of End-Stage Renal Disease and Mortality. Kidney Int. 2017, 91, 244–251. [Google Scholar] [CrossRef] [Green Version]
  83. Heerspink, H.J.L.; Greene, T.; Tighiouart, H.; Gansevoort, R.T.; Coresh, J.; Simon, A.L.; Chan, T.M.; Hou, F.F.; Lewis, J.B.; Locatelli, F.; et al. Change in Albuminuria as a Surrogate Endpoint for Progression of Kidney Disease: A Meta-Analysis of Treatment Effects in Randomised Clinical Trials. Lancet Diabetes Endocrinol. 2019, 7, 128–139. [Google Scholar] [CrossRef]
  84. Agarwal, R.; Filippatos, G.; Pitt, B.; Anker, S.D.; Rossing, P.; Joseph, A.; Kolkhof, P.; Nowack, C.; Gebel, M.; Ruilope, L.M.; et al. Cardiovascular and Kidney Outcomes with Finerenone in Patients with Type 2 Diabetes and Chronic Kidney Disease: The FIDELITY Pooled Analysis. Eur. Heart J. 2021, 43, 474–484. [Google Scholar] [CrossRef] [PubMed]
  85. Filippatos, G.; Bakris, G.L.; Pitt, B.; Agarwal, R.; Rossing, P.; Ruilope, L.M.; Butler, J.; Lam, C.S.P.; Kolkhof, P.; Roberts, L.; et al. Finerenone Reduces New-Onset Atrial Fibrillation in Patients With Chronic Kidney Disease and Type 2 Diabetes. J. Am. Coll. Cardiol. 2021, 78, 142–152. [Google Scholar] [CrossRef]
  86. Filippatos, G.; Anker, S.D.; Agarwal, R.; Pitt, B.; Ruilope, L.M.; Rossing, P.; Kolkhof, P.; Schloemer, P.; Tornus, I.; Joseph, A.; et al. Finerenone and Cardiovascular Outcomes in Patients with Chronic Kidney Disease and Type 2 Diabetes. Circulation 2021, 143, 540–552. [Google Scholar] [CrossRef]
  87. Perkovic, V.; Jardine, M.J.; Neal, B.; Bompoint, S.; Heerspink, H.J.L.; Charytan, D.M.; Edwards, R.; Agarwal, R.; Bakris, G.; Bull, S.; et al. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N. Engl. J. Med. 2019, 380, 2295–2306. [Google Scholar] [CrossRef] [Green Version]
  88. Heerspink, H.J.L.; Stefánsson, B.V.; Correa-Rotter, R.; Chertow, G.M.; Greene, T.; Hou, F.-F.; Mann, J.F.E.; McMurray, J.J.V.; Lindberg, M.; Rossing, P.; et al. Dapagliflozin in Patients with Chronic Kidney Disease. N. Engl. J. Med. 2020, 383, 1436–1446. [Google Scholar] [CrossRef] [PubMed]
  89. Rossing, P.; Filippatos, G.; Agarwal, R.; Anker, S.D.; Pitt, B.; Ruilope, L.M.; Chan, J.C.N.; Kooy, A.; McCafferty, K.; Schernthaner, G.; et al. Finerenone in Predominantly Advanced CKD and Type 2 Diabetes With or Without Sodium-Glucose Cotransporter-2 Inhibitor Therapy. Kidney Int. Rep. 2022, 7, 36–45. [Google Scholar] [CrossRef] [PubMed]
  90. Derosa, G.; Gaudio, G.; Pasini, G.; D’Angelo, A.; Maffioli, P. A Randomized, Double-Blind Clinical Trial of Canrenone vs Hydrochlorothiazide in Addition to Angiotensin II Receptor Blockers in Hypertensive Type 2 Diabetic Patients. Drug Des. Dev. Ther. 2018, 12, 2611–2616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Ito, S.; Shikata, K.; Nangaku, M.; Okuda, Y.; Sawanobori, T. Efficacy and Safety of Esaxerenone (CS-3150) for the Treatment of Type 2 Diabetes with Microalbuminuria: A Randomized, Double-Blind, Placebo-Controlled, Phase II Trial. Clin. J. Am. Soc. Nephrol. 2019, 14, 1161–1172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Ito, S.; Kashihara, N.; Shikata, K.; Nangaku, M.; Wada, T.; Okuda, Y.; Sawanobori, T. Esaxerenone (CS-3150) in Patients with Type 2 Diabetes and Microalbuminuria (ESAX-DN): Phase 3 Randomized Controlled Clinical Trial. Clin. J. Am. Soc. Nephrol. 2020, 15, 1715–1727. [Google Scholar] [CrossRef] [PubMed]
  93. Ito, S.; Kashihara, N.; Shikata, K.; Nangaku, M.; Wada, T.; Okuda, Y.; Sawanobori, T. Efficacy and Safety of Esaxerenone (CS-3150) in Japanese Patients with Type 2 Diabetes and Macroalbuminuria: A Multicenter, Single-Arm, Open-Label Phase III Study. Clin. Exp. Nephrol. 2021, 25, 1070–1078. [Google Scholar] [CrossRef] [PubMed]
  94. Zheng, Y.; Ma, S.; Huang, Q.; Fang, Y.; Tan, H.; Chen, Y.; Li, C. Meta-Analysis of the Efficacy and Safety of Finerenone in Diabetic Kidney Disease. Kidney Blood Press. Res. 2022. [CrossRef] [PubMed]
Figure 1. Actions of aldosterone through activation of the mineralocorticoid receptor (MR) in principal cells of the distal nephron. MR displays similar affinity for aldosterone and cortisol and ligand selectivity in the kidney is maintained by pre-receptor modulation. Despite the fact that cortisol reaches 100- to 1000-fold higher concentrations in several tissues, the 11-beta-hydroxysteroid-dehydrogenase-2 (11-HSD2) enzyme in tubular epithelial cells leads to conversion of cortisol to inactive cortisone, thereby rendering aldosterone the primary physiological MR ligand in the distal nephron. The genomic process begins after free diffusion of aldosterone through the cell membrane and fastening to the aldosterone ligand-binding domain of the MR. Upon ligand binding, transcription is dependent on recruitment of coregulatory proteins (coactivators or corepressors) and requires translocation of the MR-ligand complex into the nucleus. The final step of this process is upregulation of synthesis of the epithelial sodium channel (ENaC) at the apical membrane and of the Na/K–ATPase at the basolateral membrane, leading to renal sodium reabsorption and potassium secretion. In this classical genomic response, a latency of 30–60 min is expected after release or administration of aldosterone.
Figure 1. Actions of aldosterone through activation of the mineralocorticoid receptor (MR) in principal cells of the distal nephron. MR displays similar affinity for aldosterone and cortisol and ligand selectivity in the kidney is maintained by pre-receptor modulation. Despite the fact that cortisol reaches 100- to 1000-fold higher concentrations in several tissues, the 11-beta-hydroxysteroid-dehydrogenase-2 (11-HSD2) enzyme in tubular epithelial cells leads to conversion of cortisol to inactive cortisone, thereby rendering aldosterone the primary physiological MR ligand in the distal nephron. The genomic process begins after free diffusion of aldosterone through the cell membrane and fastening to the aldosterone ligand-binding domain of the MR. Upon ligand binding, transcription is dependent on recruitment of coregulatory proteins (coactivators or corepressors) and requires translocation of the MR-ligand complex into the nucleus. The final step of this process is upregulation of synthesis of the epithelial sodium channel (ENaC) at the apical membrane and of the Na/K–ATPase at the basolateral membrane, leading to renal sodium reabsorption and potassium secretion. In this classical genomic response, a latency of 30–60 min is expected after release or administration of aldosterone.
Kidneydial 02 00019 g001
Table 1. Main studies on the effects of spironolactone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
Table 1. Main studies on the effects of spironolactone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
StudyStudy
Design
Follow
Up
Nr of
Patients
Patient
Characteristics
ComparatorBack-
Ground Treatment
Main Results
Sato et al., 2003 [63]Open-label, single-arm study24 weeks13T2DM, UACR 30–300 mg/g and creatinine clearance > 60 mL/min-Trandolapril↓ UAE and LVMI for spironolactone (p < 0.05 for both)
Non-significant ↑ K (before 4.2 ± 0.3 mEq/L; after 4.3 ± 0.2 mEq/L)
Rossing et al.,
2005 [65]
Double-blind, cross-over RCT8 weeks21T2DM and UAE > 300 mg/24 hPlaceboACEi or ARB↓ UAE −33% (95%CI −41%, −25%); 24 h SBP −6 mmHg (95%CI −10, −2); 24 h DBP −4 mmHg (95%CI −6, −2); eGFR −3 mL/min/1.73 m2 (95%CI −0.3, +6.0) for spironolactone
Schjoedt et al., 2005 [66]Double-blind, cross-over RCT8 weeks20T1DM and
UAE > 300 mg/24 h
PlaceboACEi or ARB↓ UAE −30% (95%CI −41%, –17%); 24 h SBP −8 mmHg (95%CI −17, +1); 24 h DBP −3 mmHg (95%CI −7, +0.2); eGFR by −3.4 mL/min/ 1.73 m2 (−6.9, 0.1) for spironolactone
1 patient excluded due to hyperK in spironolactone
Schjoedt et al., 2006 [67]Double-blind, cross-over RCT8 weeks20T1DM or T2DM and
UAE > 2500 mg/24 h
PlaceboACEi or ARB↓ UAE −32% (95%CI −42%, −21%); 24 h SBP −6 mmHg (95%CI −10, −2); 24 h DBP −4 mmHg (95%CI −6, −2); eGFR −3 mL/min/1.73 m2 (−6.0, +1.0)
↑ K +0.2 mmol/L (95%CI −0.004, +0.5)
van den Meiracker, 2006 [68]Double-blind, parallel-group RCT1 year59T2DM and
UAE > 300 mg/24 h or UACR > 20 mg/g
PlaceboACEi or ARB↓ UAE −40.6% (95%CI −57.8%, −23.4%); 24 h SBP −7 mmHg (95%CI −12, −2); 24 h DBP −3 mmHg (95%CI −6, −1)
eGFR −12.9 mL/min/1.73 m2 (−16.5, −9.5) for spironolactone; −4.9 mL/min/1.73 m2 (−8.9, −0.8) for placebo
5 patients excluded due to hyperK in spironolactone
Saklayen et al., 2008 [69]Double-blind, cross-over RCT7 weeks30T1DM or T2DM patients with any level of proteinuriaPlaceboACEi or ARB↓ UPCR from 1.80 ± 1.78 to 0.79 ± 0.99 for spironolactone (p = 0.004); from 1.24 ± 1.13 to 1.57 ± 2.13 for placebo (p = 0.35); eGFR from 61.91 ± 23.4 to 53.94 ± 23.58 for spironolactone (p = 0.0001)
Mehdi et al.,
2009 [70]
Double-blind parallel-group RCT48 weeks81T1DM or T2DM patients and UACR > 300 mg/gPlacebo,
Losartan
100 mg
Lisinopril 80 mg↓ UACR −34% (95%CI −51%, −11.2%) for spironolactone (p = 0.007 vs. placebo); −16.8% (95%CI −37.3%, +10.5%) for losartan (p = 0.2 vs. placebo); % change in creatinine clearance −13.1% (95%CI −21.3%, −3.9%) for spironolactone; −16.8% (95%CI −23.9%, −9.1%) for losartan; −16.0% (95% CI −23.3%, −7.9%) for placebo
HyperK episodes (>6.0 mmol/L): 14 patients in spironolactone (p < 0.001 vs. placebo); 10 in losartan (p = 0.009 vs. placebo); 2 patients in placebo
Nielsen et al.,
2012 [71]
Double-blind, cross-over RCT60 days21T1D and UAE > 30 mg/dayPlaceboACEi or ARB↓ UACR −60% (range −80% to −21%); eGFR from 78 ± 6 to 72 ± 6 mL/min/1.73 m2 (p = 0.003)
HyperK episodes (>5.7 mmol/L): 2 patients in spironolactone group
Ziaee et al., 2013 [72]Parallel-group RCT12 weeks60T2DM and microalbuminuriaPlaceboEnalapril↓ UACR from 126 ± 69.3 to 59.3 ± 48.1 for spironolactone (p < 0.001); eGFR from 79.8 ± 18 to 75.6 ± 16.3 mL/min/1.73 m2 for spironolactone (p = 0.6)
Esteghamati et al.,
2013 [73]
Open-label, parallel-group RCT18 months136T2DM and UAE ≥ 30 mg/dayEnalaprilLosartan↓ UAE −60.5 mg (95%CI −148.8, −16.4) for spironolactone; +22.0 mg (95%CI −110.3, +108.9) for placebo (p = 0.017); SBP −8.89 mmHg (95%CI −15.88, −1.89) for spironolactone; −6.08 mmHg (−14.71, +2.57) for placebo (p < 0.001); DBP −4.44 mmHg (95%CI −8.10, −0.79) for spironolactone; −2.86 (−7.06, +1.34) for placebo (p = 0.001); eGFR −10.23 mL/min/1.73 m2 (95%CI −16.69, −3.76) for spironolactone; −9.08 mL/min/1.73 m2 (−16.06, −2.10) for placebo (p = 0.674)
Oxlund et al., 2013 [13]Double-blind, parallel-group RCT16 weeks119T2DM and resistant hypertensionPlaceboACEi or ARB↓ UACR −7.3 mg/g (95%CI −1093, +12.2) for spironolactone; +0 mg/g (95% +7, +146.3) for placebo (p = 0.001); placebo-corrected 24 h SBP −8.9 mmHg (95%CI −13.2, −4.6); placebo-corrected 24 h DBP −3.9 mmHg (95%CI −6.2, −1.7)
↑ K +0.26 mmol/L (95%CI +0.1, +0.4) for spironolactone; +0.02 (95%CI +0.07, +0.10) for placebo (between-group p < 0.001)
Kato et al.,
2015 [74]
Open-label parallel-group RCT8 weeks52T2DM and UACR 100–2000 mg/gPlaceboACEi or ARB↓ UACR −33% (95%CI −54%, −22%);
eGFR −3.2 ± 9.7 mL/min/1.73 m2 (p = 0.052)
Chen et al., 2018 [75]Open-label, parallel-group RCT72 weeks244T2DM and UAER 20–199 μg/minPlaceboIrbesartan 150 mg or 300 mg↓ UAΕR −30 μg/min (95%CI −54, −15) for spironolactone/irbesartan 300 mg; −30 μg/min (95%CI −51, −12) for spironolactone/irbesartan 150 mg; −23 μg/min (95%CI
−35, −12) for irbesartan 300 mg; −15 μg/min (95%CI −24, −11) for irbesartan 150 mg (between-group p < 0.001)
↓, decrease; ↑, increase; ACEi, angiotensin converting enzyme inhibitor; ARB, angiotensin receptor blocker; CI, confidence interval; DBP, diastolic blood pressure; eGFR, estimated glomerular filtration rate; hyperK, hyperkalemia; K, potassium; LVMi, left ventricular mass index; RCT, randomized controlled trial; SBP, systolic blood pressure; T2DM, type 2 diabetes mellitus; UACR, urinary albumin creatinine ratio; UAER, urinary albumin excretion rate.
Table 2. Main studies on the effects of eplerenone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
Table 2. Main studies on the effects of eplerenone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
StudyStudy
Design
Follow
Up
Nr of
Patients
Patient
Characteristics
ComparatorBack-
Ground
Treatment
Main Results
Epstein et al., 2006 [77]Double-blind, parallel-group RCT12 weeks268T2DM and UACR ≥ 50 mg/gPlaceboEnalapril 20 mg↓ UACR −41% for eplerenone 50 mg; −48.4% for eplerenone 100 mg; −7.4% for placebo (p < 0.001 vs. placebo for both)
Between-group differences in sustained hyperK (>5.5 mmol/L on two consecutive measurements) p = 0.29; severe hyperK (≥6.0 mmol/L at any timepoint) p = 0.38
Brandt-Jacobsen et al., 2020 (MIRAD trial) [79]Double-blind, parallel-group RCT26 weeks140T2DM, median UACR 17 mg/g, 12% had eGFR < 60 mL/min/1.73 m2PlaceboAntihypertensive treatment↓ UACR by −34% for eplerenone vs. placebo (p = 0.005); eGFR −3.5 mL/min/1.73 m2 for eplerenone
Between-group differences in episodes of hyperkalemia (≥5.5 mmol/L) p = 0.276
↑ K by +0.26 mmol/L for eplerenone
Mokadem et al., 2020 [78]Single-blind, parallel-group RCT24 weeks75T2DM and UACR 30–300 mg/g and stage 1 hypertensionTreatment groups:
Eplerenone/ramipril combination,
ramipril monotherapy, eplerenone monotherapy
↓ UACR −70% for eplerenone/ramipril; −37% for ramipril; −38% for eplerenone (p < 0.0001 for combination vs. both others)
HyperK episodes (>5.5 mmol/L on 2 measurements): 8% for eplerenone/ramipril; 4% for ramipril; 4% for eplerenone (for eplerenone/ramipril vs. others p = 0.5, ramipril vs. eplerenone p = 0.6); for eGFR < 60 mL/min/1.73 m2: ↑ incidence of hyperK for eplerenone/ramipril vs. others (p < 0.05)
↓, decrease; ↑, increase; eGFR, estimated glomerular filtration rate; hyperK, hyperkalemia; K, potassium; RCT, randomized controlled trial; T2DM, type 2 diabetes mellitus; UACR, urinary albumin creatinine ratio.
Table 3. Main studies on the effects of finerenone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
Table 3. Main studies on the effects of finerenone in albuminuria, renal and cardiovascular outcomes in patients with DKD.
StudyStudy
Design
Follow
UP
Nr of
Patients
Patient
Characteristics
ComparatorBack-
Ground
Treatment
Main Results
Bakris et al., 2015
(ARTS-DN)
[80]
Double-blind, parallel-group RCT90 days821 (4 different finerenone dose groups)T2DM and UACR 30 to <300 mg/g or >300 mg/g (stratified randomization)PlaceboACEi or ARB↓ placebo-corrected mean ratio of UACR at day 90 relative to baseline: finerenone 7.5 mg 0.79 (p = 0.004); finerenone 10 mg 0.76 (p = 0.001); finerenone 15 mg 0.67 (p < 0.001); finerenone 20 mg 0.62 (p < 0.001)
Significantly ↑ incidence of hyperK episodes leading to study discontinuation: finerenone 7.5 mg 2.1%, finerenone 15 mg 3.2% and finerenone 20 mg 1.7%
No significant ↑ in the risk of hyperkalemia for placebo and finerenone 10 mg
Katayama et al.,
2017 [81]
Double-blind, parallel-group RCT90 days96 (4 different finerenone dose groups)T2DM and UACR 30 to <300 mg/g or >300 mg/g (stratified randomization)PlaceboACEi or ARB↓ LS mean ratio of finerenone to baseline (0.712); LS mean ratio of finerenone to placebo (0.670) for finerenone 20 mg (p = 0.0240)
↑ K for finerenone (+0.025, +0.167 mmol/L) vs. placebo (−0.075 mmol/L)
Bakris et al.,
2020 (FIDELIO-
DKD) [23]
Double-blind, parallel-group RCT2.6 years5734T2DM and:
(a) UACR 300–5000 mg/g and eGFR 25–75 mL/min/1.73 m2 or
(b) UACR 30–300 mg/g, eGFR 25–60 mL/min/1.73 m2, diabetic retinopathy
PlaceboACEi or ARBPrimary composite endpoint of kidney failure (ESKD or eGFR < 15 mL/min/1.73 m2), eGFR decrease of ≥40%, renal death: HR 0.82; 95% CI 0.73–0.93
Secondary: kidney failure HR 0.87; 95% CI 0.72–1.05; eGFR decrease of ≥40% HR 0.81; 95% CI 0.72–0.92
Secondary composite endpoint of kidney failure (ESKD or eGFR <15 mL/min/1.73 m2), eGFR decrease of ≥57%, renal death: HR 0.76; 95% CI 0.65–0.90
Secondary composite endpoint of CV death, nonfatal MI/stroke, HHF: HR 0.86; 95% CI 0.75–0.99
Secondary: CV death HR 0.86; 95% 0.68–1.08; nonfatal MI HR 0.80; 95% CI 0.58–1.09; nonfatal stroke: HR 1.03; 95%CI 0.76–1.38; HHF HR 0.86; 95%CI 0.68–1.08HyperK leading to drug discontinuation: 2.3% for finerenone; 0.9% for placebo
Pre-specified secondary analysis: new-onset AF HR 0.71, 95%CI 0.53–0.94; fatal/nonfatal stroke after new-onset AF: HR 7.13; 95%CI 4.01–12.70
Pitt et al., 2021 (FIGARO-
DKD) [24]
Double-blind, parallel-group RCT3.4 years7437T2DM and:
(a) UACR 30–300 mg/g, eGFR ≥ 25–90 mL/min/
1.73 m2 or
(b) UACR 300–5000 mg/g, eGFR ≥ 60 mL/min/1.73 m2
PlaceboACEi or ARBPrimary composite endpoint of CV death, nonfatal MI/stroke, HHF: HR 0.87; 95%CI 0.76–0.98
Secondary: HHF HR 0.71; 95%CI 0.56–0.90
Secondary composite endpoint of kidney failure (ESKD or eGFR <15 mL/min/1.73 m2), eGFR decrease of ≥40%, renal death: HR 0.87; 95%CI 0.76–1.01
Secondary: ESKD HR 0.64, 95%CI 0.41–0.995HyperK leading to drug discontinuation: 1.2% finerenone; 0.4% placebo
↓, decrease; ↑, increase; ACEi, angiotensin converting enzyme inhibitor; ARB, angiotensin receptor blocker; CI, confidence interval; CV, cardiovascular; eGFR, estimated glomerular filtration rate; ESKD, end-stage kidney disease; HHF, hospitalization for heart failure; hyperK, hyperkalemia; K, potassium; HR, hazard ratio; LS, least-squares; RCT, randomized controlled trial; T2DM, type 2 diabetes mellitus; UACR, urinary albumin creatinine ratio.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alexandrou, M.-E.; Theodorakopoulou, M.P.; Sarafidis, P.A. Role of Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease. Kidney Dial. 2022, 2, 163-182. https://doi.org/10.3390/kidneydial2020019

AMA Style

Alexandrou M-E, Theodorakopoulou MP, Sarafidis PA. Role of Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease. Kidney and Dialysis. 2022; 2(2):163-182. https://doi.org/10.3390/kidneydial2020019

Chicago/Turabian Style

Alexandrou, Maria-Eleni, Marieta P. Theodorakopoulou, and Pantelis A. Sarafidis. 2022. "Role of Mineralocorticoid Receptor Antagonists in Diabetic Kidney Disease" Kidney and Dialysis 2, no. 2: 163-182. https://doi.org/10.3390/kidneydial2020019

Article Metrics

Back to TopTop