Next Article in Journal
Structure-Function Studies of Sponge-Derived Compounds on the Cardiac CaV3.1 Channel
Next Article in Special Issue
Scavenging of Superoxide in Aprotic Solvents of Four Isoflavones That Mimic Superoxide Dismutase
Previous Article in Journal
Zebrafish as a Potential Model for Neurodegenerative Diseases: A Focus on Toxic Metals Implications
Previous Article in Special Issue
Brain Waste Removal System and Sleep: Photobiomodulation as an Innovative Strategy for Night Therapy of Brain Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Endothelin Receptor Antagonists in Kidney Disease

by
Irene Martínez-Díaz
1,†,
Nerea Martos
1,†,
Carmen Llorens-Cebrià
1,
Francisco J. Álvarez
2,
Patricia W. Bedard
2,
Ander Vergara
1,*,
Conxita Jacobs-Cachá
1,* and
Maria José Soler
1
1
Nephrology and Transplantation Research Group, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
2
Travere Therapeutics, Inc., San Diego, CA 92130, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(4), 3427; https://doi.org/10.3390/ijms24043427
Submission received: 30 December 2022 / Revised: 25 January 2023 / Accepted: 28 January 2023 / Published: 8 February 2023
(This article belongs to the Special Issue 23rd Anniversary of IJMS: Advances in Biochemistry)

Abstract

:
Endothelin (ET) is found to be increased in kidney disease secondary to hyperglycaemia, hypertension, acidosis, and the presence of insulin or proinflammatory cytokines. In this context, ET, via the endothelin receptor type A (ETA) activation, causes sustained vasoconstriction of the afferent arterioles that produces deleterious effects such as hyperfiltration, podocyte damage, proteinuria and, eventually, GFR decline. Therefore, endothelin receptor antagonists (ERAs) have been proposed as a therapeutic strategy to reduce proteinuria and slow the progression of kidney disease. Preclinical and clinical evidence has revealed that the administration of ERAs reduces kidney fibrosis, inflammation and proteinuria. Currently, the efficacy of many ERAs to treat kidney disease is being tested in randomized controlled trials; however, some of these, such as avosentan and atrasentan, were not commercialized due to the adverse events related to their use. Therefore, to take advantage of the protective properties of the ERAs, the use of ETA receptor-specific antagonists and/or combining them with sodium-glucose cotransporter 2 inhibitors (SGLT2i) has been proposed to prevent oedemas, the main ERAs-related deleterious effect. The use of a dual angiotensin-II type 1/endothelin receptor blocker (sparsentan) is also being evaluated to treat kidney disease. Here, we reviewed the main ERAs developed and the preclinical and clinical evidence of their kidney-protective effects. Additionally, we provided an overview of new strategies that have been proposed to integrate ERAs in kidney disease treatment.

1. Introduction: The Endothelin System

Endothelin (ET) is a 21-aminoacid polypeptide described as the major vasoconstrictor of the organism. It is produced mainly by endothelial cells, but also by cells of the renal system, such as the epithelial and mesangial cells [1]. Hickey et al. were the first to describe the existence of a molecule capable of causing capillary constriction produced by the endothelium in 1985 but it was not until 1988 that ET was identified [2,3]. The ET polypeptide is present in three isoforms: ET-1, ET-2 and ET-3, with ET-1 being the greatest vasoconstrictor and the only one found at the protein level in the kidney [4]. ET-2 and ET-3 differ from ET-1 in two and five residues of the N-terminal end, respectively (Table 1), which determines the differences on the receptor-binding affinity [5]. Moreover, ET-1 is mainly released by endothelial cells, while the intestine and the kidney produce ET-2 and the neural tissue releases ET-3; the three isoforms can act in a paracrine or autocrine manner [5].
The action of ET is channelled through two membrane G-protein coupled receptors: Endothelin receptor A (ETA) and B (ETB). ETA is localized in vascular smooth muscle cells and presents more binding affinity for ET-1 and ET-2 than for ET-3, due to the differences in the amino acid sequences (Table 1). ETA activation induces a robust vasoconstrictor response and promotes cell proliferation and accumulation of the extracellular matrix. ETB is present in vascular smooth muscle cells and endothelial cells. The three ET isoforms present the same affinity for the ETB receptor, and its activation produces antiproliferative and antifibrotic effects, as well as the release of various vasodilator molecules [6]. Some experts have suggested an extended classification of the ET receptors, subdividing ETB into ETB1 and ETB2 to differentiate the receptors present on the endothelial cells and the ones present on the smooth muscle cells, respectively. Nonetheless, there is no pharmacological evidence that demonstrates a difference between the receptors expressed by these two cell types [7]. ET-1 binding to ETA causes G-proteins and phospholipase C (PLC) to join, leading to inositol triphosphate (IP3) and diacylglycerol (DAG) formation. Then, IP3 activates specific endoplasmic reticulum receptors to stimulate the release of stored Ca2+ causing a fast increase in intracellular Ca2+, which allows cell contraction and subsequent vasoconstriction. ET-1 activity through ETA receptors also involves other signalling pathways, such as the phospholipase D (PLD) or mitogen-activated protein kinase (MAPK) pathway, to carry out other physiological effects such as cell growth or mitogenesis [5]. Contrarily, ETB receptors produce their vasodilator effects through the activation of the nitric oxide synthases (NOS) system and the release of vasodilators as nitric oxide (NO) [8,9].
In the kidney, ET has an essential role in blood flow and glomerular filtration regulation and in water–sodium and acid–base balances. ETA and ETB are expressed on the glomerular podocytes, mesangial cells and on the afferent and efferent arterioles. Regarding the tubular compartment, ETB is expressed in all the regions in the renal tubule while ETA is scarcely expressed on the proximal tubule and the descending Henle’s loop [10]. In physiological conditions, ET-1 through ETA produces vasoconstriction of the afferent arteriole, reducing blood flow and, consequently, the glomerular filtration rate (GFR). Contrarily, the activation of ETB induces vasodilation, antiproliferative effects and ET-1 depuration [8,9]. In pathological conditions, such as diabetes or hypertension, the concentration of ET-1 is increased because of the hyperglycaemia, acidosis and the presence of insulin, angiotensin II and proinflammatory cytokines, which causes sustained vasoconstriction. This may contribute to deleterious effects such as hyperfiltration (mainly in early diabetic nephropathy or incipient obesity-related kidney disease [11,12,13]) or podocyte damage and, eventually, proteinuria and GFR decline (Figure 1) [14].
The endothelin receptor antagonists (ERA) are postulated as a therapeutic strategy to reduce proteinuria and delay the progression of GFR decline [14]. Promising results using ERAs in kidney disease have been obtained in recent years. The purpose of this review is to provide an overview of the main ERAs and of the mechanisms by which these drugs protect the kidney with a special focus on the results obtained in updated experimental studies and randomized clinical trials. Additionally, we provide a glance of the main novel approaches to introduce ERAs for kidney disease prevention.

2. Methods

We searched PubMed, Scopus and Google academic during November-December 2022 using the following search terms (alone of combined) to find publications related to the endothelin system and the endothelin receptor antagonists in experimental and human kidney disease: “endothelin or ET”, “endothelin receptor antagonists or ERA”, “endothelin receptor A or ETA”, “endothelin receptor B or ETB”, “kidney”, “kidney disease”, “chronic kidney disease”, ”kidney injury”, “experimental models”, “mice”, ”rat”, “podocytes”, “randomized clinical trials”. We critically reviewed the reports found and selected the relevant studies to construct the review text. For the sections “5. Preclinical experimental evidence of ERAs protective effects on kidney damage” and “6. Randomized controlled trials (RCTs) using ERAs for prevention of kidney disease progression”, we mainly focused on the works published in the last five years to not overlap previous reviews on the topic. The complete literature review strategy is available from the authors upon request. Further, we consulted https://clinicaltrials.gov during November–December 2022 to obtain information regarding unpublished ongoing clinical trials that we have stated using the corresponding National Clinical Trial (NCT) number.

3. The Endothelin Receptor Antagonists

Endothelin receptor antagonists (ERAs) are drugs that block the endothelin receptors, preventing the endothelin action. There exist different types of ERAs, which can be distinguished by their affinity for binding to ETA or ETB. In some cases, these antagonists do not present selectivity and are able to interact with both receptors. The selectivity of an ERA for each receptor subtype is determined by a competition binding assay against [125I]-ET-1 that allows to calculate the equilibrium dissociation constant of each compound to both receptors, ETA, and ETB. To establish a selectivity threshold, in 2006, Maguire and Davenport et al. [15] proposed that an ERA should present more than 100-fold selectivity for ETA or ETB to be considered selective for one or the other receptor. Those with less than 100-fold selectivity should be classified as non-selective or mixed antagonists [15]. The main ERAs are summarized in Table 2.
The majority of the antagonists here described are still under investigation in ongoing clinical trials, while others are not used in clinical practice because of lack of efficacy or due to the presence of adverse events related to their use that compromise the safety of patients. Therefore, to take advantage of these compounds with clear beneficial effects (Table 2), therapeutic approaches under study are the combination of ERAs with other nephroprotective drugs such as sodium-glucose cotransporter 2 inhibitors (SGLT2i) and the use of dual drugs such as sparsentan that blocks at the same time angiotensin-II type 1 and endothelin receptors.

3.1. ETA-Selective Receptor Antagonists

The binding of ET-1 to ETA, in pathologic conditions, can lead to vasoconstriction, inflammation, cellular injury, fibrosis, and, finally to proteinuria and loss of renal function [31]. To counteract these effects, several selective ETA-receptor antagonists have been developed as potential therapeutic agents. Some of them are currently used to treat pulmonary arterial hypertension (ambrisentanand macitentan), meanwhile others are still under study in ongoing randomized clinical studies. To date, none of the ETA-selective receptor antagonists have been approved to treat kidney disease, despite their demonstrated kidney protective effects (Table 2).
BQ-123 was the first ETA selective antagonist peptide isolated that derived from Streptomyces misakiensis fermentation products. It has been used in investigation in both animals and humans where the molecule reduced glomerular permeability [20]. Darusentan (LU 135252) is a selective endothelin receptor antagonist, with high affinity towards ETA receptor [17], with a ratio of relative selectivity of 170:1 ETA:ETB [32]. It is derived from the optimization of two initial lead structures (LU 110896 and LU 110897) found in a screening of the library of human recombinant ETA receptors [33]. Darusentan was promising because reduced blood pressure in resistant hypertension patients in early clinical studies but unfortunately failed to achieve efficacy in phase III clinical studies [32]. Other examples of ETA selective antagonists are sitaxentan, ambrisentan, avosentan, atrasentan, macitentan and zibotentan (Table 2). Currently, sitaxentan, ambrisentan and macitentan are approved to treat pulmonary arterial hypertension [21,34,35] while avosentan, atrasentan and zibotentan have been proposed as therapeutic agents in kidney disease [19,24,36]. Macitentan is a sulfamide with high affinity for ETA that has been used in pulmonary arterial hypertension since its approval in 2013 [37,38]. It belongs to the next generation of antagonists, as it was developed following the structural basis of bosentan [20], but with improvements such as a prolonged receptor binding capacity and better pharmacodynamics and pharmacokinetics [37,38]. In vivo, macitentan is metabolized by cytochrome P450 3A4 (CYP3A4) into an active metabolite, which is called aprocitentan (a non-selective ERA, see below) [26]. Avosentan is a selective ETA inhibitor which presents ~500-fold selectivity for ETA over ETB receptor [39]. It was developed for the treatment of diabetic nephropathy. A study performed by Wenzel et al. in 2009 [23] demonstrated that the addition of avosentan to the standard of care antihypertensive therapy with RAS blockers produced additional antiproteinuric effects in diabetic nephropathy patients. However, a second study to test long-term treatment with avosentan was stopped prematurely because of safety concerns [36]. Atrasentan is an oral selective ETA inhibitor with a selective ETA:ETB blockade ratio of 1200:1 [40] and 1800-fold selectivity for ETA [39]. In patients with diabetes and chronic kidney disease, atrasentan reduces the risk of renal events and albuminuria [19,22,41]. Currently, it is under study in an ongoing phase 2 clinical trial to evaluate the efficacy and safety of atrasentan in patients with proteinuric glomerular diseases (AFFINITY: Atrasentan in Patients with Proteinuric Glomerular Diseases—NCT04573920). Finally, zibotentan is a selective ETA antagonist, which shows a potent affinity to this specific receptor [42]. In a recent clinical trial, it has been demonstrated that zibotentan could be beneficial for the treatment of systemic sclerosis-associated chronic kidney disease because its effects in the improvement of estimated GFR and the absence of increased endothelin serum levels during treatment [24].

3.2. ETB-Selective Receptor Antagonists

Few ETB-selective antagonists have been developed. This can be explained by the fact that when endothelin binds ETB, it triggers beneficial effects such as vasodilation; hence inhibiting the action of ETB may not be a suitable therapeutic strategy. In addition, ETB-selective antagonists are usually less potent than ETA-selective agonists [20]. However, some small molecules have been developed to block ETB, such as non-peptide RO468443 that displays 2000-fold ETB selectivity [43], and A192621 [20]. Nevertheless, the most important ETB-selective antagonist is BQ-788, which was described for the first time by Ishikawa et al. in 1994. BQ-788 has been studied in combination with BQ-123, an ETA-selective ERA, and it causes the reduction of glomerular permeability to albumin but does not add to the effect of BQ-123 in monotherapy [39]. In cancer, BQ-788 inhibits cell growth and induces the death of melanoma cells, both in vivo and in vitro [26].

3.3. Non-Selective Endothelin Receptor Antagonists

Some ERAs can interact with either ETA or ETB receptor. Some of the non-selective ERAs are bosentan, tezosentan and aprocitentan. Bosentan is a non-peptide derivative dual endothelin receptor antagonist with affinity to both receptors ETA and ETB, but with barely higher affinity towards ETA (ETA:ETB 20:1) [22,35,40]. Currently it is used in the treatment of pulmonary arterial hypertension [34] and in paediatric idiopathic pulmonary hypertension [22]. Bosentan decreases vascular resistance, resulting in an increasing cardiac output without disrupting the heart rate. It also plays a role in the inhibition of endothelial cell proliferation [25]. Tezosentan is a dual endothelin receptor antagonist with a selectivity ratio of 30:1 ETA:ETB [44]. It was developed for the treatment of heart failure and preclinical studies have shown that tezosentan improves hemodynamics and renal function in rats [26] but does not improve dyspnoea or reduce the risk of cardiovascular events [26]. Aprocitentan (ACT-132577) is a dual inhibitor of ETA/ETB with a selective ratio of 1:16 [45,46,47]. It belongs to the sulfonamide class of molecules and is obtained by oxidative depropylation from macitentan [45,46,47]. In the PRECISION clinical trial, finished in 2022, aprocitentan lowered blood pressure in patients with resistant hypertension [27].

3.4. Other Types of Endothelin Receptor Antagonists

Sparsentan (BMS-346567) is a dual endothelin receptor/angiotensin-II type 1 receptor antagonist (DEARA) which presents high affinity for ETA (~1000-fold). It was created by combining structural elements of both irbesartan, an angiotensin II type 1 receptor antagonist, and biphenylsulfonamide, an endothelin receptor antagonist. Thus, sparsentan blocks at the same time the RAS and the endothelin system reason why it is expected to show additive renoprotective effects. Sparsentan reduces blood pressure in hypertensive patients [28]. The antiproteinuric and the possible nephroprotective effects of sparsentan, are currently studied in focal segmental glomerulosclerosis (DUPLEX study) [29] and IgA Nephropathy (PROTECT study; NCT03762850) patients in an ongoing phase 3 clinical trials.

4. Mechanisms of Renal Protection Mediated by ERAs

ET, through the activation of its receptors, may be detrimental for the kidney, as it is involved in the progression of chronic kidney disease and other conditions such as diabetes [48]. Therefore, blockade of the ET receptors with ERAs has renal protective effects. ERAs protect the kidney by several mechanisms. First, this drug class has clear effects on glomerular hemodynamics [49,50,51]. ETA receptor antagonism improves blood pressure via vasodilatation and decreases proteinuria and the filtration fraction (ratio of glomerular filtration rate over renal plasma flow), providing renoprotective effects [14]. Moreover, ETA receptor blockade may improve endothelium-dependent relaxation and vasomotion [52,53,54]. There is no difference in terms of blood pressure reduction when comparing selective ETA receptor antagonists and mixed ETA/ETB receptors antagonists, which suggests that ETB receptor blockade does not change blood pressure. This also implies that combined ETA/ETB receptors antagonists and selective ETA receptor antagonists are similar in terms of their hypotensive effects and ETB receptor antagonists are not involved in this outcome [55]. Second, ERAs also produce effects on different renal cell types that express ET-1 or its receptors [56,57,58]. Podocytes are targets of ET-1 since they express ETA [59]. In this sense, several studies have been focused on the effects of ETA receptor antagonists on these cells. After treatment with ERAs, many studies have found a reduction in the podocyte injury, which lead to the stabilization of the glomerular and podocyte structure [60,61]. Exogenous ET-1 administration induced podocyte injury in rats, which could be prevented by ETA receptor blockade [61]. Also, in a hypertensive rat model, selective ETA receptor blockade restored podocyte injury and function. Further, ERAs ameliorate the structure of the glomerular basement membrane and have beneficial effects on glomerulosclerosis and proteinuria [62]. Mesangial cells produce ET-1, although in a much smaller proportion than endothelial cells. ET-1 produced by mesangial cells can act in an autocrine way by binding to ET receptors. Via ETA it results in the contraction of mesangial cells, cell proliferation and mesangial matrix accumulation [63,64]. These deleterious effects can be blocked using ERAs [65,66]. As mentioned before, ETB is expressed in all along the renal tubule but ETA expression in the proximal tubule and the descending Henle’s loop is low [56]. ETB receptor is responsible for the clearance of ET-1 and could have important implications since it modulates the presence of this vasoconstrictor [51]. Some studies reported that treatment with an ETB-selective receptor antagonist diminished ET-1 clearance, remaining in the plasma, and increasing the response to ET-1 leading to hypertension in some patients [67]. In addition to these effects, ET-1 can induce inflammation and fibrosis [16], since overexpression of ET-1 resulted in interstitial fibrosis in transgenic mice expressing human ET-1 [68] that can be reversed only by ETA-selective receptor antagonists [69].
In summary, ETA-selective ERAs show a wide range of renoprotective effects especially by ameliorating blood pressure and modulating kidney hemodynamics although, as mentioned, ERAs have beneficial effects not mediated by its blood pressure-lowering capacity. ERAs can restore podocyte injury and its function; improve mesangial matrix accumulation, inflammation, and fibrosis, eventually reducing glomerular permeability and proteinuria.

5. Preclinical Experimental Evidence of ERAs Protective Effects on Kidney Damage

In recent years, several preclinical studies have investigated the effects of ERAs using different experimental models. The first studies on cultured mesangial cells showed that ET produced cellular contraction, hypertrophy, and extracellular matrix production [70,71]. These effects were reversed using ERAs [66], as happened in hypertensive rats [72]. Experiments using stroke-prone spontaneously hypertensive rats (SHRSP) demonstrated that the ETA receptor blockade provided renal protection by normalizing the expression of growth factors, diminishing extracellular matrix proteins, and reducing metalloproteinase-2 (MMP-2) activity [66]. Spires et al. studied the effect of atrasentan (an ETA receptor antagonist, Table 2) in streptozotocin-treated Dahl salt-sensitive (STZ-SS) and type 2 diabetic (T2DN) rats. Both rat models showed increased levels of ET-1 during the progression of the renal disease. Atrasentan diminished glomerular injury and renal fibrosis in both models but only reduced arterial pressure and proteinuria in STZ-SS. This could be explained by differences in the severity of the kidney injury of these models [73]. In any case, it illustrates that improvement of kidney damage is possible without changing arterial pressure and/or proteinuria. In this sense, Harvey et al. [74], demonstrated that ETA blocking with atrasentan (but not ETB blocking) improved integrity and viability of cultured podocytes submitted to hypoxia to mimic chronic renovascular disease [74]. In this line, Dolinina et al. tested BQ-788 (an ETB receptor antagonist, Table 2) and JKC-301 (an ETA receptor antagonist) in Sprague-Dawley rats where glomerular permeability was induced by administration of ET-1. The study demonstrated that the glomerular hyperfiltration improvement was dependent on ETA receptors, since ETA receptor blockade ameliorated glomerular hyperfiltration but not ETB receptor blockade [75]. Similar results were obtained in a study where the effects of BQ-788 and atrasentan were compared in uninephrectomized Sprague-Dawley rats on high-sodium diet (HS/UNX) and in spontaneously hypertensive rats (SHR). Both hypertensive rat models showed altered nitric oxide levels, possibly related to ETA receptor hyperactivity. Also, in the HS/UNX model ETA receptor blockade reduced blood pressure and decreased renal excretion, while ETB receptor blockade did not alter blood pressure or renal excretion. The SHR model showed a reduction in blood pressure after treatment with atrasentan. This comparison confirmed, as well, the dependence of blood pressure and renal hemodynamics on ETA receptors, since ETB receptor antagonist did not modify renal hemodynamics [76]. Indeed, ETA selective ERAs have clear renoprotective effects. In this sense sitaxentan (an ETA receptor antagonist, Table 2) improved kidney function and tubular atrophy in a rat model of chronic interstitial nephritis induced by adenine. Further, sitaxentan in combination with cinacalcet (an allosteric modulator of the calcium sensing receptor), increased renal angiotensin converting enzyme 2 (ACE2) expression, which is protective for the kidney, and normalized urinary calcium loss [77]. Similar results were obtained in the study of Caires et al. who tested bosentan and macitentan (Table 2) in normotensive and hypertensive rats with cyclosporin A (CsA)-induced kidney damage. CsA is nephrotoxic and has a vasoconstrictive effect, which was partially reversed only by bosentan. However, both, bosentan and macitentan were able to improve the hemodynamic changes induced by CsA in hypertensive rats by decreasing blood pressure. Furthermore, bosentan and macitentan reduced the generation of reactive oxygen species produced by CsA. Thus, the ERAs used in these experiments had similar effects although bosentan seemed to be better at reversing the hemodynamic changes [78]. Ambrisentan (an ETA receptor antagonist, Table 2) and bosentan showed similar kidney protective effects in an ischemia-reperfusion rat model in terms of reduction of kidney apoptosis, tissue damage and inflammation probably mediated by an increase of nitric oxide levels. Another study using the normotensive Wistar Kyoto (WKY) rats compared the effects of macitentan and sitaxentan. The results of these experiments revealed that sitaxentan prevented sunitinib-induced hypertension in the same manner as macitentan indicating that the increase in blood pressure was mediated by ETA receptors. Sitaxentan also improved albuminuria and diminished prostacyclin levels [79].
The effects of ERAs have also been studied on top of RAS blockers, the current standard of care for many chronic kidney diseases [80,81,82]. Atrasentan combined with losartan (an angiotensin-II type 1 receptor blocker) improved podocyte number and structure and decreased proteinuria in BTBR ob/ob mice [83] similarly to what happens in human [19]. Other studies combined two RAS blockers (trandolapril and losartan) with atrasentan in a rat model of chronic kidney disease, showing an additional beneficial effect of the combination of ERAs with RAS blockers. This combination of drugs increased the survival rate and reduced proteinuria and renal glomerular damage [84]. Also, Gagliardini et al., combined avosentan (an ETA receptor antagonist, Table 2) with lisinopril (an angiotensin converting enzyme inhibitor) in uninephrectomized streptozotocin-induced diabetic rats. Combined therapy was able to improve proteinuria, protected from glomerular and tubulointerstitial damage, restored podocyte number, nephrin levels and glomerular permeability. The combination also improved the deleterious changes in the peritubular capillaries and renal interstitial blood perfusion, which could lead to amelioration of the tubular function [85]. Preclinical studies in mice and rats using sparsentan, a new dual AT1/ETA receptor antagonist with affinity to ETA and angiotensin II (type 1) receptors (Table 2), showed that this dual inhibition protects the glomeruli from podocyte loss and podocyte foot effacement. The effects also include maintenance of the glomerulus basement membrane, glomerular glycocalyx integrity and reduction of blood pressure [86,87]. In addition, studies using multiphoton microscopy imaging in Confetti mice with focal segmental glomerulosclerosis induced by transient receptor potential channel 6 (TRPC6) overexpression, showed greater preservation of the kidney function in mice treated with sparsentan in comparison with the mice that received no drug or losartan [88]. ERAs have also been studied in combination with sodium-glucose type 2 cotransporter (SGLT2) inhibitors (SGLT2i) in several diabetic mice models because of the potential for SGLT2i to reduce the volume overload induced by ERAs. Atrasentan combined with dapagliflozin (an SGLT2i) did not improve albuminuria, glomerular filtration rate, kidney inflammation or fibrosis but ameliorated of glomerulosclerosis and podocyte injury in a mouse model of type 2 diabetic kidney disease. This suggests that the dual therapy approach can have therapeutic potential [89]. A recent study of Vergara, A. et al. [90] tested the capacity of an SGLT2 inhibitor (empagliflozin) and/or an ERA (atrasentan) on top of RAS blockade with ramipril to protect the diabetic kidney in experimental diabetic nephropathy using db/db mice. This study revealed that triple therapy with empagliflozin, atrasentan and ramipril maintained the impact of each therapy alone and added to organ protection. Empagliflozin combined with ramipril or in triple therapy with atrasentan ameliorated hyperfiltration, but only the triple combination exerted greater protection against podocyte loss. The combined therapy not only protected against kidney injury but also provided cardiac protection in terms of decrease of cardiomyocyte hypertrophy. Additionally, the add-on triple therapy further enhanced the intrarenal ACE2/Angiotensin(1-7)/Mas protective arm of the RAS. These data suggest that triple therapy with empagliflozin, atrasentan and ramipril have a synergistic cardiorenal protective effects in experimental diabetic nephropathy. Thus, the combination with RAS blockers and/or SGLT2i may promote the use of the ERAs in the clinical practice as it has shown add-on effects in experimental models and has the potential to mitigate adverse events produced by ERAs in monotherapy. This therapeutic approach is currently being evaluated in randomized controlled trials.

6. Randomized Controlled Trials (RCTs) Using ERAs for Prevention of Kidney Disease Progression

The largest trials testing ERAs have been performed in type 2 diabetic patients (Table 3). In these studies, ERAs have shown to reduce albuminuria and slightly decrease blood pressure [19,23]. The effect of selective endothelin antagonist on albuminuria is consistent across different studies, obtaining a 30–40% reduction on urine albumin-to-creatinine ratio (UACR) in the groups that received the active treatment. However, blood pressure reduction is moderate and shows different results between RCTs. Overall, selective ERAs seem to reduce 3–5 mmHg both systolic and diastolic blood pressure (SBP and DBP, respectively). The effects on BP vary among the employed ERA, with the greatest reductions described for darusentan (9.9 mmHg reduction in SBP and 4.6 mmHg reduction in DBP) [91]. Nevertheless, the latter study included patients with resistant hypertension, which may have contributed to the larger differences in the active treatment arms [91]. In addition, the SONAR study showed that BP reduction is more evident when initiating the treatment and becomes milder after chronic treatment [19].
Regarding GFR preservation, selective ERAs have displayed protective effects or no effect among the different RCTs performed to date. The SONAR trial, which treated responder patients (patients that showed a decrease in UACR of at least 30% with no substantial fluid retention during the enrichment period) for a median follow-up of 2.2 years, showed that 0.75 mg of atrasentan on top of the RAS blockade was able to preserve 0.65 mL/min/1.73 m2 of GFR and to prevent the doubling of serum creatinine during the treatment period [19]. In the same line, in patients with systemic sclerosis, zibotentan was able to preserve 4.3 mL/min/1.73 m2 of GFR after 6.5 months of treatment [24]. The only ERA that showed a significant decrease in GFR that could be related to the type of patients included and the greater BP reduction was darusentan [91].
When analyzing major renal events, only ASCEND and SONAR trials were designed to find differences in a primary composite kidney outcome [19,36]. The ASCEND trial had to be prematurely stopped because an increased number of deaths due to cardiovascular causes in the group of patients receiving the active treatment [36]. As death was included within the main composite outcome, the study was unable to find significant differences between the groups receiving avosentan and the group receiving placebo. The increased number of CV deaths was also linked to an increased number of adverse events: fluid overload, heart failure and anemia. However, if we only consider end-stage renal disease (ESRD) or doubling of serum creatinine as events, the group of patients receiving avosentan showed a lower risk compared to those treated with placebo (HR 0.63, 95%CI: 0.42–0.95). To overcome the evident adverse events related to the inhibition of ETA receptor-mediated sodium and water excretion, the SONAR trial only included responder patients that did not show adverse events during an initial enrichment period [19]. In the latter study, atrasentan was able to reduce the number of renal events when compared to placebo. Nevertheless, previously described adverse events such as fluid overload, heart failure or anemia were again more frequent in the group treated with atrasentan. In this line, the addition of the new renoprotective SGLT2i to the treatment with ERAs in type 2 diabetic patients could prevent the development of fluid retention or anemia, as the former drug class has diuretic effects and increases hemoglobin levels [94,95]. A recent post-hoc analysis of patients receiving atrasentan and SGLT2i in the SONAR trial revealed that weight increase (a surrogate marker of fluid overload) was reduced in patients receiving both atrasentan and SGLT2i [96]. New trials with prespecified kidney outcomes that evaluate the synergistic effects of the combination will shed light upon the future of ERAs in the treatment of chronic kidney disease. The currently ongoing ZENITH-CKD trial (NCT04724837), for example, will evaluate the efficacy of the combination of zibotentan and dapagliflozin in the treatment of CKD.
Additionally, the use of ERAs is being extended to kidney diseases with albuminuria such as primary FSGS or IgA nephropathy, where the existence of previous cardiovascular comorbidities is less frequent and the risk of adverse events also lower. Sparsentan, a dual angiotensin II type 1 and endothelin type A (ETA) receptor antagonist, was already tested in the DUET trial that included patients with primary focal segmental glomerulosclerosis (FSGS) [97]. After eight weeks of treatment, sparsentan obtained greater reductions in proteinuria, and was superior to irbesartan, achieving partial remission of the disease (28% vs. 9%). The promising effects of sparsentan will be further confirmed by the ongoing trials on primary FSGS (DUPLEX study, NCT03493685) and IgA nephropathy (PROTECT and SPARTAN studies, NCT03762850 and NCT04663204, respectively). The ALIGN (NCT04573478) will also give insights about the impact of the combination of atrasentan and RAS blockade for the treatment of IgA nephropathy.

7. Conclusions

As reviewed here, the concentration of ET-1 is increased in pathological conditions, such as diabetes or hypertension, causing sustained vasoconstriction that ultimately leads to kidney damage. The ERAs show clear renoprotective effects in preclinical experimental models and in human mainly by hemodynamic effects but also by restoring podocyte injury, reducing mesangial matrix accumulation, fibrosis and inflammation which reduces glomerular permeability and proteinuria. However, the use of ERAs in clinical practice to prevent kidney disease is narrow because some ERAs failed to demonstrate efficacy in phase III randomized clinical trials and/or produced adverse events such as oedemas. To overcome these limitations, the combination of ERAs with SGLT2i have been proposed as well as the use of the dual angiotensin-II type 1/endothelin receptor blockers. The utility of these therapeutic approaches to treat kidney disease is currently being tested in ongoing randomized controlled trials.

Author Contributions

Writing—original draft preparation; I.M.-D., N.M., C.L.-C. and A.V. Writing—review and editing; F.J.Á., P.W.B., A.V., C.J.-C. and M.J.S.; Figure design: A.V. All authors have read and agreed to the published version of the manuscript.

Funding

The authors have funding from Fondo de Investigación Sanitaria-Feder, ISCIII, PI21/01292, RICORS RD21/0005/0031, and Marató TV3 421/C/2020, Marató TV3 759/U/2020, Marató TV3 215/C/2021.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

F.J.Á. and P.W.B. are employees of Travere Therapeutics. A.V. reports personal fees from MUNDIPHARMA, and non-financial support from MUNDIPHARMA, SANOFI and NOVONORDISK outside this work. C.J.C. declares travel support and a research grant from TRAVERE THERAPEUTICS outside this work. M.J.S. reports grants from BOEHRINGER, personal fees from NOVONORDISK, JANSSEN, BOHERINGER ASTRAZENECA, FRESENIUS, MUNDIPHARMA, PFIZER, ICU, GE Healthcare BAYER, TRAVERE THERAPEUTICS and VIFOR, and non-financial support from ELI LILLY and ESTEVE outside this work.

References

  1. Barton, M.; Yanagisawa, M. Endothelin: 20 Years from Discovery to Therapy. Can. J. Physiol. Pharmacol. 2008, 86, 485–498. [Google Scholar] [CrossRef] [PubMed]
  2. Hickey, K.A.; Rubanyi, G.; Paul, R.J.; Highsmith, R.F. Characterization of a Coronary Vasoconstrictor Produced by Cultured Endothelial Cells. Am. J. Physiol. Cell Physiol. 1985, 248, C550–C556. [Google Scholar] [CrossRef] [PubMed]
  3. Yanagisawa, M.; Kurihara, H.; Kimura, S.; Tomobe, Y.; Kobayashi, M.; Mitsui, Y.; Yazaki, Y.; Goto, K.; Masaki, T. A Novel Potent Vasoconstrictor Peptide Produced by Vascular Endothelial Cells. Nature 1988, 332, 411–415. [Google Scholar] [CrossRef] [PubMed]
  4. Karet, F.E.; Davenport, A.P. Localization of Endothelin Peptides in Human Kidney. Kidney Int. 1996, 49, 382–387. [Google Scholar] [CrossRef] [PubMed]
  5. Inoue, A.; Yanagisawa, M.; Kimura, S.; Kasuya, Y.; Miyauchi, T.; Goto, K.; Masaki, T. The Human Endothelin Family: Three Structurally and Pharmacologically Distinct Isopeptides Predicted by Three Separate Genes. Proc. Natl. Acad. Sci. USA 1989, 86, 2863–2867. [Google Scholar] [CrossRef] [PubMed]
  6. Schiffrin, E.L. Role of Endothelin-1 in Hypertension and Vascular Disease. Am. J. Hypertens. 2001, 14, S83–S89. [Google Scholar] [CrossRef] [PubMed]
  7. Maguire, J.J.; Davenport, A.P. Endothelin Receptors and Their Antagonists. Semin. Nephrol. 2015, 35, 125–136. [Google Scholar] [CrossRef] [PubMed]
  8. Edwards, R.M.; Trizna, W.; Ohlstein, E.H. Renal Microvascular Effects of Endothelin. Am. J. Physiol.-Ren. Physiol. 1990, 259, F217–F221. [Google Scholar] [CrossRef]
  9. Schildroth, J.; Rettig-Zimmermann, J.; Kalk, P.; Steege, A.; Fähling, M.; Sendeski, M.; Paliege, A.; Lai, E.Y.; Bachmann, S.; Persson, P.B.; et al. Endothelin Type A and B Receptors in the Control of Afferent and Efferent Arterioles in Mice. Nephrol. Dial. Transplant. 2011, 26, 779–789. [Google Scholar] [CrossRef] [PubMed]
  10. Chow, L.H.; Subramanian, S.; Nuovo, G.J.; Miller, F.; Nord, E.P. Endothelin Receptor MRNA Expression in Renal Medulla Identified by in Situ RT-PCR. Am. J. Physiol.-Ren. Physiol. 1995, 269, F449–F457. [Google Scholar] [CrossRef]
  11. Helal, I.; Fick-Brosnahan, G.M.; Reed-Gitomer, B.; Schrier, R.W. Glomerular Hyperfiltration: Definitions, Mechanisms and Clinical Implications. Nat. Rev. Nephrol. 2012, 8, 293–300. [Google Scholar]
  12. Tonneijck, L.; Muskiet, M.H.A.; Smits, M.M.; van Bommel, E.J.; Heerspink, H.J.L.; van Raalte, D.H.; Joles, J.A. Glomerular Hyperfiltration in Diabetes: Mechanisms, Clinical Significance, and Treatment. J. Am. Soc. Nephrol. 2017, 28, 1023–1039. [Google Scholar] [CrossRef] [PubMed]
  13. Yang, Y.; Xu, G. Update on Pathogenesis of Glomerular Hyperfiltration in Early Diabetic Kidney Disease. Front. Endocrinol. 2022, 13, 872918. [Google Scholar]
  14. Goddard, J.; Johnston, N.R.; Hand, M.F.; Cumming, A.D.; Rabelink, T.J.; Rankin, A.J.; Webb, D.J. Endothelin-A Receptor Antagonism Reduces Blood Pressure and Increases Renal Blood Flow in Hypertensive Patients with Chronic Renal Failure: A Comparison of Selective and Combined Endothelin Receptor Blockade. Circulation 2004, 109, 1186–1193. [Google Scholar] [CrossRef]
  15. Davenport, A.P.; Maguire, J.J. Endothelin. In The Vascular Endothelium I; Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2006; Volume 176, pp. 295–329. [Google Scholar] [CrossRef]
  16. Saleh, M.A.; Boesen, E.I.; Pollock, J.S.; Savin, V.J.; Pollock, D.M. Endothelin-1 Increases Glomerular Permeability and Inflammation Independent of Blood Pressure in the Rat. Hypertension 2010, 56, 942–949. [Google Scholar] [CrossRef]
  17. Liang, F.; Glascock, C.B.; Schafer, D.L.; Sandoval, J.; Cable, L.A.; Melvin, L.; Hartman, J.C.; Pitts, K.R. Darusentan Is a Potent Inhibitor of Endothelin Signaling and Function in Both Large and Small Arteries. Can. J. Physiol. Pharmacol. 2010, 88, 840–849. [Google Scholar] [CrossRef]
  18. de Zeeuw, D.; Coll, B.; Andress, D.; Brennan, J.J.; Tang, H.; Houser, M.; Correa-Rotter, R.; Kohan, D.; Heerspink, H.J.L.; Makino, H.; et al. The Endothelin Antagonist Atrasentan Lowers Residual Albuminuria in Patients with Type 2 Diabetic Nephropathy. J. Am. Soc. Nephrol. 2014, 25, 1083–1093. [Google Scholar] [CrossRef]
  19. Heerspink, H.J.L.; Parving, H.H.; Andress, D.L.; Bakris, G.; Correa-Rotter, R.; Hou, F.F.; Kitzman, D.W.; Kohan, D.; Makino, H.; McMurray, J.J.V.; et al. Atrasentan and Renal Events in Patients with Type 2 Diabetes and Chronic Kidney Disease (SONAR): A Double-Blind, Randomised, Placebo-Controlled Trial. Lancet 2019, 393, 1937–1947. [Google Scholar] [CrossRef] [PubMed]
  20. Davenport, A.P.; Hyndman, K.A.; Dhaun, N.; Southan, C.; Kohan, D.E.; Pollock, J.S.; Pollock, D.M.; Webb, D.J.; Maguire, J.J. Endothelin. Pharmacol. Rev. 2016, 68, 357–418. [Google Scholar] [CrossRef] [PubMed]
  21. Scott, L.J. Sitaxentan: In Pulmonary Arterial Hypertension. Drugs 2007, 67, 761–770. [Google Scholar] [CrossRef]
  22. Enevoldsen, F.C.; Sahana, J.; Wehland, M.; Grimm, D.; Infanger, M.; Krüger, M. Endothelin Receptor Antagonists: Status Quo and Future Perspectives for Targeted Therapy. J. Clin. Med. 2020, 9, 824. [Google Scholar] [PubMed] [Green Version]
  23. Wenzel, R.R.; Littke, T.; Kuranoff, S.; Jürgens, C.; Bruck, H.; Ritz, E.; Philipp, T.; Mitchell, A. Avosentan Reduces Albumin Excretion in Diabetics with Macroalbuminuria. J. Am. Soc. Nephrol. 2009, 20, 655–664. [Google Scholar] [CrossRef] [PubMed]
  24. Stern, E.P.; Host, L.v.; Wanjiku, I.; Escott, K.J.; Gilmour, P.S.; Ochiel, R.; Unwin, R.; Burns, A.; Ong, V.H.; Cadiou, H.; et al. Zibotentan in Systemic Sclerosis-Associated Chronic Kidney Disease: A Phase II Randomised Placebo-Controlled Trial. Arthritis Res. Ther. 2022, 24, 130. [Google Scholar] [CrossRef] [PubMed]
  25. Wang, Y.; Chen, S.; Du, J. Bosentan for Treatment of Pediatric Idiopathic Pulmonary Arterial Hypertension: State-of-the-Art. Front. Pediatr. 2019, 7, 302. [Google Scholar] [CrossRef] [PubMed]
  26. Mekuria, A.B.; Demelash Kifle, Z.; Abdelwuhab, M. Endothelin System and Therapeutic Application of Endothelin Receptor Antagonists. J. Clin. Exp. Pharmacol. 2021, 10, 272. [Google Scholar]
  27. Schlaich, M.P.; Bellet, M.; Weber, M.A.; Danaietash, P.; Bakris, G.L.; Flack, J.M.; Dreier, R.F.; Sassi-Sayadi, M.; Haskell, L.P.; Narkiewicz, K.; et al. Dual Endothelin Antagonist Aprocitentan for Resistant Hypertension (PRECISION): A Multicentre, Blinded, Randomised, Parallel-Group, Phase 3 Trial. Lancet 2022, 400, 1927–1937. [Google Scholar] [CrossRef] [PubMed]
  28. Murugesan, N.; Gu, Z.; Fadnis, L.; Tellew, J.E.; Baska, R.A.F.; Yang, Y.; Beyer, S.M.; Monshizadegan, H.; Dickinson, K.E.; Valentine, M.T.; et al. Dual Angiotensin II and Endothelin A Receptor Antagonists: Synthesis of 2′-Substituted N-3-Isoxazolyl Biphenylsulfonamides with Improved Potency and Pharmacokinetics. J. Med. Chem. 2005, 48, 171–179. [Google Scholar] [CrossRef]
  29. Komers, R.; Diva, U.; Inrig, J.K.; Loewen, A.; Trachtman, H.; Rote, W.E. Study Design of the Phase 3 Sparsentan Versus Irbesartan (DUPLEX) Study in Patients With Focal Segmental Glomerulosclerosis. Kidney Int. Rep. 2020, 5, 494–502. [Google Scholar] [CrossRef] [PubMed]
  30. Siramshetty, V.B.; Grishagin, I.; Nguyen, D.T.; Peryea, T.; Skovpen, Y.; Stroganov, O.; Katzel, D.; Sheils, T.; Jadhav, A.; Mathé, E.A.; et al. NCATS Inxight Drugs: A Comprehensive and Curated Portal for Translational Research. Nucleic Acids Res. 2022, 50, D1307–D1316. [Google Scholar] [CrossRef] [PubMed]
  31. Kohan, D.E.; Pollock, D.M. Endothelin Antagonists for Diabetic and Non-Diabetic Chronic Kidney Disease. Br. J. Clin. Pharmacol. 2013, 76, 573–579. [Google Scholar] [CrossRef]
  32. Enseleit, F.; Lüscher, T.F.; Ruschitzka, F. Darusentan, a Selective Endothelin A Receptor Antagonist, for the Oral Treatment of Resistant Hypertension. Ther. Adv. Cardiovasc. Dis. 2010, 4, 231–240. [Google Scholar] [CrossRef]
  33. Riechers, H.; Albrecht, H.-P.; Amberg, W.; Baumann, E.; Bernard, H.; Böhm, H.-J.; Klinge, D.; Kling, A.; Müller, S.; Raschack, M.; et al. Expedited Articles Discovery and Optimization of a Novel Class. of Orally Active Nonpeptidic Endothelin-A Receptor Antagonists. J. Med. Chem. 1996, 39, 2123–2128. [Google Scholar] [CrossRef]
  34. Rivera-Lebron, B.N.; Risbano, M.G. Ambrisentan: A Review of Its Use in Pulmonary Arterial Hypertension. Ther. Adv. Respir. Dis. 2017, 11, 233–244. [Google Scholar]
  35. Zhao, Q.; Guo, N.; Chen, J.; Parks, D.; Tian, Z. Comparative Assessment of Efficacy and Safety of Ambrisentan and Bosentan in Patients with Pulmonary Arterial Hypertension: A Meta-Analysis. J. Clin. Pharm. Ther. 2022, 47, 146–156. [Google Scholar]
  36. Mann, J.F.E.; Green, D.; Jamerson, K.; Ruilope, L.M.; Kuranoff, S.J.; Littke, T.; Viberti, G. Avosentan for Overt Diabetic Nephropathy. J. Am. Soc. Nephrol. 2010, 21, 527–535. [Google Scholar] [CrossRef]
  37. Patel, T.; McKeage, K. Macitentan: First Global Approval. Drugs 2014, 74, 127–133. [Google Scholar] [CrossRef]
  38. Sidharta, P.N.; Krähenbühl, S.; Dingemanse, J. Pharmacokinetic and Pharmacodynamic Evaluation of Macitentan, a Novel Endothelin Receptor Antagonist for the Treatment of Pulmonary Arterial Hypertension. Expert Opin. Drug Metab. Toxicol. 2015, 11, 437–449. [Google Scholar] [CrossRef]
  39. Anguiano, L.; Riera, M.; Pascual, J.; Soler, M.J. Endothelin Blockade in Diabetic Kidney Disease. J. Clin. Med. 2015, 4, 1171–1192. [Google Scholar]
  40. Zhou, Y.; Chi, J.; Huang, Y.; Dong, B.; Lv, W.; Wang, Y.G. Efficacy and Safety of Endothelin Receptor Antagonists in Type 2 Diabetic Kidney Disease: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Diabet. Med. 2021, 38, e14411. [Google Scholar]
  41. Chavez, E.; Rodriguez, J.; Drexler, Y.; Fornoni, A. Novel Therapies for Alport Syndrome. Front. Med. 2022, 9, 848389. [Google Scholar] [CrossRef]
  42. Morris, C.D.; Rose, A.; Curwen, J.; Hughes, A.M.; Wilson, D.J.; Webb, D.J. Specific Inhibition of the Endothelin A Receptor with ZD4054: Clinical and Pre-Clinical Evidence. Br. J. Cancer 2005, 92, 2148–2152. [Google Scholar] [CrossRef]
  43. Breu, V.; Clozel, M.; Burri, K.; Hirth, G.; Neidhart, W.; Ramuz, H. In Vitro Characterisation of Ro 46-8443, the First Non-Pep Tide Antagonist Selective for the Endothelin ETB Receptor. FEBS Lett. 1996, 383, 37–41. [Google Scholar] [CrossRef]
  44. Persson, B.P.; Rossi, P.; Weitzberg, E.; Oldner, A. Inhaled Tezosentan Reduces Pulmonary Hypertension in Endotoxin-Induced Lung Injury. Shock 2009, 32, 427–434. [Google Scholar] [CrossRef]
  45. Iglarz, M.; Binkert, C.; Morrison, K.; Fischli, W.; Gatfield, J.; Treiber, A.; Weller, T.; Bolli, M.H.; Boss, C.; Buchmann, S.; et al. Pharmacology of Macitentan, an Orally Active Tissue-Targeting Dual Endothelin Receptor Antagonist. J. Pharmacol. Exp. Ther. 2008, 327, 736–745. [Google Scholar] [CrossRef]
  46. Sidharta, P.N.; Dingemanse, J. Effect of Multiple-Dose Aprocitentan Administration on the Pharmacokinetics of Midazolam in Healthy Male Subjects. Eur. J. Drug Metab. Pharmacokinet. 2020, 45, 227–234. [Google Scholar] [CrossRef]
  47. Sidharta, P.N.; Melchior, M.; Kankam, M.K.; Dingemanse, J. Single- and Multiple-Dose Tolerability, Safety, Pharmacokinetics, and Pharmacodynamics of the Dual Endothelin Receptor Antagonist Aprocitentan in Healthy Adult and Elderly Subjects. Drug Des. Dev. Ther. 2019, 13, 949–964. [Google Scholar] [CrossRef] [Green Version]
  48. Kohan, D.E.; Barton, M. Endothelin and Endothelin Antagonists in Chronic Kidney Disease. Kidney Int. 2014, 86, 896–904. [Google Scholar] [CrossRef]
  49. Inscho, E.W.; Imig, J.D.; Cook, A.K.; Pollock, D.M. ET A and ET B Receptors Differentially Modulate Afferent and Efferent Arteriolar Responses to Endothelin. Br. J. Pharmacol. 2005, 146, 1019–1026. [Google Scholar] [CrossRef]
  50. Masaki, T.; Kimura, S.; Yanagisawa, M.; Goto, K. Molecular and Cellular Mechanism of Endothelin Regulation: Implications for Vascular Function. Circulation 1991, 84, 1457–1468. [Google Scholar]
  51. Dupuis, J.; Goresky, C.A.; Fournier, A. Pulmonary Clearance of Circulating Endothelin-1 in Dogs in Vivo: Exclusive Role of ET(B) Receptors. J. Appl. Physiol. 1996, 81, 1510–1515. [Google Scholar] [CrossRef]
  52. Barton, M.; Haudenschild, C.C.; D’Uscio, L.V.; Shaw, S.; Münter, K.; Lüscher, T.F. Endothelin ETA Receptor Blockade Restores NO-Mediated Endothelial Function and Inhibits Atherosclerosis in Apolipoprotein E-Deficient Mice. Proc. Natl. Acad. Sci. USA 1998, 95, 14367–14372. [Google Scholar] [CrossRef] [PubMed]
  53. Halcox, J.P.J.; Nour, K.R.A.; Zalos, G.; Quyyumi, A.A. Coronary Vasodilation and Improvement in Endothelial Dysfunction with Endothelin ETA Receptor Blockade. Circ. Res. 2001, 89, 969–976. [Google Scholar] [CrossRef]
  54. D’Uscio, L.V.; Barton, M.; Shaw, S.; Lüscher, T.F. Chronic ETA Receptor Blockade Prevents Endothelial Dysfunction of Small Arteries in Apolipoprotein E-Deficient Mice. Cardiovasc. Res. 2002, 53, 487–495. [Google Scholar] [CrossRef] [PubMed]
  55. Li, J.; Turgeon, A.; Schiffrin, E.L. Effect of Chronic Treatment with Two Different ETA Selective Endothelin Receptor Antagonists on Blood Pressure and Small Artery Structure of Deoxycorticosterone Acetate (DOCA)-Sant Hypertensive Rats. Am. J. Hypertens. 1998, 11, 554–562. [Google Scholar]
  56. Wendel, M.; Knels, L.; Kummer, W.; Koch, T. Distribution of Endothelin Receptor Subtypes ETA and ET B in the Rat Kidney. J. Histochem. Cytochem. 2006, 54, 1193–1203. [Google Scholar] [CrossRef]
  57. Simonson, M.S. Endothelins: Muntifunctional Renal Peptides. Physiol. Rev. 1993, 73, 375–411. [Google Scholar]
  58. Barton, M.; Yanagisawa, M. Endothelin: 30 Years from Discovery to Therapy. Hypertension 2019, 74, 1232–1265. [Google Scholar] [CrossRef]
  59. Rebibou, J.M.; He, C.J.; Delarue, F.; Peraldi, M.N.; Adida, C.; Rondeau, E.; Sraer, J.D. Functional Endothelin 1 Receptors on Human Glomerular Podocytes and Mesangial Cells. Nephrol. Dial. Transplant. 1992, 7, 288–292. [Google Scholar] [CrossRef]
  60. Benigni, A.; Zoja, C.; Corna, D.; Orisio, S.; Longaretti, L.; Bertani, T.; Remuzzi, G. A Specific Endothelin Subtype A Receptor Antagonist Protects against Injury in Renal Disease Progression. Kidney Int. 1993, 44, 440–444. [Google Scholar] [CrossRef] [Green Version]
  61. Ortmann, J.; Amann, K.; Brandes, R.P.; Kretzler, M.; Münter, K.; Parekh, N.; Traupe, T.; Lange, M.; Lattmann, T.; Barton, M. Role of Podocytes for Reversal of Glomerulosclerosis and Proteinuria in the Aging Kidney after Endothelin Inhibition. Hypertension 2004, 44, 974–981. [Google Scholar] [CrossRef]
  62. Opočenský, M.; Kramer, H.J.; Bäcker, A.; Vernerová, Z.; Eis, V.; Červenka, L.; Chábová, V.C.; Tesař, V.; Vaněčková, I. Late-Onset Endothelin-A Receptor Blockade Reduces Podocyte Injury in Homozygous Ren-2 Rats despite Severe Hypertension. Hypertension 2006, 48, 965–971. [Google Scholar] [CrossRef]
  63. Kohan, D.E. Endothelins in the Normal and Diseased Kidney. Am. J. Kidney Dis. 1997, 29, 2–26. [Google Scholar] [CrossRef]
  64. Sorokin, A.; Kohan, D.E. Physiology and Pathology of Endothelin-1 in Renal Mesangium. Am. J. Physiol. Renal Physiol. 2003, 285, F579–F589. [Google Scholar] [CrossRef]
  65. Gómez-Garre, D.; Largo, R.; Liu, X.H.; Gutierrez, S.; López-Armada, M.J.; Palacios, I.; Egido, J. An Orally Active ET(A)/ET(B) Receptor Antagonist Ameliorates Proteinuria and Glomerular Lesions in Rats with Proliferative Nephritis. Kidney Int. 1996, 50, 962–972. [Google Scholar] [CrossRef]
  66. Tostes, R.C.A.; Touyz, R.M.; He, G.; Ammarguellat, F.; Schiffrin, E.L. Endothelin A Receptor Blockade Decreases Expression of Growth Factors and Collagen and Improves Matrix Metalloproteinase-2 Activity in Kidneys from Stroke-Prone Spontaneously Hypertensive Rats. J. Cardiovasc. Pharmacol. 2002, 39, 892–900. [Google Scholar] [CrossRef]
  67. Fukuroda, T.; Fujikawa, T.; Ozaki, S.; Ishikawa, K.; Yano, M.; Nishikibe, M. Clearance of Circulating Endothelin-1 by Etb Receptors in Rats. Biochem. Biophys. Res. Commun. 1994, 199, 1461–1465. [Google Scholar] [CrossRef]
  68. Hocher, B.; Thöne-Reineke, C.; Rohmeiss, P.; Schmager, F.; Slowinski, T.; Burst, V.; Siegmund, F.; Quertermous, T.; Bauer, C.; Neumayer, H.H.; et al. Endothelin-1 Transgenic Mice Develop Glomerulosclerosis, Interstitial Fibrosis, and Renal Cysts but Not Hypertension. J. Clin. Investig. 1997, 99, 1380–1389. [Google Scholar] [CrossRef]
  69. Saleh, M.A.; Boesen, E.I.; Pollock, J.S.; Savin, V.J.; Pollock, D.M. Endothelin Receptor A-Specific Stimulation of Glomerular Inflammation and Injury in a Streptozotocin-Induced Rat Model of Diabetes. Diabetologia 2011, 54, 979–988. [Google Scholar] [CrossRef]
  70. Takeda, M.; Breyer, M.D.; Noland, T.D.; Homma, T.; Hoover, R.L.; Inagami, T.; Kon, V. Endothelin-1 Receptor Antagonist: Effects on Endothelin- and Cyclosporine-Treated Mesangial Cells. Kidney Int. 1992, 41, 1713–1719. [Google Scholar] [CrossRef] [Green Version]
  71. Orth, S.R.; Amann, K.; Gehlen, F.; Unger, L.; Wagner, J.; Raschack, M.; Ritz, E. Adult Human Mesangial Cells (HMCs) Express Endothelin-B-Receptors Which Mediate Endothelin-1-Induced Cell Growth. J. Cardiovasc. Pharmacol. 2000, 36, S232–S237. [Google Scholar] [CrossRef]
  72. Nakamura, T.; Kurashina, T.; Saito, Y.; Sumino, H.; Akuzawa, N.; Aizawa, H.; Sakamoto, H.; Ono, Z.; Nagai, R. ET(A) Receptor Antagonist Ameliorates Nephrosclerosis and Left Ventricular Hypertrophy Induced in Rat by Prolonged Inhibition of Nitric Oxide Synthesis. Hypertens. Res. Clin. Exp. 1998, 21, 251–257. [Google Scholar] [CrossRef]
  73. Spires, D.; Poudel, B.; Shields, C.A.; Pennington, A.; Fizer, B.; Taylor, L.; McPherson, K.C.; Cornelius, D.C.; Williams, J.M. Prevention of the Progression of Renal Injury in Diabetic Rodent Models with Preexisting Renal Disease with Chronic Endothelin A Receptor Blockade. Am. J. Physiol. Renal Physiol. 2018, 315, F977–F985. [Google Scholar] [CrossRef]
  74. Harvey, T.W.; Engel, J.E.; Chade, A.R. Vascular Endothelial Growth Factor and Podocyte Protection in Chronic Hypoxia: Effects of Endothelin-A Receptor Antagonism. Am. J. Nephrol. 2016, 43, 74–84. [Google Scholar] [CrossRef]
  75. Dolinina, J.; Rippe, A.; Öberg, C.M. Sustained, Delayed, and Small Increments in Glomerular Permeability to Macromolecules during Systemic ET-1 Infusion Mediated via the ETa Receptor. Am. J. Physiol. Renal Physiol. 2019, 316, F1173–F1179. [Google Scholar] [CrossRef]
  76. Bądzyńska, B.; Vaneckova, I.; Sadowski, J.; Hojná, S.; Kompanowska-Jezierska, E. Effects of Systemic and Renal Intramedullary Endothelin-1 Receptor Blockade on Tissue NO and Intrarenal Hemodynamics in Normotensive and Hypertensive Rats. Eur. J. Pharmacol. 2021, 910, 174445. [Google Scholar] [CrossRef]
  77. Törmänen, S.; Pörsti, I.; Lakkisto, P.; Tikkanen, I.; Niemelä, O.; Paavonen, T.; Mustonen, J.; Eräranta, A. Endothelin A Receptor Blocker and Calcimimetic in the Adenine Rat Model of Chronic Renal Insufficiency. BMC Nephrol. 2017, 18, 323. [Google Scholar] [CrossRef]
  78. Caires, A.; Fernandes, G.S.; Leme, A.M.; Castino, B.; Pessoa, E.A.; Fernandes, S.M.; Fonseca, C.D.; Vattimo, M.F.; Schor, N.; Borges, F.T. Endothelin-1 Receptor Antagonists Protect the Kidney against the Nephrotoxicity Induced by Cyclosporine-A in Normotensive and Hypertensive Rats. Braz. J. Med. Biol. Res. 2018, 51, e6373. [Google Scholar] [CrossRef]
  79. Colafella, K.M.M.; Neves, K.B.; Montezano, A.C.; Garrelds, I.M.; Van Veghel, R.; De Vries, R.; Uijl, E.; Baelde, H.J.; Van Den Meiracker, A.H.; Touyz, R.M.; et al. Selective ETA vs. Dual ETA/B Receptor Blockade for the Prevention of Sunitinib-Induced Hypertension and Albuminuria InWKY Rats. Cardiovasc. Res. 2020, 116, 1779–1790. [Google Scholar] [CrossRef]
  80. Xie, X.; Liu, Y.; Perkovic, V.; Li, X.; Ninomiya, T.; Hou, W.; Zhao, N.; Liu, L.; Lv, J.; Zhang, H.; et al. Renin-Angiotensin System Inhibitors and Kidney and Cardiovascular Outcomes in Patients with CKD: A Bayesian Network Meta-Analysis of Randomized Clinical Trials. Am. J. Kidney Dis. 2016, 67, 728–741. [Google Scholar] [CrossRef] [Green Version]
  81. Mishima, E.; Haruna, Y.; Arima, H. Renin-Angiotensin System Inhibitors in Hypertensive Adults with Non-Diabetic CKD with or without Proteinuria: A Systematic Review and Meta-Analysis of Randomized Trials. Hypertens. Res. 2019, 42, 469–482. [Google Scholar] [CrossRef]
  82. Fu, E.L.; Clase, C.M.; Evans, M.; Lindholm, B.; Rotmans, J.I.; Dekker, F.W.; van Diepen, M.; Carrero, J.J. Comparative Effectiveness of Renin-Angiotensin System Inhibitors and Calcium Channel Blockers in Individuals With Advanced CKD: A Nationwide Observational Cohort Study. Am. J. Kidney Dis. 2021, 77, 719–729.e1. [Google Scholar] [CrossRef] [PubMed]
  83. Hudkins, K.L.; Wietecha, T.A.; Steegh, F.; Alpers, C.E. Beneficial Effect on Podocyte Number in Experimental Diabetic Nephropathy Resulting from Combined Atrasentan and RAAS Inhibition Therapy. Am. J. Physiol. Renal Physiol. 2020, 318, F1295–F1305. [Google Scholar] [CrossRef] [PubMed]
  84. Čertíková Chábová, V.; Vernerová, Z.; Kujal, P.; Husková, Z.; Škaroupková, P.; Tesař, V.; Kramer, H.J.; Kompanowska-Jezierska, E.; Walkowska, A.; Sadowski, J.; et al. Addition of ET A Receptor Blockade Increases Renoprotection Provided by Renin—Angiotensin System Blockade in 5/6 Nephrectomized Ren-2 Transgenic Rats. Life Sci. 2014, 118, 297–305. [Google Scholar] [CrossRef] [PubMed]
  85. Gagliardini, E.; Corna, D.; Zoja, C.; Sangalli, F.; Carrara, F.; Rossi, M.; Conti, S.; Rottoli, D.; Longaretti, L.; Remuzzi, A.; et al. Unlike Each Drug Alone, Lisinopril If Combined with Avosentan Promotes Regression of Renal Lesions in Experimental Diabetes. Am. J. Physiol. Renal Physiol. 2009, 297, F1448–F1456. [Google Scholar] [CrossRef]
  86. Nagasawa, H.; Suzuki, H.; Jenkinson, C.; Ueda, S.; Fukao, Y.; Nakayama, M.; Otsuka, T.; Okuma, T.; Clapper, W.; Komers, R.; et al. Sparsentan, the Dual Endothelin Angiotensin Receptor Antagonist (Deara), Attenuates Albuminuria and Protects from the Development of Renal Injury to a Greater Extent Than Losartan in the Gddy Mouse Model of Iga Nephropathy: A 16-Week Study. ERA Congr. 2022, 37, gfac067.060. [Google Scholar] [CrossRef]
  87. Bedard, P.; Jenkinson, C.; Komers, R. Sparsentan Protects the Glomerular Basement Membrane and Glycocalyx, and Attenuates Proteinuria in a Rat Model of Focal Segmental Glomerulosclerosis (FSGS). ERA Congr. 2022, 37, gfac067.054. [Google Scholar] [CrossRef]
  88. Gyarmati, G.; Deepak, S.K.; Shroff, U.N.; Izuhara, A.; Komers, R.; Bedard, P.W.; Peti-Peterdi, J. Sparsentan Improves Glomerular Endothelial and Podocyte Functions and Augments Protective Tissue Repair in a Mouse Model of Focal Segmental Glomerulosclerosis (FSGS). In Proceedings of the American Society of Nephrology (ASN) Kidney Week 2022—Session: Glomerular Diseases: From Bench to Bedside, Orlando, FL, USA, 3–6 November 2022. [Google Scholar]
  89. Stuart, D.; Peterson, C.S.; Hu, C.; Revelo, M.P.; Huang, Y.; Kohan, D.E.; Ramkumar, N. Lack of Renoprotective Effects of Targeting the Endothelin A Receptor and (or) Sodium Glucose Transporter 2 in a Mouse Model of Type 2 Diabetic Kidney Disease. Can. J. Physiol. Pharmacol. 2022, 100, 763–771. [Google Scholar] [CrossRef]
  90. Vergara, A.; Jacobs-Cacha, C.; Llorens-Cebria, C.; Ortiz, A.; Martinez-Diaz, I.; Martos, N.; Dominguez-Báez, P.; Van den Bosch, M.M.; Bermejo, S.; Pieper, M.P.; et al. Enhanced Cardiorenal Protective Effects of Combining SGLT2 Inhibition, Endothelin Receptor Antagonism and RAS Blockade in Type 2 Diabetic Mice. Int. J. Mol. Sci. 2022, 23, 12823. [Google Scholar] [CrossRef]
  91. Weber, M.A.; Black, H.; Bakris, G.; Krum, H.; Linas, S.; Weiss, R.; Linseman, J.V.; Wiens, B.L.; Warren, S.; Lindholm, L.H. A Selective Endothelin-Receptor Antagonist to Reduce Blood Pressure in Patients with Treatment-Resistant Hypertension: A Randomised, Double-Blind, Placebo-Controlled Trial. Lancet 2009, 374, 1423–1431. [Google Scholar] [CrossRef]
  92. Kohan, D.E.; Pritchett, Y.; Molitch, M.; Wen, S.; Garimella, T.; Audhya, P.; Andress, D.L. Addition of Atrasentan to Renin-Angiotensin System Blockade Reduces Albuminuria in Diabetic Nephropathy. J. Am. Soc. Nephrol. 2011, 22, 763–772. [Google Scholar] [CrossRef]
  93. Dhaun, N.; MacIntyre, I.M.; Kerr, D.; Melville, V.; Johnston, N.R.; Haughie, S.; Goddard, J.; Webb, D.J. Selective Endothelin-a Receptor Antagonism Reduces Proteinuria, Blood Pressure, and Arterial Stiffness in Chronic Proteinuric Kidney Disease. Hypertension 2011, 57, 772–779. [Google Scholar] [CrossRef]
  94. Griffin, M.; Rao, V.S.; Ivey-Miranda, J.; Fleming, J.; Mahoney, D.; Maulion, C.; Suda, N.; Siwakoti, K.; Ahmad, T.; Jacoby, D.; et al. Empagliflozin in Heart Failure: Diuretic and Cardiorenal Effects. Circulation 2020, 142, 1028–1039. [Google Scholar] [CrossRef] [PubMed]
  95. Oshima, M.; Neuen, B.L.; Jardine, M.J.; Bakris, G.; Edwards, R.; Levin, A.; Mahaffey, K.W.; Neal, B.; Pollock, C.; Rosenthal, N.; et al. Effects of Canagliflozin on Anaemia in Patients with Type 2 Diabetes and Chronic Kidney Disease: A Post-Hoc Analysis from the CREDENCE Trial. Lancet Diabetes Endocrinol. 2020, 8, 903–914. [Google Scholar] [CrossRef] [PubMed]
  96. Heerspink, H.J.L.; Kohan, D.E.; de Zeeuw, D. New Insights from SONAR Indicate Adding Sodium Glucose Co-Transporter 2 Inhibitors to an Endothelin Receptor Antagonist Mitigates Fluid Retention and Enhances Albuminuria Reduction. Kidney Int. 2021, 99, 346–349. [Google Scholar] [CrossRef] [PubMed]
  97. Trachtman, H.; Nelson, P.; Adler, S.; Campbell, K.N.; Chaudhuri, A.; Derebail, V.K.; Gambaro, G.; Gesualdo, L.; Gipson, D.S.; Hogan, J.; et al. DUET: A Phase 2 Study Evaluating the Efficacy and Safety of Sparsentan in Patients with FSGS. J. Am. Soc. Nephrol. 2018, 29, 2745–2754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Scheme of the endothelin system. ET-1 acts thought its binding to ETA and ETB producing opposite effects in the kidney. The effects caused by the activation of ETA are shown in red and the effects of ETB activation are shown in blue. In pathological conditions, the hyperglycaemia, acidosis and the presence of insulin, angiotensin II and proinflammatory cytokines causes the increase of ET-1 concentration, which produces deleterious effects on renal function, such as vasoconstriction and endothelial damage, inflammation, fibrosis, podocyte damage or albuminuria.
Figure 1. Scheme of the endothelin system. ET-1 acts thought its binding to ETA and ETB producing opposite effects in the kidney. The effects caused by the activation of ETA are shown in red and the effects of ETB activation are shown in blue. In pathological conditions, the hyperglycaemia, acidosis and the presence of insulin, angiotensin II and proinflammatory cytokines causes the increase of ET-1 concentration, which produces deleterious effects on renal function, such as vasoconstriction and endothelial damage, inflammation, fibrosis, podocyte damage or albuminuria.
Ijms 24 03427 g001
Table 1. ET1, ET2 and ET3 amino acid sequences. ET is a 21-amino acid peptide present in three different isoforms in the organism: ET-1, ET-2 and ET-3 [5]; the differences in amino acid sequence between them are shown in bold.
Table 1. ET1, ET2 and ET3 amino acid sequences. ET is a 21-amino acid peptide present in three different isoforms in the organism: ET-1, ET-2 and ET-3 [5]; the differences in amino acid sequence between them are shown in bold.
ET IsoformAmino Acid Sequence
ET-1NH2—CSCSSLMDKECVYFCHLDIIW—CH3
ET-2NH2—CSCSSWLDKECVYFCHLDIIW—CH3
ET-3NH2—CTCFTYLDKECVYFCHLDIIW—CH3
Table 2. Main endothelin receptor antagonists (ERAs), the selectivity for the ET receptors, year of development, the current status and the protective effects of each of them.
Table 2. Main endothelin receptor antagonists (ERAs), the selectivity for the ET receptors, year of development, the current status and the protective effects of each of them.
ERAAffinityYear of Development ¥Status ¥Protective EffectsDiseases TestedReferences
BQ-123ETA1995--Potent vasodilatation in the peripheral circulation
Reduction of albumin permeability
--Saleh, M., et al., 2010 [16]
DarusentanETA
~1000-fold
1996InvestigationalInhibition of vasoconstrictionResistant Hypertension
Chronic Heart Failure
Liang, F., et al., 2010 [17]
AtrasentanETA
~1800-fold
1996InvestigationalReduction of albuminuria
Risk of renal events
Diabetic Nephropathyde Zeeuw, D., et al., 2014 [18]
Heerspink, H., et al., 2019 [19]
Davenport A.P. et al., 2016 [20]
SitaxentanETA
~6500-fold
1997--Reduction of tubular atrophy
Influences kidney hemodynamics and improves kidney function
Pulmonary Arterial Hypertension
Chronic Renal Insufficiency
Scott, L.J., et al., 2007 [21]
AmbrisentanETA
>4000-fold
2004Approved in 2007Inhibition of vasoconstriction effectsPulmonary Arterial Hypertension Enevoldsen, F.C., et al., 2020 [22]
MacitentanETA
~1000-fold
--Approved in 2013Stimulate vasodilation,
Marked antitumoral effects in an experimental model
of multidrug-resistant ovarian tumors
Pulmonary Arterial HypertensionDavenport A.P., et al., 2016 [20]
AvosentanETA
~500-fold
2006InvestigationalReduction of albuminuriaDiabetic Nephropathy Wenzel, R.R., et al., 2009 [23]
ZibotentanETA2010InvestigationalImprovement in eGFR
Not evidence of an increment of serum endothelin levels
Reduction of blood pressure
Chronic Kidney Disease
Potential use for Alzheimer Disease
Heart failure, hormone resistant prostate cancer and other cancers
Stern, E.P., et al., 2022 [24]
BQ-788ETB1994--Reduction of albumin permeability in combination with BQ-123--Saleh, M., et al., 2010 [16]
BosentanETA/ETB1999Approved in 2001Decrease vascular resistance
Inhibition of endothelial cells proliferation
Pulmonary Arterial Hypertension Wang, Y., et al., 2019 [25]
TezosentanETA/ETB2001--Decrease serum creatinine
Increases GFR
Maintained renal architecture in kidneys after ischemia
Pulmonary Arterial Hypertension
Acute Heart Failure
Mekuria, et al., 2021 [26]
AprocitentanETA/ETB2015InvestigationalDecrease blood pressureResistant HypertensionSchlaich, M.P., et al., 2022 [27]
Sparsentan ETA *
~1000-fold
2005InvestigationalPromote proteinuria reduction
Nephroprotective effects
Focal Segmental Glomerulosclerosis and IgA NephropathyMurugesan N., et al., 2005 [28]
Komers, R., et al. 2020 [29]
* Dual ETA and angiotensin-II type1 receptor antagonist. ¥ Year of development and status of each drug was searched in “NCATS Inxight Drugs” database in December 2022 [30].
Table 3. Renal effects displayed by selective endothelin receptor antagonists (ERAs) in different randomized controlled trials.
Table 3. Renal effects displayed by selective endothelin receptor antagonists (ERAs) in different randomized controlled trials.
NStudy PopulationDiabetes (%)Baseline GFR (mL/min/1.73 m2)Intervention/Control/
Follow-Up
Kidney
Endpoints
Albuminuria/Proteinuria
Reduction
BP Reduction
(ERA-Placebo)
GFR Difference
(ERA-Placebo)
Author/Study/Year
379GFR > 30 mL/min/1.73 m2.
Resistant hypertension.
153
(40%)
79.0Darusentan 50, 100 or 300 mg/daily
Placebo
3.5 months
NR30.4 mg/g (UACR)−9.9 mmHg in SBP(95%CI: −12.3–−5.7)
−4.6 mmHg in DBP
(95%CI: −7.0–−2.2)
−3.7
(95%CI: −6.9–−0.5)
Weber, M.A. et al. [91]
2009
139221–80 years.
Creatinine 1.2–3 mg/dL.
UACR ≥ 309 mg/g.
Diabetic.
1392
(100%)
33.3Avosentan 25 or 50 mg/daily
Placebo
4 months
HR 0.87
(95%CI 0.6–1.2) a
565.5 mg/g (UACR)
31.7% UACR reduction *
−5.1 mmHg in SBP−3.7 mmHg in DBP0.15
(95%CI: −1.3–1.9)
Mann, J.F.E. et al. [36]
ASCEND 2010
89GFR > 20 mL/min/1.73 m2.
UACR 100–3000 mg/g.
Type 2 diabetes.
89
(100%)
52.8Atrasentan 0.25, 0.75, 1.25 mg/daily
Placebo
2 months
NR27.5% UACR reduction *
Not significant reduction
with 0.25 mg
−8.2 mmHg in SBP−6.6 mmHg in DBPNot significant reduction with 0.25 mgNRKohan, D.E. et al. [92]
2011
2718–70 years.
CKD stages 1 to 4.
Non-diabetic.
0
(0%)
54.0Sitaxsentan 100 mg/daily
Placebo and nifedipine 30 mg/daily
1.5 months
NR0.56 g/day
(24-h proteinuria)
336.3 mg/g (UPCR)
≈−5 mmHg reduction in SBP and DBPNRDhaun, N. et al. [93]
2011
211>18 years.
GFR 30 to 75 mL/min/1.73 m2.
UACR 300–5000 mg/g.
Type 2 diabetes.
211
(100%)
49.3Atrasentan 0.75 or 1.25 mg/daily
Placebo
3 months
NR301.5 mg/g (UACR)0.5 mmHg in SBP
(95%CI: −5.0–6.0)
1 mmHg in DBP
(95%CI: −2.8–4.8)
−0.5
(95%CI: −5.3–4.3)
Zeeuw, D. et al. [18]
RADAR 2014
264818–85 years.
GFR 25 to 75 mL/min/1.73 m2.UACR 300–5000 mg/g.
Type 2 diabetes
Responders (30% decrease in UACR).
2648 (100%)43.9Atrasentan 0.75 mg/daily
Placebo
26.4 months
HR 0.65
(95%CI 0.5–0.9) b
33.6% UACR reduction *
(95%CI: 29.1–38.2)
−1.6 mmHg SBP reduction
(95%CI: 0.7–2.5)
0.65
(95%CI: 0.3–1.0)
Heerspink, et al. [19]
SONAR 2019
13>18 years.
Systemic sclerosis.
CKD stages 2 to 3a.
0
(0%)
52.4Zibotentan 10 mg/daily
Placebo
6.5 months
NRNRNR4.3
(95%CI: 2.6–11.3)
Stern, et al. [24]
ZEBRA 1 2022
a Doubling of serum creatinine, end-stage kidney disease or death. b Doubling of serum creatinine, end-stage kidney disease or death due to kidney failure. * UACR percentage reduction compared to place (the reduction observed in the placebo group has been subtracted to the reduction in the active treatment groups). BP: Blood Pressure; SBP: systolic blood pressure; DBP: Diastolic blood Pressure; CKD: Chronic Kidney Disease; GFR: Glomerular Filtration Rate; UACR: urine albumin-to-creatinine ratio; UPCR: urine protein-to-creatinine ratio; NR: not reported.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Martínez-Díaz, I.; Martos, N.; Llorens-Cebrià, C.; Álvarez, F.J.; Bedard, P.W.; Vergara, A.; Jacobs-Cachá, C.; Soler, M.J. Endothelin Receptor Antagonists in Kidney Disease. Int. J. Mol. Sci. 2023, 24, 3427. https://doi.org/10.3390/ijms24043427

AMA Style

Martínez-Díaz I, Martos N, Llorens-Cebrià C, Álvarez FJ, Bedard PW, Vergara A, Jacobs-Cachá C, Soler MJ. Endothelin Receptor Antagonists in Kidney Disease. International Journal of Molecular Sciences. 2023; 24(4):3427. https://doi.org/10.3390/ijms24043427

Chicago/Turabian Style

Martínez-Díaz, Irene, Nerea Martos, Carmen Llorens-Cebrià, Francisco J. Álvarez, Patricia W. Bedard, Ander Vergara, Conxita Jacobs-Cachá, and Maria José Soler. 2023. "Endothelin Receptor Antagonists in Kidney Disease" International Journal of Molecular Sciences 24, no. 4: 3427. https://doi.org/10.3390/ijms24043427

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop