Clinical Trials Results for Endothelin System: Pathophysiological role in Chronic Heart Failure, Acute Coronary Syndromes and MI – Marker of Disease Severity or Genetic Determination?
Curator: Aviva Lev-Ari, PhD, RN
UPDATED on 8/17/2018
Ambrisentan (U.S. trade name Letairis; E.U. trade name Volibris; India trade name Pulmonext by MSN labs) is a drug indicated for use in the treatment of pulmonary hypertension.
The peptide endothelin constricts muscles in blood vessels, increasing blood pressure. Ambrisentan, which relaxes those muscles, is an endothelin receptor antagonist, and is selective for the type A endothelin receptor (ETA).[1] Ambrisentan significantly improved exercise capacity (6-minute walk distance) compared with placebo in two double-blind, multicenter trials (ARIES-1 and ARIES-2).[2]
Ambrisentan was approved by the U.S. Food and Drug Administration (FDA) and European Medicines Agency, and designated an orphan drug, for the treatment of pulmonary hypertension.[3][4][5][6][7]
Ambrisentan is an endothelin receptor antagonist used in the therapy of pulmonary arterial hypertension (PAH). Ambrisentan has been associated with a low rate of serum enzyme elevations during therapy, but has yet to be implicated in cases of clinically apparent acute liver injury.
Ambrisentan was first approved by the U.S. Food and Drug Administration (FDA) on Jun 15, 2007, then approved by the European Medicines Agency (EMA) on Apr 21, 2008 and approved by Pharmaceuticals and Medical Devices Agency of Japan (PMDA) on Jul 23, 2010. In 2000, Abbott, originator of ambrisentan, granted Myogen (acquired by Gilead in 2006) a license to the compound for the treatment of PAH. In 2006, GlaxoSmithKline obtained worldwide rights to market the compound for PAH worldwide, with the exception of the U.S. It is marketed as Letairis® by Gilead in US.
Ambrisentan is an endothelin receptor antagonist, and is selective for the type A endothelin receptor (ETA). It is indicated for the treatment of pulmonary arterial hypertension (PAH) (WHO Group 1) to improve exercise ability and delay clinical worsening. Studies establishing effectiveness included predominantly patients with WHO Functional Class II-III symptoms and etiologies of idiopathic or heritable PAH (64%) or PAH associated with connective tissue diseases (32%).
Letairis® is available as film-coated tablet for oral use, containing 5 or 10 mg of free Ambrisentan. The recommended starting dose is 5 mg once daily with or without food, and increase the dose to 10 mg once daily if 5 mg is tolerated.
Introduction to Endothelin
Endothelin (ET) derived from vascular endothelial cells (ECs), which consists of a 21 amino acid peptide, has a strong and persistent vasoconstrictive action (1). ET has three family peptides (ET-1, ET-2, and ET-3). As the distribution and properties of these peptides are different, each peptide is believed to play specific physiological roles. ET has two types of receptor: the ETA receptor with a high affinity for ET-1 and ET-2 is mainly located on muscle cells, whereas the ETB receptor with an affinity for all three peptides lies on endothelial, epithelial, endocrine, and nerve cells. Of the three ET isoforms,
ET-1 plays a much more important role in the regulation of vascular tone than the others and has a powerful effect on the cardiovascular system. Thus, the role of ET-1 and its receptors as the etiology or precipitating factors in various cardiovascular diseases (CVD) has been investigated (2, 3). In addition, numerous studies have reported effective treatment targeted at ET-1 in pulmonary hypertension, salt-sensitive hypertension, diabetes, and acute and chronic kidney diseases using ETconverting enzyme (ECE) inhibitors and ET-receptor antagonists (2, 4). Several animal models genetically lacking ET-1 and ET receptors have also been used as a tool for determining the physiological and pathophysiological roles of ET-1 and ET receptors in CVD (5 – 10).
Fig. 1.
Schematic illustration of ET-1 production and ET receptor–mediated actions on vascular endothelial cells and smooth muscle cells. G: G protein, ROS: reactive oxygen species, CaM: calmodulin, AA: arachidonic acid, PGI2: prostaglandin I2, AC: adenylate cyclase, sGC: soluble guanylate cyclase.
Figure Source: Journal of Pharmacological Sciences, 119, 302 – 313 (2012)
Introduction to the ET system
Endothelial Cells (ECs) are known as the main physiological source of vascular ET-1. Vascular smooth muscle cells (VSMCs), macrophages, leukocytes, cardiomyocytes, and fibroblasts are also capable of ET-1 production (11 – 13).
Several studies have indicated that various physical and chemical factors such as thrombin, angiotensin II, cytokines, hypoxia, and shear stress stimulate ET-1 gene expression in ECs by DNA binding of transcription factors including activator protein-1, GATA-2, Smad, nuclear factor-kappa B (NF-κB), and hypoxia inducible factor-1 (14 – 18). On the other hand, ET-1 is synthesized as an inactive 203-amino-acid precursor, preproET-1, which is proteolytically cleaved to yield a second inactive 39 (or 38)-amino-acid segment called ‘big’ ET-1.
The last part of the proteolytic process is mainly carried out by ECE (ECE-1 and ECE-2) and leads to the production of the bioactive form of 21-amino-acid peptide ET-1. As ET-1 release from ECs is constitutive, ET-1 biosynthesis and release appear to be mainly controlled via regulation of gene transcription and/or ECE activity.
On the other hand, although another ETB-receptor subtype (ETB2) located on VSMCs exerts vasoconstriction, it has become clear that ETB2 receptor–induced vasoconstriction is negligible under normal conditions but becomes more important in some kinds of diseases such as atherosclerosis and essential hypertension (24 – 26).
- inhibit generation of ET-1 (Jeng et al., 2002) and
- block the action of ET-1 (Battistini et al., 2006)
- selective antagonists in man (Haynes and Webb, 1994) and from
- knockout and transgenic animals
The recent licensing of
- bosentan
- sitaxsentan and
- ambrisentan
for treatment of PAH is the most obvious demonstration of the clinical benefit derived from therapeutic manipulation of the ET-1 system in cardiovascular disease. This development of one of the first effective treatments for a condition with poor prognosis has obvious clinical significance and is likely to be extended to include PAH associated with connective tissue disorders.
Thus, ET antagonists are already realizing their potential in treatment of cardiovascular diseases, while early clinical data suggest these compounds may prove beneficial in other conditions, such as resistant hypertension, chronic kidney disease and SAH. In contrast, a potential role in conditions associated with vascular remodelling (restenosis, chronic obstructive pulmonary disease and transplant graft rejection) remains speculative and requires further investigation. It should also be noted that the clinical experience with ET antagonists in patients with cardiovascular disease remains relatively limited and the design of new trials could be improved using knowledge gained from previous studies, particularly with regard to drug dose and selectivity. These successes must obviously be balanced against the failure of ET antagonists to realize their potential in the treatment of heart failure, and the fact that teratogenic effects have restricted their possible use to treatment of conditions where childbearing potential is unlikely to be an issue.
Several reasons have been proposed to account for the disappointing outcomes in clinical trials as compared to investigations using animal models of disease, including
- inadequate models or a bias in publication towards positive outcomes;
- incorrect dose/timing of administration;
- the need to show additional benefit over existing treatments; and
- ET activation being a consequence rather than a cause of the condition.
Whatever the reason, this experience urges caution in extrapolating data obtained in vitro and in animals to humans. It is hoped that additional information will emerge from unpublished clinical trials that will shed light on previous failures (Kelland and Webb, 2006), and that the combination of powerful pharmacological and molecular approaches will help us to better understand the role of ETA and ETB receptors in health and disease so as to fully realize the clinical potential created by the identification of the powerful vasoconstrictor peptide, ET-1.
Further studies have addressed the role of ET receptor antagonism in erectile dysfunction and aneurysmal SAH, with mixed results. A double-blind pilot study of 53 patients with mild-to-moderate erectile dysfunction demonstrated no benefit of the ETA-selective antagonist BMS-193884 (100mg by mouth) over placebo (Kim et al., 2002).
The ETA-selective antagonist clazosentan was specifically designed for intravenous use in conditions characterized by cerebral vasoconstriction. Its potential in treating severe aneurysmal SAH has recently been addressed in a phase IIa pilot study for the Clazosentan to Overcome Neurological iSChaemia and Infarction OccUrring after Sub-arachnoid haemorrhage (CONSCIOUS-1) trial (Vajkoczy et al., 2005). This ‘pre-CONSCIOUS-1′ study documented a reduction in the frequency and severity of cerebral vasospasm following SAH.
- resistant hypertension,
- chronic kidney disease,
- connective tissue disease and
- sub-arachnoid haemorrhage
Figure 3
Generation and action of endothelin-1 (ET-1) in the vascular wall. The 21-amino-acid peptide, ET-1, is the eventual product of a gene on chromosome 6 that encodes preproET-1 protein. This is converted to proET-1 on secretion into the cytoplasm, which Endothelin-1 is generated from precursor peptides via a two-step proteolytic pathway. Transcription of a gene on chromosome 6 generates mRNA encoding the 212-amino-acid peptide, preproET-1, which, once translated, is stripped of its signal sequence and secreted into the cytoplasm as proET-1 (Inoue et al., 1989). ProET-1 is further cleaved by a furin-like endopeptidase to the 38-amino-acid precursor molecule big ET-1, which circulates in plasma at low concentration but is not thought to possess significant bioactivity (Yanagisawa et al., 1988). Removal of a further 17 COOH-terminal residues, classically but not exclusively by ET-converting enzymes (ECE), results in formation of the mature 21-amino-acid ET-1 (Hirata et al., 1990).
Figure Source: Br J Pharmacol. 2008 March; 153(6): 1105–1119.
The STATE OF SCIENCE for the ET System has been UPDATED by a 2012 study published by the Japanese Pharmacological Society in the Journal of Pharmacological Sciences, 2012.
Since the discovery of ET-1, many researchers have elucidated the physiological and pathophysiological role of ET-1 and ET receptors in the cardiovascular system over the past 20 years. Among many non-peptide and orally available ET-receptor antagonists developed so far, the nonselective ETA/ETB-receptor antagonist bosentan and selective ETA-receptor antagonist ambrisentan are now clinically utilized as agents for pulmonary artery hypertension. There is a possibility that ambrisentan could be widely used in the treatment of pulmonary hypertension because of less interactions with other drugs or side effects such as liver dysfunction. In addition, future clinical applications may provide new findings about which antagonist is more effective, a nonselective ETA/ ETB-receptor or selective ETA-receptor antagonist.
On the other hand, although the selective ETA-receptor antagonist sitaxsentan, which was released in Europe and the United States, was recently forced to be withdrawn because of a high risk of liver failure, it is hoped in the future that other ET-receptor antagonists, including macitentan and zibotentan, currently being developed can be utilized in clinical treatment targeted at the cardiovascular ET-1 system.
In Pathophysiological Roles of Endothelin Receptors in Cardiovascular Diseases
J Pharmacol Sci 119, 302 – 313 (2012) the Japanese Team reported the following important developments:
POINT # 1:
Endothelin (ET)-1 derived from endothelial cells has a much more important role in cardiovascular system regulation than the ET-2 and ET-3 isoforms. Numerous lines of evidence indicate that ET-1 possesses a number of biological activities leading to cardiovascular diseases (CVD) including hypertension and atherosclerosis. Physiological and pathophysiological responses to ET-1 in various tissues are mediated by interactions with ETA- and ETB-receptor subtypes. Both subtypes on vascular smooth muscle cells mediate vasoconstriction, whereas the ETB-receptor subtype on endothelial cells contributes to vasodilatation and ET-1 clearance. Although selective ETA- or nonselective ETA/ETB-receptor antagonisms have been assumed as potential strategies for the treatment of several CVD based on clinical and animal experiments, it remains unclear which antagonisms are suitable for individuals with CVD because upregulation of the nitric oxide system via the ETB receptor is responsible for vasoprotective effects such as vasodilatation and anti-cell proliferation. In this review, we have summarized the current understanding regarding the role of ET receptors, especially the ETB receptor, in CVD.
POINT # 2:
The downstream effects of ET-1 are mediated by two G-protein-coupled receptors ETA and ETB. In the vasculature, the
- ETA receptor on VSMCs mediates vasoconstriction and cell proliferation, whereas the
- Endothelial ETB receptor (generally called ETB1) exerts opposite effects.
Stimulation of the ETB1 receptor leads to the release of vasodilators such as
- nitric oxide (NO) and
- prostaglandin I2 and
- clearance of ET-1 from the circulation within the lungs, kidneys, and liver (19 – 23).
Many vascular relaxing or contraction factors produced in the blood vessel wall maintain normal endothelial function by mutually antagonistic actions. In particular, there are various reports regarding the interaction of ET-1 and NO (27). For instance, ET-1 binding to the ETB1 receptor leads to phosphoinositide 3-kinase (PI3K) activation and subsequent production of phosphatidylinositol- 3,4,5-trisphosphate, which results in recruitment and activation of protein kinase B / Akt (28). This PI3K/Akt pathway is responsible for the phosphorylation and activation of endothelial NO synthase (eNOS). On the other hand, ET-1 also reduces eNOS expression and its activity through increases in hydrogen peroxide by the ETA receptor (29). Therefore, reduced ETB-receptor function and/or overactivation of the ETA receptor eliminate the protective function by NO in vessels and promote the pathological formation of various circulatory diseases
POINT # 3:
3.1 Activation of PKC
Locally generated ET-1 contributes to tissue repair or remodeling of the infarcted heart in an autocrine/paracrine manner, thereby exerting an immediate beneficial effect on damaged tissue (33, 34). Other studies reported that ET-1 administered prior to the onset of ischemia exhibited cardioprotective effects (35, 36). Exogenous ET-1 mimics the cardioprotective effect of pre-conditioning against infarction, apparently via ETA receptor– mediated activation of PKC and a mitochondrial type of ATP-sensitive K+ channel (37, 38). On the other hand, a substantial and long lasting rise in ET-1 induces myocardial hypertrophy, which is associated with a maladaptive effect on myocardial structure and function, thereby leading to fatal events (39 – 42).
The ET-1 / ETA receptor pathway also promotes myocardial fibrosis by enhancing
- cardiac fibroblast proliferation,
- adhesion molecule expression,
- and extracellular matrix deposition (43, 44).
Therefore, the use of ET-receptor antagonists, mostly targeting the ETA receptor, provides beneficial effects in chronic heart failure as evidenced by a reduced infarct size, improved reperfusion coronary flow, or protection during ischemia/reperfusion (39 – 42).
3.2 Sympathetic overactivity and norepinephrine (NE) release
Activation of the ETB receptor may likely prevent post-ischemic cardiac dysfunction through the attenuation of NE release.
In myocardial ischemia, sympathetic overactivity accompanied by excessive norepinephrine (NE) release is associated with cardiac dysfunction and arrhythmia, thereby exaggerating primary ischemia and initiating a malignant cycle that can cause further myocardial damage and high-risk cardiac dysfunction (45, 46). Both ETA and ETB receptors exist in the sympathetic nerve varicosities of guinea pig hearts and exhibit opposite effects on NE release in association with reperfusion arrhythmias: ETA receptors evoke NE release, whereas ETB receptors prevent it (47).
Previous studies showed that a selective ETA-receptor antagonist or the combination of ETA- and ETB-receptor antagonists suppressed excessive NE release from sympathetic nerve endings in postischemic rat hearts and improved cardiac dysfunction after ischemia/reperfusion (9, 47). In addition, exogenous ET-1 induced excessive NE release and subsequent cardiac dysfunction, counteracted with the Na+/H+ exchanger (NHE) inhibitor 5-(N-ethyl-N-isopropyl)-amiloride (EIPA). Thus, the excessive NE overflow triggered by the ET-1 / ETA / NHE pathway seems to be contributive to post-ischemic cardiac dysfunction in rats.
On the other hand, both pharmacological blockade and genetic deficiency of ETB receptors exaggerated the post-ischemic excessive NE release and cardiac dysfunction. Oikonomidis et al. demonstrated that NE levels during the early phase of myocardial infarction are much higher in ETB-deficient rats than wild-type rats and this contributes to the incidence of ventricular arrhythmogenesis, thereby suggesting that the ETB receptor exerts a suppressive effect in sympathetic hyperactivity during the early phase of myocardial infarction (48). Furthermore, activation of the ETB receptor with sarafotoxin-6c substantially
- reduced NE release,
- myocardial infarct size, and
- ischemic arrhythmias (47, 49).
3.3 Big ET-1 must be converted to ET-1 and exerts similar physiological functions
Big ET-1 exerts several physiological actions similar to ET-1, but it must be converted to ET-1 via enzymatic degradation by ECE (50). ECE expression and its enzyme activities are increased in myocardial infarction (51), thereby suggesting that a selective ECE inhibitor may be useful in ischemic cardiac diseases at the clinical level, which warrants further attention. In fact, SM-19712 and FR901533, both of which are highly selective ECE inhibitors, exert a desirable influence on myocardial infarction by decreasing plasma concentrations of ET-1 (52, 53).
On the other hand, exogenously applied big ET-1 has qualitatively similar effects to ET-1 in the cardiovascular system in vivo and in vitro (54 – 57). Against this background, Sharif et al. demonstrated that exogenously applied ET-1 exhibited opposite effects to endogenously released ET-1 on ischemic ventricular arrhythmias (58).
The Japanese Team also reported that exogenous big ET-1 suppressed ischemia/reperfusion-induced NE overflow and improved cardiac dysfunction observed after reperfusion, in spite of the fact that ET-1 content in coronary effluent from the heart exposed to ischemia/reperfusion was increased by exogenous big ET-1 application (59). In addition, treatment with big ET-1 in the presence of A-192621, a selective ETB-receptor antagonist, failed to exert beneficial effects against ischemia/reperfusion-induced NE overflow and subsequent cardiac dysfunction. Thus, ET-1 generated from exogenously applied big ET-1 preferentially may act on ETB receptors rather than ETA receptors, leading to an increase in NO production and subsequent suppression of NE overflow.
POINT # 4:
Neointimal Formaion and Baloon Angioplasty Intervention
Cardiovascular hypertrophy and remodeling are not simply a response to elevated blood pressure. Various vasoactive substances, such as angiotensin II, are implicated in the development of these structural changes (72). ET-1 has potent mitogenic and hypertrophic properties, mainly via stimulation of ETA receptors (73). ETB receptor– mediated actions also protect against cardiovascular hypertrophy via endothelial NO generation, which inhibits mitogenesis and the proliferation of VSMCs (74).
Balloon angioplasty and stent insertion are now widely used for the treatment of coronary arterial disease. Although these procedures improve regional myocardial blood flow by dilating stenotic coronary vessels, one major drawback of this therapeutic approach is restenosis after the procedure because of the proliferation of VSMCs and neointimal formation triggered by mechanical damage to ECs. Several growth factors or vasoactive peptides are related to the process of neointimal formation. In a clinical study, expressions of ECE, ET-1, and ET receptors were enhanced in neointimal VSMCs after percutaneous coronary intervention in human coronary arteries (75). In addition, increases in ET-1 levels were observed in the coronary circulation after percutaneous transluminal coronary angioplasty (76). Anggrahini et al. recently demonstrated that ET-1 derived from ECs mainly contributes to the process of vascular remodeling in the model of flow cessation (10). Thus, ET-1 is closely related to the pathogenesis of restenosis after angioplasty. Similar results have been reported in animal models with restenosis such as balloon injury (77, 78).
Murakoshi et al. showed that vascular remodeling caused by the cessation of blood flow was markedly accelerated in the carotid artery of ETB receptor–knockout mice, and long-term treatment with an ETB-receptor antagonist worsened vascular remodeling in wild-type mice (6).
In contrast, selective ETA-receptor blockade could attenuate this vascular remodeling in the same animals. There has also been a report showing that ET-1 contributes to the remodeling of mesenteric resistance arteries in diabetes via activation of ETA receptors, and ETB receptor–mediated actions provide vasoprotective effects (79). Our previous report has demonstrated that vascular remodeling is markedly attenuated by treatment with a selective ETA receptor antagonist, whereas pharmacological blockade of ETB receptors aggravates neointimal hyperplasia after balloon injury (80). Treatment with an ETA/ETB dual receptor antagonist also suppresses neointimal hyperplasia and the efficacy of treatment is comparable with that of a selective ETA-receptor antagonist (80), thereby suggesting that the antagonism of ETB receptors does not seem to impair the positive effects of concomitant ETA receptor antagonism. Furthermore, we also confirmed similar results in ETB-deficient rats. Therefore, antagonism of the ET-1 / ETA receptor pathway appears to be essential for preventing neointimal hyperplasia after balloon injury, irrespective of the presence of ETB receptor–mediated actions.
POINT # 5:
Pulmonary Hypertention
The lungs are known to synthesize ET-1 and possess ETA and ETB receptors, both of which are involved in physiological and pathophysiological actions of ET-1 in the lung. In particular, endothelial ETB receptors in lungs are responsible for circulating ET-1 clearance, with close to 50% removal during pulmonary transit in humans (81).
These findings suggest that an ETA/ETB-receptor antagonist brings reasonable validity by suppressing overactivation of the ET-1 / ETA receptor system and vasoconstriction via the ETB2 receptor. Moreover, it is less likely that the ETB1 receptor actively functions as a protective factor through increases in NO production.
POINT # 6:
Hypertension: Salt-sensitive type
ETB receptor–mediated actions are protective in the pathogenesis of salt-sensitive hypertension. These results also suggest that the antagonism of the ETA receptor is essential for protection from cardiovascular disease including salt-sensitive hypertension, irrespective of the presence of the ETB receptor. This view may explain the findings that selective ETA-receptor antagonists and nonselective ETA/ETB-receptor antagonists similarly improve salt-sensitive hypertension and related tissue injuries.
POINT # 7:
Gender Differences in CVD
ETB receptor–mediated actions seem to occur downstream of the vasoprotective effects of estrogen, although the relationship between ETB receptor– and estrogen receptor– signaling systems remains unclear. On the other hand, neointimal hyperplasia observed in female ETB deficient rats is almost completely suppressed by ETA or ETA/ETB-receptor antagonists. Thus, augmentation of ETA receptor–mediated actions under ETB-receptor dysfunction seems to be responsible for the abolition of sex differences in vascular remodeling.
Sex differences are considered to be caused by the vasoprotective effect of estrogen (108 – 110). In fact, several clinical studies showed that postmenopausal women who receive estrogen replacement therapy (ERT) have a substantially lower risk of incidence of cardiovascular disease (111, 112). The protective effect of estrogen on the cardiovascular system is closely related to the up-regulation of endothelial NO production and downregulation of adhesion molecule activity, smooth muscle proliferation/migration, and superoxide production (113 – 115).
It is reasonable to assume that 17β-estradiol is mainly involved in sex differences in the ET system. Sex differences in ET-receptor density, as well as in the ratio of ET-receptor subtypes, have been also investigated. Ergul et al. reported that men’s saphenous veins have a larger number of ET receptors and an increased ratio of ETA to ETB receptors compared to women’s saphenous veins and that these differences were reflected by the sex differences in ET-1-induced vascular contractile responses (127). On the other hand, although several animal studies also indicated that ET receptors are involved
in the sex differences in the incidence of CVD, the effect of estrogen on these receptors is quite contradictory.
For example, Nuedling et al. demonstrated upregulation of the ETB receptor in the heart of ovariectomized female spontaneously hypertensive rats, which could be reversed by exogenous estrogen replacement (128). They also confirmed downregulation of the ETB receptor by 17β-estradiol in cultured cardiomyocytes. Others reported similar results showing that vascular mRNA expression of ETB, but not ETA receptors in DOCA-salt–induced hypertensive rats was higher in males than that observed in females (129, 130).
Table 1
Comparison of ECE inhibitors in clinical development and experimental use
Drug | Development phase | Selectivity | IC50 (nM) | Reference | ||
---|---|---|---|---|---|---|
ECE/NEP | ECEa | NEP | ACE | |||
TMC 66 | Preclinical | 2900 | Asai et al. (1999) | |||
Daglutril (SLV 306, KC 12615) | II | Tabrizchi (2003) | ||||
CGS 26303, CGS 26393 | Preclinical | 0.0033 | 410 | 0.9 | DeLombaert et al. (1994) | |
Phosphoramidon | Preclinical | 0.0097 | 3500 | 34 | 78![]() |
Kukkola et al. (1995) |
CGS 35601, CGS 37808 | Preclinical | 0.036 | 55 | 2 | 22 | Battistini et al. (2005) |
CGS 31447 | Preclinical | 0.23 | 21 | 4.8 | Shetty et al. (1998) | |
SCH 54470 | Preclinical | 1.3 | 70 | 90 | 2.5 | McKittrick et al. (1996) |
CGS 34043 | Preclinical | 19 | 5.8 | 110 | DeLombaert et al. (2000) | |
WS 75624 B | Preclinical | 42 | 79 | 3300 | Tsurumi et al. (1995b) | |
B 90063 | Preclinical | 70 | 1000 | 70![]() |
Takaishi et al. (1998) | |
CGS 35066 | Preclinical | 100 | 22 | 2300 | Trapani et al. (2000) | |
PD 069185, PD 159790 | Preclinical | >200 | 900 | >180![]() |
Ahn et al. (1998) | |
SM 19712 | Preclinical | >240 | 42 | >10![]() |
>10![]() |
Umekawa et al. (2000) |
FR 901533, WS 79089 B | Preclinical | >700 | 140 | >100![]() |
Tsurumi et al. (1995a) | |
Ro 68-7629 | Preclinical | >90![]() |
1.1 | >100![]() |
>100![]() |
Muller et al. (2002) |
Table 2
Comparison of ET receptor antagonists in clinical use, clinical development and experimental use
Drug | Trade name | Development phase | Selectivity | Ki (nM) | Bioavailability | Eliminationt1/2 (hours) | Reference | |
---|---|---|---|---|---|---|---|---|
ETA/ETB | ETA | ETB | ||||||
(a) Mixed ETA/ETB receptor antagonists | ||||||||
Tezosentan (Ro 61-0612) | Veletri | III | 1.2 | 18 | 21 | i.v. | 2.1 (rat) | Clozel et al. (1999) |
PD 145065 | Preclinical | 3.8 | 4a | 15a | Doherty et al. (1993) | |||
PD 142893 | Preclinical | 10 | 15a | 150a | Cody et al. (1992) | |||
Bosentan (Ro 47-0203) | Tracleer | IV | 20 | 4.7 | 95 | Oral | 3.7–7.5 (human) | Clozel et al. (1994); Weber et al. (1996) |
TAK 044 | II | 34 | 3.8a | 130a | i.v. | 0.5–1.0 (human) | Watanabe et al. (1995);Haynes et al. (1996) | |
Avosentan (SPP 301) | III | 50 | Oral | 7.5–15.2 (human) | Dieterle et al. (2004)b | |||
(b) Selective ETA receptor antagonists | ||||||||
Enrasentan (SB 217242) | III | 100 | 1.1 | 111 | Oral | 3.3 (rat) | Ohlstein et al. (1996) | |
Darusentan (LU 135252) | III | 170 | 6 | 1000 | Oral | 12.0 (rat) | Riechers et al. (1996);Cernacek et al. (1998) | |
Ambrisentan (LU 208075) | Letairis | IV | 200 | 1 | 195 | Oral | 9.0–15.0 (PAH patients) | Riechers et al. (1996); Galiéet al. (2005) |
YM 598 | II | 390 | 3.1a | 1200a | Oral | 2.5 (rat) | Harada et al. (2001) | |
S 0139 | II | 1000 | 1 | 1000 | Oral | Mihara et al. (1994) | ||
Clazosentan (Ro 61-1790) | IIb | 1300 | 9.5c | 6.4c | i.v. | 0.8 (monkey) | Roux et al. (1997) | |
Atrasentan (A 147627) | Xinlay | III | 2000 | 0.069 | 139 | Oral | 2.5 (monkey) | Opgenorth et al. (1996) |
BQ 123 | Preclinical | 2500 | 7.3a | 18![]() |
i.v. | Ihara et al. (1992) | ||
ZD 4054 | IIa | >4800 | 21a | >100![]() |
Oral | 9.1–9.7 (human) | Morris et al. (2005) | |
Sitaxsentan (TBC 11251) | Thelin | IV | 7000 | 1.4a | 9800a | Oral | 5.9–7.5 (rat) | Wu et al. (1997) |
BMS 193884 | II | 13![]() |
1.4 | 18![]() |
Oral | 9.0 (monkey) | Murugesan et al. (2000) | |
FR 139317 | Preclinical | 7300 | 1 | 7300 | i.v. | Aramori et al. (1993) | ||
Edonentan (BMS 207940) | IIa | 81![]() |
0.01 | 810 | Oral | 17.0 (monkey) | Murugesan et al. (2003) | |
TBC 3711 | I | 440![]() |
0.08a | Oral | 5.3 (rat) | Wu et al. (2004) | ||
(c) Selective ETB receptor antagonists | ||||||||
BQ 788 | Preclinical | 1100 | 1300a | 1.2a | i.v. | Ishikawa et al. (1994) | ||
A 192621 | Preclinical | 1300 | 8200a | 6.4a | Oral | 5.0 (rat) | von Geldern et al. (1999) |
Table 3
Summary and completed clinical trials of ET receptor antagonists in indicated cardiovascular diseases
Condition | Antagonist | Study | Outcome |
---|---|---|---|
Pulmonary arterial hypertension | Bosentan | BREATHE-1 | Improvement in primary and secondary end points, well tolerated |
BREATHE-4 | Improvement in end points, well tolerated | ||
Sitaxsentan | Open label | ||
STRIDE 1–6 | Sustained improvement, well tolerated | ||
Ambrisentan | AMB-220 | Improvement in end points, sustained for >2 years, well tolerated | |
ARIES-2 | Improvement in end points, well tolerated | ||
Heart failure | |||
Chronic | Bosentan | Pilot | Short-term haemodynamic improvements |
ENABLE-1; -2 | No benefit, early worsening of symptoms | ||
REACH-1 | No benefit, toxic effects, trial stopped, trend to reduced mortality | ||
Enrasentan | ENCOR | No benefit, increased adverse events, trend to increased mortality | |
Darusentan | EARTH | No benefit, increased adverse events | |
HEAT-CHF | Some haemodynamic benefit, adverse events (including death) at higher dose | ||
BMS 193884 | No data, trial probably discontinued | ||
Edonentan | No data, trial probably discontinued | ||
Acute | Tezosentan | RITZ-1 | Safety trial, well tolerated, some haemodynamic improvement |
RITZ-2 | Acute improvement in haemodynamics, dose dependence | ||
RITZ-3 | Follow-up to RITZ-2, never conducted | ||
RITZ-4 | No benefit and adverse events | ||
204 | Dose-dependent improvements in haemodynamics, decreased urine production | ||
VERITAS-1, -2 | Limited benefit, trials discontinued | ||
Hypertension | Bosentan | Reduction of SBP and DBP in essential HT | |
Darusentan | HEAT-HTN | Dose-dependent reduction of SBP and DBP in essential HT | |
DAR-201 | Reduction of SBP and DBP in refractory HT | ||
Aneurysmal SAH | Clazosentan | Pre-CONSCIOUS-1 | Reduced frequency and severity of cerebral vasospasm |
Erectile dysfunction | BMS 193884 | No improvement over placebo | |
Scleroderma (ulcers) | Bosentan | RAPIDS-1, -2 | Significant reduction in ulcers, well-tolerated |
Table 4
[Source: J Pharmacol Sci 119, 302 – 313 (2012)]
Randomized clinical trials evaluating endothelin receptor antagonists in
Chronic Heart Failure
ETR antagonist(selectivity) | Study | Patients | Outcome |
Bosentan(ETA/ETB) | REACH-1 | N=370 NYHA IIIb-IV, LVEF<35% | No improvements in end point |
ENABLE I | N=1613 NYHA , LVEF<35% | No improvements in end point | |
ENABLE II | Same NYHA, LVEF<35% | No improvements in end point | |
Tozosertan(ETA/ETB) | RITZ 1 | N=675 ADHF | No difference in all end points |
RITZ-2 | N=292 ADHF | Increase in Cardiac Index decrease in PCWP | |
RITZ-4 | N=193 ADHF w/ACS | No difference in all end points | |
RITZ-5 | N=84 ACHF | No improvements in end point | |
Darusentan(ETA) | HEAT | N=157 NYHA II, LVEF<35% | Improvement in Cardiac Index |
EARTH | N=642 NYHA II-IV, LVEF<35% | No difference in end points | |
Enrasentan(ETA/ETB) | ENCOR | N=419 NYHA II-III, LVEF<35% | Better outcome in placebo group |
REACH-1: Research on Endothelin Antagonism in Chronic Heart Failure,
ENABLE: Endothelin Antagonist Bosentan for Lowering Cardiac Events
RITZ: Randomized Intravenous TeZosentan,
HEAT: Heart Failure ET[A] Receptor Blockade Trial,
EARTH: EndothelinA Receptor Antagonist Trial in Heart Failure,
ENCOR: ENrasentan COoperative Randomized, ETR: endothelin receptor, NYHA: New York Heart Association,
Abbreviations: ADHF: acute decompensated heart failure, ACS: acute coronary syndrome, ACHF: acute congestive heart failure, LVEF: left ventricular ejection fraction.
Source:
Pathophysiological Roles of Endothelin Receptors in Cardiovascular Disease
Big Endothelin in chronic heart failure: marker of disease severity or genetic determination?
Source
1st Internal Cardio-angiological Department, St. Anne’s Hospital, Brno, Czech Republic.
Abstract
The first objective of the study was to compare the levels of big endothelin and endothelin-1 and other noninvasive parameters used for evaluation of disease severity in patients with stable chronic heart failure (CHF). Endothelin-1 and big endothelin plasma concentrations were measured in 124 chronic heart failure patients. The second objective of the study was to prove an association between endothelin-1 and big endothelin plasma levels and two frequent polymorphisms in the endothelin-1 coding gene (6p21-23) -3A/-4A and G (8002) A in patients with chronic heart failure. Thirdly, we tried to associate other noninvasive parameters of CHF, especially cardiothoracic index (CTI), NYHA classification, signs of pulmonary congestion (PC) and ejection fraction (EF) with determined genotypes of the two ET-1 polymorphic variants. There were significant differences between big endothelin levels in NYHA II versus IV (P<0.001) and NYHA III versus IV (P<0.001) and endothelin-1 in NYHA II versus IV (P<0.001) and NYHA III versus IV (P<0.001). No associations between plasma levels of endothelin-1 and big endothelin and polymorphisms G (8002) A and -3A/-4A in gene coding endothelin-1 were found. In patients with CHF with CTI above 60% the number of carriers of genotypes with ET-1 8002A (AA and AG genotypes) increases. Concerning on the -3A/-4A ET-1 polymorphism, we observed a significant difference in genotype distribution as well as in allelic frequency in the group of patients with CTI above 60% between patients without and with pulmonary congestion. The allelic frequency of 3A allele is twice elevated in the patients with pulmonary congestion (37.8 vs. 78.1%, respectively).
- ALL THE TABLES ABOVE AND NARRATIVE based chiefly on the following two seminal papers:
J Pharmacol Sci 119, 302 – 313 (2012)
https://www.jstage.jst.go.jp/article/jphs/119/4/119_12R01CR/_pdf
Pathophysiological Roles of Endothelin Receptors in Cardiovascular Diseases
Mamoru Ohkita1, Masashi Tawa1,2, Kento Kitada1,3, and Yasuo Matsumura1,*
1Laboratory of Pathological and Molecular Pharmacology, Osaka University of Pharmaceutical Sciences,
4-20-1 Nasahara, Takatsuki, Osaka 569-1094, Japan
2Department of Pharmacology, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
3Department of Pharmacology, Kagawa University Medical School,
1750–1 Ikenobe, Miki-cho, Kita-gun, Kagawa 761-0793, Japan
The endothelin system as a therapeutic target in cardiovascular disease: great expectations or bleak house?
N S Kirkby,1 P W F Hadoke,1 A J Bagnall,1 and D J Webb1,*
http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2275436/#bib140
http://onlinelibrary.wiley.com/doi/10.1038/sj.bjp.0707516/full
http://onlinelibrary.wiley.com/doi/10.1038/sj.bjp.0707516/references
References from
Journal of Pharmacological Sciences
© The Japanese Pharmacological Society
J Pharmacol Sci 119, 302 – 313 (2012)
https://www.jstage.jst.go.jp/article/jphs/119/4/119_12R01CR/_pdf
The refences below are accessable online at
https://www.jstage.jst.go.jp/article/jphs/119/4/119_12R01CR/_article/references
[click on the left hand side on PubMed]
[IF “Live Link N/A”, source is accessible at Libraries, only]
1 Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M,
Mitsui Y, et al. A novel potent vasoconstrictor peptide produced
by vascular endothelial cells. Nature. 1988;332:411–415.Live Link N/A
2 Levin ER. Endothelins as cardiovascular peptides. Am J Nephrol.
1996;16:246–251. Live Link N/A
3 Rich S, McLaughlin VV. Endothelin receptor blockers in cardiovascular
disease. Circulation. 2003;108:2184–2190.
4 Agapitov AV, Haynes WG. Role of endothelin in cardiovascular
disease. J Renin Angiotensin Aldosterone Syst. 2002;3:1–15.
5 Matsumura Y, Kuro T, Kobayashi Y, Konishi F, Takaoka M,
Wessale JL, et al. Exaggerated vascular and renal pathology in
endothelin-B receptor-deficient rats with deoxycorticosterone
acetate-salt hypertension. Circulation. 2000;102:2765–2773.
6 Murakoshi N, Miyauchi T, Kakinuma Y, Ohuchi T, Goto K,
Yanagisawa M, et al. Vascular endothelin-B receptor system in
vivo plays a favorable inhibitory role in vascular remodeling after
injury revealed by endothelin-B receptor-knockout mice. Circulation.
7 Elmarakby AA, Dabbs Loomis E, Pollock JS, Pollock DM. ETA
receptor blockade attenuates hypertension and decreases reactive
oxygen species in ETB receptor-deficient rats. J Cardiovasc
Pharmacol. 2004;44:S7–S10. Live Link N/A
8 Nishida M, Okada Y, Akiyoshi K, Eshiro K, Takoaka M, Gariepy
CE, et al. Role of endothelin ETB receptor in the pathogenesis of
monocrotaline-induced pulmonary hypertension in rats.Eur J
Pharmacol. 2004;496:159–165. Live Link N/A
9 Yamamoto S, Matsumoto N, Kanazawa M, Fujita M, Takaoka M,
Gariepy CE, et al. Differential contributions of endothelin-A and
endothelin-B receptors in postischemic cardiac dysfunction and
norepinephrine overflow in rat hearts. Circulation. 2005;111:
302–309. Live Link N/A
10 Anggrahini DW, Emoto N, Nakayama K, Widyantoro B, Adiarto
S, Iwasa N, et al. Vascular endothelial cell-derived endothelin-1
mediates vascular inflammation and neointima formation following
blood flow cessation. Cardiovasc Res. 2009;82:143–151. Live Link N/A
11 Nunez DJR, Brown MJ, Davenport AP, Neylon CB, Schofield JP,
310 M Ohkita et al
Wyse RK. Endothelin-1 messenger-RNA is widely expressed in
porcine and human tissues. J Clin Invest. 1990;85:1537–1541.
12 Resink TJ, Hahn AWA, Scott-Burden T, Powell J, Weber E,
Buhler FR. Inducible endothelin messenger RNA expression and
peptide secretion in cultured human vascular smooth muscle
cells. Biochem Biophys Res Commun. 1990;168:1303–1310.
13 Firth JD, Ratcliffe PJ. Organ distribution of the 3 rat endothelin
messenger RNAs and the effects of ischemia on renal gene expression.
J Clin Invest. 1992;90:1023–1031.
14 Wilson DB, Dorfman DM, Orkin SH. A nonerythroid GATAbinding
protein is required for function of the human preproendothelin-
1 promoter in endothelial cells. Mol Cell Biol. 1990;
15 Hu J, Discher DJ, Bishopric NH, Webster KA. Hypoxia regulates
expression of the endothelin-1 gene through a proximal hypoxiainducible
factor-1 binding site on the antisense strand. Biochem
Biophys Res Commun. 1998;245:894–899.
16 Rodriguez-Pascual F, Redondo-Horcajo M, Lamas S. Functional
cooperation between Smad proteins and activator protein-1 regulates
transforming growth factor-β-mediated induction of endothelin-
1 expression. Circ Res. 2003;92:1288–1295. Live Link N/A
17 Quehenberger P, Bierhaus A, Fasching P, Muellner C, Klevesath
M, Hong M, et al. Endothelin 1 transcription is controlled by
nuclear factor κB in AGE-stimulated cultured endothelial cells.
18 Wort SJ, Ito M, Chou PC, Mc Master SK, Badiger R, Jazrawi E,
et al. Synergistic induction of endothelin-1 by tumor necrosis
factor a and interferon γ is due to enhanced NF-κB binding and
histone acetylation at specific κB sites. J Biol Chem. 2009;
19 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.
20 Dupuis J, Goresky CA, Fournier A. Pulmonary clearance of circulating
endothelin-1 in dogs in vivo: exclusive role of ETB receptors.
J Appl Physiol. 1996;81:1510–1515. Live Link N/A
21 Dupuis J, Schwab AJ, Simard A, Cernacek P, Stewart DJ,
Goresky CA. Kinetics of endothelin-1 binding in the dog liver
microcirculation in vivo. Am J Physiol. 1999;277:G905–G914. Live Link N/A
22 Tran Duc AT, Schwab AJ, Simard A, Villeneuve L, Dupuis J.
Reduction in hepatic endothelin-1 clearance in cirrhosis. Clin Sci
(Lond). 2003;105:227–234. Live Link N/A
23 Johnström P, Fryer TD, Richards HK, Harris NG, Barret O, Clark
JC, et al. Positron emission tomography using 18F-labelled endothelin-
1 reveals prevention of binding to cardiac receptors
owing to tissue-specific clearance by ETB receptors in vivo. Br J
Pharmacol. 2005;144:115–122. Live Link N/A
24 Iwasa S, Fan J, Shimokama T, Nagata M, Watanabe T. Increased
immunoreactivity of endothelin-1 and endothelin B receptor in
human atherosclerotic lesions. A possible role in atherogenesis.
Atherosclerosis. 1999;146:93–100. Live Link N/A
25 Cardillo C, Kilcoyne CM, Waclawiw M, Cannon RO 3rd, Panza
JA. Role of endothelin in the increased vascular tone of patients
with essential hypertension. Hypertension. 1999;33:753–758.
26 Dimitrijevic I, Edvinsson ML, Chen Q, Malmsjö M, Kimblad
PO, Edvinsson L. Increased expression of vascular endothelin
type B and angiotensin type 1 receptors in patients with ischemic
heart disease. BMC Cardiovasc Disord. 2009;9:40. Live Link N/A
27 Bourque SL, Davidge ST, Adams MA. The interaction between
endothelin-1 and nitric oxide in the vasculature: new perspectives.
Am J Physiol Regul Integr Comp Physiol. 2011;300: R1288–R1295. Live Link N/A
28 Liu S, Premont RT, Kontos CD, Huang J, Rockey DC. Endothelin-
1 activates endothelial cell nitric-oxide synthase via heterotrimeric
G-protein βγ subunit signaling to protein kinase B/Akt. J
Biol Chem. 2003;278:49929–49935.
29 Wedgwood S, Black SM. Endothelin-1 decreases endothelial
NOS expression and activity through ETA receptor-mediated
generation of hydrogen peroxide. Am J Physiol Lung Cell Mol
Physiol. 2005;288:L480–L487. Live Link N/A
30 Matsuyama K, Yasue H, Okumura K, Saito Y, Nakao K,
Shirakami G, et al. Increased plasma level of endothelin-1-like
immunoreactivity during coronary spasm in patients with coronary
spastic angina. Am J Cardiol. 1991;68:991–995.
31 Miyauchi T, Yanagisawa M, Tomizawa T, Sugishita Y, Suzuki N,
Fujino M, et al. Increased plasma concentrations of endothelin-1
and big endothelin-1 in acute myocardial infarction. Lancet.
32 Malatino LS, Grassi R, Stancanelli B. Release of immunoreactive
endothelin from the heart during percutaneous transluminal
coronary angioplasty. Am Heart J. 1993;126:700–702.
33 Brunner F, DuToit EF, Opie LH. Endothelin release during
ischaemia and reperfusion of isolated perfused rat hearts. J Mol
Cell Cardiol. 1992;24:1291–1305. Live Link N/A
34 Hasdai D, Kornowski R, Battler A. Endothelin and myocardial
ischemia. Cardiovasc Drugs Ther. 1994;8:589–599. Live Link N/A
35 Bugge E, Ytrehus K. Endothelin-1 can reduce infarct size through
protein kinase C and KATP channels in the isolated rat heart. Cardiovasc
36 Wang P, Gallagher KP, Downey JM, Cohen MV. Pretreatment
with endothelin-1 mimics ischemic preconditioning against infarction
in isolated rabbit heart. J Mol Cell Cardiol. 1996;28: 579–588. Live Link N/A
37 Hide EJ, Piper J, Thiemermann C. Endothelin-1-induced reduction
of myocardial infarct size by activation of ATP-sensitive
potassium channels in a rabbit model of myocardial ischaemia
and reperfusion. Br J Pharmacol. 1995;116:2597–2602.
38 Gourine AV, Molosh AI, Poputnikov D, Bulhak A, Sjöquist PO,
Pernow J. Endothelin-1 exerts a preconditioning-like cardioprotective
effect against ischaemia/reperfusion injury via the ETA
receptor and the mitochondrial KATP channel in the rat in vivo. Br
J Pharmacol. 2005;144:331–337.
39 Brunner F, Opie LH. Role of endothelin-A receptors in ischemic
contracture and reperfusion injury. Circulation. 1998;97: 391–398. Live Link N/A
40 Galiuto L, DeMaria AN, Del Balzo U, May-Newman K, Flaim
SF, Wolf PL, et al. Ischemia-reperfusion injury at the microvascular
level: treatment by endothelin A-selective antagonist and
evaluation by myocardial contrast echocardiography. Circulation.
2000;102:3111–3116. Live Link N/A
41 Pernow J, Wang QD. Endothelin in myocardial ischaemia and
reperfusion. Cardiovasc Res. 1997;33:518–526.
42 Reriani M, Raichlin E, Prasad A, Mathew V, Pumper GM, Nelson
RE, et al. Long-term administration of endothelin receptor antagonist
improves coronary endothelial function in patients with
early atherosclerosis. Circulation. 2010;122:958–966.
43 Intengan HD, Schiffrin EL. Vascular remodeling in hypertension:
roles of apoptosis, inflammation, and fibrosis. Hypertension.
44 Callera GE, Montezano AC, Touyz RM, Zorn TM, Carvalho
Endothelin in Cardiovascular Diseases 311 MH, Fortes ZB, et al. ETA receptor mediates altered leukocyteendothelial cell interaction and adhesion molecules expression in
DOCA-salt rats. Hypertension. 2004;43:872–879. Live Link N/A
45 Braunwald E, Sobel BE. Coronary blood flow and myocardial
ischemia. In: Braunwald, E, editor. Heart disease, a textbook of
cardiovascular medicine. Philadelphia: WB Saunders; 1988.
p. 1191–1221. Live Link N/A
46 Schömig A, Haass M, Richardt G. Catecholamine release and
arrhythmias in acute myocardial ischemia. Eur Heart J. 1991;
12 Suppl F:38–47. Live Link N/A
47 Isaka M, Kudo A, Imamura M, Kawakami H, Yasuda K. Endothelin
receptors, localized in sympathetic nerve terminals of
the heart, modulate norepinephrine release and reperfusion arrhythmias.
Basic Res Cardiol. 2007;102:154–162. Live Link N/A
48 Oikonomidis DL, Tsalikakis DG, Baltogiannis GG, Tzallas AT,
Xourgia X, Agelaki MG, et al. Endothelin-B receptors and ventricular
arrhythmogenesis in the rat model of acute myocardial
infarction. Basic Res Cardiol. 2010;105:235–245. Live Link N/A
49 Crockett TR, Gray GA, Kane KA, Wainwright CL. Sarafotoxin
6c (S6c) reduces infarct size and preserves mRNA for the ETB
receptor in the ischemic/reperfused myocardium of anesthetized
rats. J Cardiovasc Pharmacol. 2004;44:148–154. Live Link N/A
50 Giannessi D, Del Ry S, Vitale RL. The role of endothelins and
their receptors in heart failure. Pharmacol Res. 2001;43:
51 Bohnemeier H, Pinto YM, Horkay F, Tóth M, Juhász-nagy A,
Orzechowski HD, et al. Endothelin converting-enzyme-1 mRNA
expression in human cardiovascular disease. Clin Exp Hypertens.
1998;20:417–437. Live Link N/A
52 Umekawa K, Hasegawa H, Tsutsumi Y, Sato K, Matsumura Y,
Ohashi N. Pharmacological characterization of a novel sulfonylureid-
pyrazole derivative, SM-19712, a potent nonpeptidic
inhibitor of endothelin converting enzyme.
Jpn J Pharmacol. 2000;84:7–15.
53 Martin P, Tzanidis A, Stein-Oakley A, Krum H. Effect of a highly
selective endothelin-converting enzyme inhibitor on cardiac remodeling
in rats after myocardial infarction. J Cardiovasc Pharmacol.
54 Matsumura Y, Hisaki K, Takaoka M, Morimoto S. Phosphoramidon,
a metalloproteinase inhibitor, suppresses the hypertensive
effect of big endothelin-1. Eur J Pharmacol. 1990;185:103–106.
55 Trapani AJ, De Lombaert S, Kuzmich S, Jeng AY. Inhibition of
big ET-1-induced pressor response by an orally active dual inhibitor
of endothelin-converting enzyme and neutral endopeptidase
24.11. J Cardiovasc Pharmacol. 1995;26:S69–S71.
56 Fecteau MH, Honoré JC, Plante M, Labonté J, Rae GA,
D’Orléans-Juste P. Endothelin-1 (1-31) is an intermediate in the
production of endothelin-1 after big endothelin-1 administration
in vivo. Hypertension. 2005;46:87–92. Live Link N/A
57 Grover GJ, Sleph PG, Fox M, Trippodo NC. Role of endothelin-1
and big endothelin-1 in modulating coronary vascular tone, contractile
function and severity of ischemia in rat hearts.
58 Sharif I, Kane KA, Wainwright CL. Endothelin and ischaemic
arrhythmias-antiarrhythmic or arrhythmogenic? Cardiovasc Res.
1998;39:625–632. Live Link N/A
59 Tawa M, Fukumoto T, Ohkita M, Yamashita N, Geddawy A,
Imamura T, et al. Effects of exogenous big endothelin-1 on postischemic
cardiac dysfunction and norepinephrine overflow in rat
hearts. Hypertens Res. 2011;34:218–224.
60 Khamaisi M, Raz I, Shilo V, Rosenberger C, Dahan R, Abassi Z,
et al. Diabetes and radiocontrast media increase endothelin converting
enzyme-1 in the kidney. Kidney Int. 2008;74:91–100. Live Link N/A
61 Valdenaire O, Lepailleur-Enouf D, Egidy G, Thouard A, Barret
A, Vranckx R, et al. A fourth isoform of endothelin-converting
enzyme (ECE-1) is generated from an additional promoter. Eur J
62 Emoto N, Nurhantari Y, Alimsardjono H, Xie J, Yamada T,
Yanagisawa M, et al. Constitutive lysosomal targeting and degradation
of bovine endothelin-converting enzyme-1a mediated by
novel signals in its alternatively spliced cytoplasmic tail. J Biol
63 Packer M. Multicentre, double-blind, placebo-controlled study
of long-term endothelin blockade with bosentan in chronic heart
failure – results of the REACH-1 trial [Abstract]. Circulation.
1998;98:I-3. Live Link N/A
64 Teerlink JR, Massie B, Cleland J, Tzivoni D, for the RITZ-1 Investigators.
A double-blind, parallel-group, multi-center, placebocontrolled
study to investigate the efficacy and safety of tezosentan
in reducing symptoms in patients with acute decompensated
heart failure [Abstract]. Circulation. 2001;104:II-526. Live Link N/A
65 Torre-Amione G, Young JB, Colucci WS, Lewis BS, Pratt C,
Cotter G, et al. Hemodynamic and clinical effects of tezosentan,
an intravenous dual endothelin receptor antagonist, in patients
hospitalized for acute decompensated heart failure. J Am Coll
66 O’Connor CM, Gattis WA, Adams KF Jr, Hasselblad V, Chandler
B, Frey A, et al. Randomized Intravenous Tezosentan Study-4
Investigators. Tezosentan in patients with acute heart failure and
acute coronary syndromes: results of the Randomized Intravenous
TeZosentan Study (RITZ-4). J Am Coll Cardiol. 2003;
67 Kaluski E, Kobrin I, Zimlichman R, Marmor A, Krakov O, Milo
O, et al. RITZ-5: randomized intravenous Tezosentan (an endothelin-
A/B antagonist) for the treatment of pulmonary edema:
a prospective, multicenter, double-blind, placebo-controlled
study. J Am Coll Cardiol. 2003;41:204–210. Live Link N/A
68 Lüscher TF, Enseleit F, Pacher R, Mitrovic V, Schulze MR,
Willenbrock R, et al. Hemodynamic and neurohumoral effects of
selective endothelin A (ET(A)) receptor blockade in chronic
heart failure: the Heart Failure ET(A) Receptor Blockade Trial
(HEAT). Circulation. 2002;106:2666–2672. Live Link N/A
69 Anand I, McMurray J, Cohn JN, Konstam MA, Notter T, Quitzau
K, et al. Long-term effects of darusentan on left-ventricular remodelling
and clinical outcomes in the Endothelin A Receptor
Antagonist Trial in Heart Failure (EARTH): randomised, doubleblind,
placebo-controlled trial. Lancet. 2004;364:347–354.
70 Abraham WT, Ascheim D, Demarco T, Klapholz M, Kukin M,
Miller A, et al. Effects of enrasentan, a non selective endothelin
receptor antagonist, in class II to III heart failure: results of the
Enrasentan Cooperative Randomized (ENCOR) Evaluation
[Abstract]. J Am Coll Cardiol. 2001;38:612. Live Link N/A
71 Battistini B, Berthiaume N, Kelland NF, Webb DJ, Kohan DE.
Profile of past and current clinical trials involving endothelin receptor
antagonists: the novel “-sentan” class of drug. Exp Biol
Med. 2006;231:653–695. Live Link N/A
72 Gibbons GH, Dzau VJ. The emerging concept of vascular remodeling.
N Engl J Med. 1994;330:1431–1438.
73 Ohlstein EH, Arleth A, Bryan H, Elliott JD, Sung CP. The selective
endothelin ETA receptor antagonist BQ123 antagonizes en312
M Ohkita et al
dothelin-1-mediated mitogenesis.Eur J Pharmacol. 1992;225:
74 Garg UC, Hassid A. Nitric oxide-generating vasodilators and
8-bromo-cyclic guanosine monophosphate inhibit mitogenesis
and proliferation of cultured rat vascular smooth muscle cells. J
Clin Invest. 1989;83:1774–1777.
75 Shirai N, Naruko T, Ohsawa M, Ikura Y, Sugama Y, Hirayama M,
et al. Expression of endothelin-converting enzyme, endothelin-1
and endothelin receptors at the site of percutaneous coronary intervention
in humans. J Hypertens. 2006;24:711–721.
76 Takase H, Sugiyama M, Nakazawa A, Toriyama T, Hayashi K,
Goto T, et al. Increased endogenous endothelin-1 in coronary
circulation is associated with restenosis after coronary angioplasty.
Can J Cardiol. 2003;19:902–906. Live Link N/A
77 Douglas SA, Louden C, Vickery-Clark LM, Storer BL, Hart T,
Feuerstein GZ, et al. A role for endogenous endothelin-1 in
neointimal formation after rat carotid artery balloon angioplasty.
Protective effects of the novel nonpeptide endothelin receptor
antagonist SB 209670. Circ Res. 1994;75:190–197.
78 Wang X, Douglas SA, Louden C, Vickery-Clark LM, Feuerstein
GZ, Ohlstein EH. Expression of endothelin-1, endothelin-3, endothelin-
converting enzyme-1, and endothelin-A and endothelin-
B receptor mRNA after angioplasty-induced neointimal formation
in the rat. Circ Res. 1996;78:322–328.
79 Sachidanandam K, Portik-Dobos V, Harris AK, Hutchinson JR,
Muller E, Johnson MH, et al. Evidence for vasculoprotective effects
of ETB receptors in resistance artery remodeling in diabetes.
80 Kitada K, Yui N, Matsumoto C, Mori T, Ohkita M, Matsumura Y.
Inhibition of endothelin ETB receptor system aggravates neointimal
hyperplasia after balloon injury of rat carotid artery. J Pharmacol
81 Dupuis J, Stewart DJ, Cernacek P, Gosselin G. Human pulmonary
circulation is an important site for both clearance and production
of endothelin-1. Circulation. 1996;94:1578–1584. Live Link N/A
82 Stewart DJ, Levy R, Cernacek P, Langleben D. Increased plasma
endothelin-1 in pulmonary hypertension: marker or mediator of
disease? Ann Intern Med. 1991;114:464–469.
83 Cody RJ, Haas GJ, Binkley PF, Capers Q, Kelley R. Plasma endothelin
correlates with the extent of pulmonary hypertension in
patients with chronic congestive heart failure. Circulation.
84 Iglarz M, Clozel M. At the heart of tissue: endothelin system and
end-organ damage. Clin Sci. 2010;119:453–463. Live Link N/A
85 Shao D, Park JE, Wort SJ. The role of endothelin-1 in the pathogenesis
of pulmonary arterial hypertension. Pharmacol Res.
86 Frasch HF, Marshall C, Marshall BE. Endothelin-1 is elevated in
monocrotaline pulmonary hypertension.Am J Physiol. 1999;
276:L304–L310. Live Link N/A
87 Miyauchi T, Yorikane R, Sakai S, Sakurai T, Okada M, Nishikibe
M, et al. Contribution of endogenous endothelin-1 to the progression
of cardiopulmonary alterations in rats with monocrotalineinduced
pulmonary hypertension. Circ Res. 1993;73:887–897.
88 Jasmin JF, Cernacek P, Dupuis J. Activation of the right ventricular
endothelin (ET) system in the monocrotaline model of pulmonary
hypertension: response to chronic ETA receptor blockade.
Clin Sci. 2003;105:647–653. Live Link N/A
89 Shubeita HE, McDonough PM, Harris AN, Knowlton KU,
Glembotski CC, Brown JH, et al. Endothelin induction of inositol
phospholipid hydrolysis, sarcomere assembly, and cardiac gene
expression in ventricular myocytes. J Biol Chem. 1990;265:
90 Hill NS, Warburton RR, Pietras L, Klinger JR. Nonspecific endothelin-
receptor antagonist blunts monocrotaline-induced pulmonary
hypertension in rats. J Appl Physiol. 1997;83: 1209–1215. Live Link N/A
91 Jasmin JF, Lucas M, Cernacek P, Dupuis J. Effectiveness of a
nonselective ETA/B and a selective ETA antagonist in rats with
monocrotaline-induced pulmonary hypertension. Circulation.
92 Tilton RG, Munsch CL, Sherwood SJ, Chen SJ, Chen YF, Wu C,
et al. Attenuation of pulmonary vascular hypertension and cardiac
hypertrophy with sitaxsentan sodium, an orally active ET(A) receptor
antagonist. Pulm Pharmacol Ther. 2000;13:87–97. Live Link N/A
93 Bonvallet ST, Zamora MR, Hasunuma K, Sato K, Hanasato N,
Anderson D, et al. BQ123, an ETA-receptor antagonist, attenuates
hypoxic pulmonary hypertension in rats. Am J Physiol.
1994;266:H1327–H1331. Live Link N/A
94 Sato K, Morio Y, Morris KG, Rodman DM, McMurtry IF.
Mechanism of hypoxic pulmonary vasoconstriction involves ETA
receptor-mediated inhibition of KATP channel. Am J Physiol.
2000;278:L434–L442. Live Link N/A
95 Nishida M, Eshiro K, Okada Y, Takaoka M, Matsumura Y. Roles
of endothelin ETA and ETB receptors in the pathogenesis of
monocrotaline-induced pulmonary hypertension.J Cardiovasc
Pharmacol. 2004;44:187–191. Live Link N/A
96 Fujita K, Matsumura Y, Kita S, Miyazaki Y, Hisaki K, Takaoka
M, et al. Role of endothelin-1 and the ETA receptor in the maintenance
of deoxycorticosterone acetate-salt-induced hypertension.
Br J Pharmacol. 1995;114:925–930.
97 Larivière R, Thibault G, Schiffrin EL. Increased endothelin-1
content in blood vessels of deoxycorticosterone acetate-salt hypertensive
but not in spontaneously hypertensive rats. Hypertension.
98 Larivière R, Day R, Schiffrin EL. Increased expression of endothelin-
1 gene in blood vessels of deoxycorticosterone acetatesalt
hypertensive rats. Hypertension. 1993;21:916–920.
99 Barton M, d’Uscio LV, Shaw S, Meyer P, Moreau P, Lüscher TF.
ETA receptor blockade prevents increased tissue endothelin-1,
vascular hypertrophy, and endothelial dysfunction in salt-sensitive
hypertension. Hypertension. 1998;31(pt2):499–504. Live Link N/A
100 Li JS, Larivière R, Schiffrin EL. Effect of a nonselective endothelin
antagonist on vascular remodeling in deoxycorticosterone
acetate-salt hypertensive rats: evidence for a role of endothelin
in vascular hypertrophy. Hypertension. 1994;24:183–188.
101 Fujita K, Matsumura Y, Miyazaki Y, Takaoka M, Morimoto S.
Effects of the endothelin ETA-receptor antagonist FR139317 on
development of hypertension and cardiovascular hypertrophy in
deoxycorticosterone acetate-salt hypertensive rats. Jpn J Pharmacol.
102 Matsumura Y, Hashimoto N, Taira S, Kuro T, Kitano R, Ohkita
M, et al. Different contribution of endothelin-A and endothelin-B
receptors in the pathogenesis of deoxycorticosterone acetatesalt–
induced hypertension in rats. Hypertension. 1999;33:
103 Pollock DM, Pollock JS. Evidence for endothelin involvement in
the response to high salt. Am J Physiol Renal Physiol. 2001;
281:F144–F150. Live Link N/A
104 Clozel M, Breu V. The role of ETB receptors in normotensive and
Endothelin in Cardiovascular Diseases 313
hypertensive rats as revealed by the non-peptide selective ETB
receptor antagonist Ro 46-8443. FEBS Lett. 1996;383:42–45.
105 Hashimoto N, Kuro T, Fujita K, Azuma S, Matsumura Y. Endothelin
ETB receptor-mediated action on systemic and renal hemodynamics
and urine formation in deoxycorticosterone acetatesalt-
induced hypertensive rats. Biol Pharm Bull. 1998;21:
106 Bush TL, Barrett-Connor E. Noncontraceptive estrogen use and
cardiovascular disease. Epidemiol Rev. 1985;7:89–104. Live Link N/A
107 Godsland IF, Wynn V, Crook D, Miller NE. Sex, plasma lipoproteins,
and atherosclerosis: prevailing assumptions and outstanding
questions. Am Heart J. 1987;114:1467–1503.
108 Stampfer MJ, Colditz GA, Willett WC, Manson JE, Rosner B,
Speizer FE, et al. Postmenopausal estrogen therapy and cardiovascular
disease. Ten-year follow-up from the nurses’ health
study. N Engl J Med. 1991;325:756–762. Live Link N/A
109 Farhat MY, Lavigne MC, Ramwell PW. The vascular protective
effects of estrogen. FASEB J. 1996;10:615–624.
110 Mendelsohn ME, Karas RH. The protective effects of estrogen
on the cardiovascular system. N Engl J Med. 1999;340:
111 Walsh BW, Schiff I, Rosner B, Greenberg L, Ravnikar V, Sacks
FM. Effects of postmenopausal estrogen replacement on the
concentrations and metabolism of plasma lipoproteins. N Engl J
112 Grady D, Rubin SM, Petitti DB, Fox CS, Black D, Ettinger B,
et al. Hormone therapy to prevent disease and prolong life in
postmenopausal women. Ann Intern Med. 1992;117:1016–1037.
113 Tolbert T, Oparil S. Cardiovascular effects of estrogen. Am J
114 Miller AP, Chen YF, Xing D, Feng W, Oparil S. Hormone replacement
therapy and inflammation interactions in cardiovascular
disease. Hypertension. 2003;42:657–663. Live Link N/A
115 Florian M, Freiman A, Magder S. Treatment with 17-β-estradiol
reduces superoxide production in aorta of ovariectomized rats.
116 Hulley S, Grady D, Bush T, Furberg C, Herrington D, Riggs B,
et al. Randomized trial of estrogen plus progestin for secondary
prevention of coronary heart disease in postmenopausal women.
117 Rossouw JE, Anderson GL, Prentice RL, LaCroix AZ,
Kooperberg C, Stefanick ML, et al. Risks and benefits of estrogen
plus progestin in healthy postmenopausal women: principal
results from the women’s health initiative randomized controlled
trial. JAMA. 1998;288:321–333. Live Link N/A
118 Akishita M, Kozaki K, Eto M, Yoshizumi M, Ishikawa M, Toba
K, et al. Estrogen attenuates endothelin-1 production by bovine
endothelial cells via estrogen receptor. Biochem Biophys Res
119 Bilsel AS, Moini H, Tetik E, Aksungar F, Kaynak B, Ozer A.
17β-Estradiol modulates endothelin-1 expression and release in
human endothelial cells. Cardiovasc Res. 2000;46:579–584. Live Link N/A
120 Tan Z, Wang TH, Yang D, Fu XD, Pan JY. Mechanisms of
17β-estradiol on the production of ET-1 in ovariectomized rats.
121 Miyauchi T, Yanagisawa M, Iida K, Ajisaka R, Suzuki N, Fujino
M, et al. Age and sex-related variation of plasma endothelin-1
concentration in normal and hypertensive subjects. Am Heart J.
122 Best PJ, Berger PB, Miller VM, Lerman A. The effect of estrogen
replacement therapy on plasma nitric oxide and endothelin-1
levels in postmenopausal women. Ann Intern Med. 1998;128:
123 Maeda S, Tanabe T, Miyauchi T, Otsuki T, Sugawara J, Iemitsu
M, et al. Aerobic exercise training reduces plasma endothelin-1
concentration in older women. J Appl Physiol. 2003;95:
336–341. Live Link N/A
124 Polderman KH, Stehouwer CD, van Kamp GJ, Schalkwijk CG,
Gooren LJ. Modulation of plasma endothelin levels by the menstrual
cycle. Metabolism. 2000;49:648–650. Live Link N/A
125 Carbonne B, Mignot TM, Tsatsaris V, Ferré F. Changes in plasma
and amniotic fluid endothelin levels during pregnancy: facts or
artifacts? Eur J Obstet Gynecol Reprod Biol. 1998;76:15–19. Live Link N/A
126 Polderman KH, Stehouwer CD, van Kamp GJ, Dekker GA,
Verheugt FW, Gooren LJ. Influence of sex hormones on plasma
endothelin levels. Ann Intern Med. 1993;118:429–432.
127 Ergul A, Shoemaker K, Puett D, Tackett RL. Gender differences
in the expression of endothelin receptors in human saphenous
veins in vitro. J Pharmacol Exp Ther. 1998;285:511–517.
128 Nuedling S, van Eickels M, Alléra A, Doevendans P, Meyer R,
Vetter H, et al. 17β-Estradiol regulates the expression of endothelin
receptor type B in the heart. Br J Pharmacol. 2003;140: 195–201. Live Link N/A
129 David FL, Carvalho MH, Cobra AL, Nigro D, Fortes ZB,
Rebouças NA, et al. Ovarian hormones modulate endothelin-1
vascular reactivity and mRNA expression in DOCA-salt hypertensive
rats. Hypertension. 2001;38:692–696. Live Link N/A
130 David FL, Montezano AC, Rebouças NA, Nigro D, Fortes ZB,
Carvalho MH, et al. Gender differences in vascular expression of
endothelin and ETA/ETB receptors, but not in calcium handling
mechanisms, in deoxycorticosterone acetate-salt hypertension.
Braz J Med Biol Res. 2002;35:1061–1068. Live Link N/A
131 Pedersen SH, Nielsen LB, Pedersen NG, Nilas L, Ottesen B.
Hormone therapy modulates ET(A) mRNA expression in the
aorta of ovariectomised New Zealand White rabbits. Gynecol
Endocrinol. 2009;25:175–182. Live Link N/A
132 Pedersen SH, Nielsen LB, Mortensen A, Nilas L, Ottesen B.
Progestin oppose the effects of estradiol on the endothelin-1 receptor
type B in coronary arteries from ovariectomized hyperlipidemic
rabbits. Menopause. 2008;15:503–510. Live Link N/A
133 Kitada K, Yui N, Koyama M, Kimura K, Suzuki R, Tanaka R,
et al. Endothelin ETB receptor is involved in sex differences in
the development of balloon injury-induced neointimal formation.
J Pharmacol Exp Ther. 2011;336:533–539.
References from
Br J Pharmacol. 2008 March; 153(6): 1105–1119.
GO TO
http://onlinelibrary.wiley.com/doi/10.1038/sj.bjp.0707516/references
CLICK on any reference on [PubMed] for all references on the following list:
- Abassi ZA, Tate JE, Golomb E, Keiser HR. Role of neutral endopeptidase in the metabolism of endothelin. Hypertension. 1992;20:89–95. [PubMed]
- Ahn D, Ge YQ, Stricklett PK, Gill P, Taylor D, Hughes AK, et al. Collecting duct-specific knockout of endothelin-1 causes hypertension and sodium retention. J Clin Invest. 2004;114:504–511.[PMC free article] [PubMed]
- Ahn K, Sisneros AM, Herman SB, Pan SM, Hupe D, Lee C, et al. Novel selective quinazoline inhibitors of endothelin converting enzyme-1. Biochem Biophys Res Commun. 1998;243:184–190. [PubMed]
- Amiri F, Virdis A, Neves MF, Iglarz M, Seidah NG, Touyz RM, et al. Endothelium-restricted over-expression of human endothelin-1 causes vascular remodeling and endothelial dysfunction.Circulation. 2004;110:2233–2240. [PubMed]
- Anand I, McMurray J, Cohn JN, Konstam MA, Notter T, Quitzau K, et al. Long-term effects of darusentan on left-ventricular remodelling and clinical outcomes in the Endothelin(A) Receptor antagonist Trial in Heart failure (EARTH): randomised, double-blind, placebo-controlled trial.Lancet. 2004;364:347–354. [PubMed]
- Anggard E, Galton S, Rae G, Thomas R, McLoughlin L, DeNucci G, et al. The fate of radioiodinated endothelin-1 and endothelin-3 in the rat. J Cardiovasc Pharmacol. 1989;13:S46–S49. [PubMed]
- Arai H, Hori S, Aramori I, Ohkubo H, Nakanishi S. Cloning and expression of a cDNA-encoding an endothelin receptor. Nature. 1990;348:730–732. [PubMed]
- Aramori I, Nirei H, Shoubo M, Sogabe K, Nakamura K, Kojo H, et al. Subtype selectivity of a novel endothelin antagonist, FR139317, for the two endothelin receptors in transfected Chinese hamster ovary cells. Mol Pharmacol. 1993;43:127–131. [PubMed]
- Asai Y, Nonaka N, Suzuki SI, Nishio M, Takahashi K, Shima H, et al. TMC-66, a new endothelin converting enzyme inhibitor produced by Streptomyces sp. A5008. J Antibiot. 1999;52:607–612.[PubMed]
- Bagnall AJ, Kelland NF, Gulliver-Sloan F, Davenport AP, Gray GA, Yanagisawa M, et al. Deletion of endothelial cell endothelin B receptors does not affect blood pressure or sensitivity to salt.Hypertension. 2006;48:286–293. [PubMed]
- Barnes K, Brown C, Turner AJ. Endothelin-converting enzyme—ultrastructural localization and its recycling from the cell surface. Hypertension. 1998;31:3–9. [PubMed]
- Barst RJ, Langleben D, Frost A, Horn EM, Oudiz R, Shapiro S, et al. Sitaxsentan therapy for pulmonary arterial hypertension. Am J Respir Crit Care Med. 2004;169:441–447. [PubMed]
- Barst RJ, Rich S, Widlitz A, Horn EM, McLaughlin V, McFarlin J. Clinical efficacy of sitaxsentan, an endothelin-A receptor antagonist, in patients with pulmonary arterial hypertension—open-label pilot study. Chest. 2002;121:1860–1868. [PubMed]
- Battistini B, Berthiaume N, Kelland NF, Webb DJ, Kohan DE. Profile of past and current clinical trials involving endothelin receptor antagonists: the novel ‘-sentan’ class of drug. Exp Biol Med.2006;231:653–695.
- Battistini B, Daull P, Jeng AY. CGS 35601, a triple inhibitor of angiotensin converting enzyme, neutral endopeptidase and endothelin converting enzyme. Cardiovasc Drug Rev. 2005;23:317–330.[PubMed]
- Bloch KD, Hong CC, Eddy RL, Shows TB, Quertermous T. cDNA cloning and chromosomal assignment of the endothelin-2 gene—vasoactive intestinal contractor peptide is rat endothelin-2.Genomics. 1991;10:236–242. [PubMed]
- Bonderman D, Nowotny R, Skoro-Sajer N, Jakowitsch J, Adlbrecht C, Klepetko A, et al. Bosentan therapy for inoperable chronic thromboembolic pulmonary hypertension. Chest. 2005;128:2599–2603. [PubMed]
- Burrell KM, Molenaar P, Dawson PJ, Kaumann AJ. Contractile and arrhythmic effects of endothelin receptor agonists in human heart in vitro: blockade with SB 209670. J Pharmacol Exp Ther.2000;292:449–459. [PubMed]
- Cardillo C, Kilcoyne CM, Waclawiw M, Cannon RO, III, Panza JA. Role of endothelin in the increased vascular tone of patients with essential hypertension. Hypertension. 1999;33:753–758. [PubMed]
- Cernacek P, Franchi L, Dupuis O, Rouleau JL, Levy M. Radioreceptor assay of an endothelin A receptor antagonist in plasma and urine. Clin Chem. 1998;44:1666–1673. [PubMed]
- Channick RN, Simonneau G, Sitbon O, Robbins IM, Frost A, Tapson VF, et al. Effects of the dual endothelin-receptor antagonist bosentan in patients with pulmonary hypertension: a randomised placebo-controlled study. Lancet. 2001;358:1119–1123. [PubMed]
- Clozel M, Breu V, Gray GA, Kalina B, Loffler BM, Burri K, et al. Pharmacological characterization of bosentan, a new potent orally-active nonpeptide endothelin receptor antagonist. J Pharmacol Exp Ther. 1994;270:228–235. [PubMed]
- Clozel M, Ramuz H, Clozel JP, Breu V, Hess P, Loffler BM, et al. Pharmacology of tezosentan, new endothelin receptor antagonist designed for parenteral use. J Pharmacol Exp Ther. 1999;290:840–846. [PubMed]
- Cody WL, Doherty AM, He JX, Depue PL, Rapundalo ST, Hingorani GA, et al. Design of a functional hexapeptide antagonist of endothelin. J Med Chem. 1992;35:3301–3303. [PubMed]
- Cotter G, Kaluski E, Stangl K, Pacher R, Richter C, Milo-Cotter O, et al. The hemodynamic and neuro-hormonal effects of low doses of tezosentan (an endothelin A/B receptor antagonist) in patients with acute heart failure. Eur J Heart Fail. 2004;6:601–609. [PubMed]
- Cotter G, Kiowski W, Kaluski E, Kobrin I, Milovanov O, Marmor A, et al. Tezosentan (an intravenous endothelin receptor A/B antagonist) reduces peripheral resistance and increases cardiac power therefore preventing a steep decrease in blood pressure in patients with congestive heart failure.Eur J Heart Fail. 2001;3:457–461. [PubMed]
- Cowburn PJ, Cleland JGF, McArthur JD, MacLean MR, McMurray JJV, Dargie HJ. Short-term haemodynamic effects of BQ-123, a selective endothelin ETA-receptor antagonist, in chronic heart failure. Lancet. 1998;352:201–202.
- Davenport AP. International union of pharmacology. XXIX. Update on endothelin receptor nomenclature. Pharmacol Rev. 2002;54:219–226. [PubMed]
- Davenport AP, Maguire JJ. Is endothelin-induced vasoconstriction mediated only by ET(A) receptors in humans. Trends Pharmacol Sci. 1994;15:9–11. [PubMed]
- DeLombaert S, Blanchard L, Stamford LB, Tan J, Wallace EM, Satoh Y, et al. Potent and selective non-peptidic inhibitors of endothelin-converting enzyme-1 with sustained duration of action. J Med Chem. 2000;43:488–504. [PubMed]
- DeLombaert S, Ghai RD, Jeng AY, Trapani AJ, Webb RL. Pharmacological profile of a non-peptidic dual inhibitor of neutral endopeptidase-24.11 and endothelin-converting enzyme. Biochem Biophys Res Commun. 1994;204:407–412. [PubMed]
- Dhaun N, Goddard J, Webb DJ. The endothelin system and its antagonism in chronic kidney disease.J Am Soc Nephrol. 2006;17:943–955. [PubMed]
- Dickstein K, De Voogd HJ, Miric MP, Willenbrock R, Mitrovic V, Pacher R, et al. Effect of single doses of SLV306, an inhibitor of both neutral endopeptidase and endothelin-converting enzyme, on pulmonary pressures in congestive heart failure. Am J Cardiol. 2004;15:237–239.
- Dieterle W, Mann J, Kutz K. Pharmacokinetics and pharmacodynamics of the ETA-selective endothelin receptor antagonist SPP301 in healthy human subjects. J Clin Pharmacol. 2004;44:59–66. [PubMed]
- Doherty AM, Cody WL, He JX, Depue PL, Cheng XM, Welch KM, et al. In-vitro and in-vivo studies with a series of hexapeptide endothelin antagonists. J Cardiovasc Pharmacol. 1993;22:S98–S102.
- D’Orleans-Juste P, Labonte J, Bkaily G, Choufani S, Plante M, Honore JC. Function of the endothelin(B) receptor in cardiovascular physiology and pathophysiology. Pharmacol Ther.2002;95:221–238. [PubMed]
- Dupuis J, Jasmin JF, Prie S, Cernacek P. Importance of local production of endothelin-1 and of the ETB receptor in the regulation of pulmonary vascular tone. Pulm Pharmacol Ther. 2000;13:135–140. [PubMed]
- Emoto N, Yanagisawa M. Endothelin-converting enzyme-2 is a membrane-bound, phosphoramidon-sensitive metalloprotease with acidic pH optimum. J Biol Chem. 1995;270:15262–15268. [PubMed]
- Fattinger K, Funk C, Pantze M, Weber C, Reichen J, Stieger B, et al. The endothelin antagonist bosentan inhibits the canalicular bile salt export pump: a potential mechanism for hepatic adverse reactions. Clin Pharmacol Ther. 2001;69:223–231. [PubMed]
- Fernandez-Patron C, Radomski MW, Davidge ST. Vascular matrix metalloproteinase-2 cleaves big endothelin-1 yielding a novel vasoconstrictor. Circ Res. 1999;85:906–911. [PubMed]
- Ferro CJ, Spratt JC, Haynes WG, Webb DJ. Inhibition of neutral endopeptidase causes vasoconstriction of human resistance vessels in vivo. Circulation. 1998;97:2323–2330. [PubMed]
- Firth JD, Ratcliffe PJ. Organ distribution of the 3 rat endothelin messenger RNAs and the effects of ischemia on renal gene expression. J Clin Invest. 1992;90:1023–1031. [PMC free article] [PubMed]
- Frelin C, Ladoux A, Marsault R, Vigne P. Functional-properties of high-affinity and low-affinity receptor subtypes for endothelin-3. J Cardiovasc Pharmacol. 1991;17:S131–S133. [PubMed]
- Fukuroda T, Ozaki S, Ihara M, Ishikawa K, Yano M, Nishikibe M. Synergistic inhibition by BQ-123 and BQ-788 of endothelin-1-induced contractions of the rabbit pulmonary artery. Br J Pharmacol.1994;113:336–338. [PMC free article] [PubMed]
- Furchgott RF, Zawadzki JV. The obligatory role of endothelial-cells in the relaxation of arterial smooth-muscle by acetylcholine. Nature. 1980;288:373–376. [PubMed]
- Galié N, Badesch D, Oudiz R, Simonneau G, McGoon MD, Keogh AM, et al. Ambrisentan therapy for pulmonary arterial hypertension. J Am Coll Cardiol. 2005;46:529–535. [PubMed]
- Galié N, Hinderliter AL, Torbicki A, Fourme T, Simonneau G, Pulido T, et al. Effects of the oral endothelin-receptor antagonist bosentan on echocardiographic and Doppler measures in patients with pulmonary arterial hypertension. J Am Coll Cardiol. 2003;41:1380–1386. [PubMed]
- Gariepy CE, Ohuchi T, Williams SC, Richardson JA, Yanagisawa M. Salt-sensitive hypertension in endothelin-B receptor-deficient rats. J Clin Invest. 2000;105:925–933. [PMC free article][PubMed]
- Gavras H, Brunner HR, Turini GA, Kershaw GR, Tifft CP, Cuttelod S, et al. Antihypertensive effect of oral angiotensin converting-enzyme inhibitor SQ 14225 in man. N Engl J Med. 1978;298:991–995.[PubMed]
- Gavras I, Gavras H. Angiotenisn II—possible adverse effects on arteries, heart, brain and kidney: experimental, clinical and epidemiological evidence The Renin–Angiotensin System 1993. Gower Medical Publishing: London; In: Robertson JIS and Nicholls MG (eds).
- Gayraud M. Raynaud’s phenomenon. Joint bone. Spine. 2007;74:1–8.
- Ge YQ, Bagnall A, Stricklett PK, Strait K, Webb DJ, Kotelevtsev Y, et al. Collecting duct-specific knockout of the endothelin B receptor causes hypertension and sodium retention. Am J Physiol Renal Physiol. 2006;291:F1274–F1280. [PubMed]
- Gillespie MN, Owasoyo JO, McMurtry IF, O’Brien RF. Sustained coronary vasoconstriction provoked by a peptidergic substance released from endothelial-cells in culture. J Pharmacol Exp Ther.1986;236:339–343. [PubMed]
- Goddard J, Eckhart C, Johnston NR, Cumming AD, Rankin AJ, Webb DJ. Endothelin A receptor antagonism and angiotensin-converting enzyme inhibition are synergistic via an endothelin B receptor-mediated and nitric oxide-dependent mechanism. J Am Soc Nephrol. 2004a;15:2601–2610. [PubMed]
- Goddard J, Johnston NR, Hand MF, Cumming AD, Rabelink TJ, Rankin AJ, et al. 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. 2004b;109:1186–1193. [PubMed]
- Grandjean T, Rivier JL. Cardio-circulatory effects of beta-adrenergic blockade in organic heart disease. Comparison between propranolol and CIBA 39 089-Ba. Br Heart J. 1968;30:50–59.[PMC free article] [PubMed]
- Gray GA, Webb DJ. The endothelin system and its potential as a therapeutic target in cardiovascular disease. Pharmacol Ther. 1996;72:109–148. [PubMed]
- Gregan B, Jurgensen J, Papsdorf G, Furkert J, Schaefer M, Beyermann M, et al. Ligand-dependent differences in the internalization of endothelin A and endothelin B receptor heterodimers. J Biol Chem. 2004;279:27679–27687. [PubMed]
- Harada H, Kazami J, Watanuki S, Tsuzuki R, Sudoh K, Fujimori A, et al. Ethenesulfonamide and ethanesulfonamide derivatives, a novel class of orally active endothelin-A receptor antagonists.Bioorg Med Chem. 2001;9:2955–2968. [PubMed]
- Harrison VJ, Corder R, Anggard EE, Vane JR. Evidence for vesicles that transport endothelin-1 in bovine aortic endothelial cells. J Cardiovasc Pharmacol. 1993;22:S57–S60.
- Haynes WG, Ferro CJ, O’Kane KPJ, Somerville D, Lomax CC, Webb DJ. Systemic endothelin receptor blockade decreases peripheral vascular resistance and blood pressure in humans. Circulation.1996;93:1860–1870. [PubMed]
- Haynes WG, Webb DJ. Endothelin—a long-acting local constrictor hormone. Br J Hosp Med.1992;47:340–349. [PubMed]
- Haynes WG, Webb DJ. Contribution of endogenous generation of endothelin-1 to basal vascular tone.Lancet. 1994;344:852–854. [PubMed]
- Hickey KA, Rubanyi G, Paul RJ, Highsmith RF. Characterization of a coronary vasoconstrictor produced by cultured endothelial cells. Am J Physiol. 1985;248:C550–C556. [PubMed]
- Hirata Y, Yoshimi H, Takagi Y, Kanno K, Eguchi S, Marumo F. Interaction of endothelin isopeptides with vascular endothelin-1 receptor. Biomed Res—Tokyo. 1990;11:195–198.
- Hoang MV, Turner AJ. Novel activity of endothelin-converting enzyme: hydrolysis of bradykinin.Biochem J. 1997;327:23–26. [PMC free article] [PubMed]
- Humbert M, Cabane J. Successful treatment of systemic sclerosis digital ulcers and pulmonary arterial hypertension with endothelin receptor antagonist bosentan. Rheumatology. 2003;42:191–193.
- Ihara M, Ishikawa K, Fukuroda T, Saeki T, Funabashi K, Fukami T, et al. In vitro biological profile of a highly potent novel endothelin (et) antagonist BQ-123 selective for the ET(A) receptor. J Cardiovasc Pharmacol. 1992;20:S11–S14.
- Ikura T, Sawamura T, Shiraki T, Hosokawa H, Kido T, Hoshikawa H, et al. cDNA cloning and expression of bovine endothelin-converting enzyme. Biochem Biophys Res Commun.1994;203:1417–1422. [PubMed]
- Inoue A, Yanagisawa M, Kimura S, Kasuya Y, Miyauchi T, Goto K, et al. The human endothelin family—3 structurally and pharmacologically distinct isopeptides predicted by 3 separate genes. Proc Natl Acad Sci USA. 1989;86:2863–2867.
- Ishikawa K, Ihara M, Noguchi K, Mase T, Mino N, Saeki T, et al. Biochemical and pharmacological profile of a potent and selective endothelin B-receptor antagonist, BQ-788. Proc Natl Acad Sci USA. 1994;91:4892–4896.
- Ivy DD, Parker TA, Abman SH. Prolonged endothelin B receptor blockade causes pulmonary hypertension in the ovine fetus. Am J Physiol Lung Cell Mol Physiol. 2000;279:L758–L765.[PubMed]
- Ivy DD, Yanagisawa M, Gariepy CE, Gebb SA, Colvin KL, McMurtry IF. Exaggerated hypoxic pulmonary hypertension in endothelin B receptor-deficient rats. Am J Physiol Lung Cell Mol Physiol. 2002;282:L703–L712. [PubMed]
- Jandeleit-Dahm KAM. Dual ACE/NEP inhibitors—more than playing the ACE card. J Human Hypertens. 2006;20:478–481. [PubMed]
- Janes RW, Peapus DH, Wallace BA. The crystal structure of human endothelin. Nat Struct Biol.1994;1:311–319. [PubMed]
- Jeng AY, Mulder P, Kwan AL, Battistini B. Non-peptidic endothelin-converting enzyme inhibitors and their potential therapeutic applications. Can J Physiol Pharmacol. 2002;80:440–449. [PubMed]
- Johnström P, Fryer TD, Richards HK, Harris NG, Barret O, Clark JC, et al. Positron emission tomography using 18F-labelled endothelin-1 reveals prevention of binding to cardiac receptors owing to tissue-specific clearance by ET B receptors in vivo. Br J Pharmacol. 2005;144:115–122.[PMC free article] [PubMed]
- Kalra PR, Moon JCC, Coats AJS. Do results of the ENABLE (Endothelin Antagonist Bosentan for Lowering Cardiac Events in Heart Failure) study spell the end for non-selective endothelin antagonism in heart failure. Int J Cardiol. 2002;85:195–197. [PubMed]
- Kaluski E, Kobrin I, Zimlichman R, Marmor A, Krakov O, Milo O, et al. RITZ-5: randomized intravenous tezosentan (an endothelin-A/B antagonist) for the treatment of pulmonary edema—a prospective, multicenter, double-blind, placebo-controlled study. J Am Coll Cardiol. 2003;41:204–210. [PubMed]
- Karet FE, Davenport AP. Localization of endothelin peptides in human kidney. Kidney Int.1996;49:382–387. [PubMed]
- Karne S, Jayawickreme CK, Lerner MR. Cloning and characterization of an endothelin-3 specific receptor (ET(C) receptor) from Xenopus laevis dermal melanophores. J Biol Chem.1993;268:19126–19133. [PubMed]
- Kawai N, Yamamoto T, Yamamoto H, McCarron RM, Spatz M. Functional characterization of endothelin receptors on cultured brain capillary endothelial cells of the rat. Neurochem Int.1997;31:597–605. [PubMed]
- Kelland NF, Webb DJ. Clinical trials of endothelin antagonists in heart failure: a question of dose. Exp Biol Med. 2006;231:696–699.
- Kim NN, Dhir V, Azadzoi KM, Traish AM, Flaherty E, Goldstein I. Pilot study of the endothelin—a receptor selective antagonist BMS-193884 for the treatment of erectile dysfunction. J Androl.2002;23:76–83. [PubMed]
- Kiowski W, Sutsch G, Hunziker P, Muller P, Kim J, Oechslin E, et al. Evidence for endothelin-1-mediated vasoconstriction in severe chronic heart failure. Lancet. 1995;346:732–736. [PubMed]
- Klipper E, Levy N, Gilboa T, Muller L, Meidan R. Identification of a novel alternatively spliced variant endothelin converting enzyme-1 lacking a transmembrane domain. Exp Biol Med. 2006;231:723–728.
- Kloog Y, Ambar I, Sokolovsky M, Kochva E, Wollberg Z, Bdolah A. Sarafotoxin, a novel vasoconstrictor peptide–phosphoinositide hydrolysis in rat heart and brain. Science. 1988;242:268–270.[PubMed]
- Kohan DE. Endothelins in the normal and diseased kidney. Am J Kidney Dis. 1997;29:2–26.[PubMed]
- Korn JH, Mayes M, Cerinic MM, Rainisio M, Pope J, Hachulla E, et al. Digital ulcers in systemic sclerosis—prevention by treatment with bosentan, an oral endothelin receptor antagonist. Arthritis Rheum. 2004;50:3985–3993. [PubMed]
- Krum H, Viskoper RJ, Lacourciere Y, Budde M, Charlon V. The effect of an endothelin-receptor antagonist, bosentan, on blood pressure in patients with essential hypertension. N Engl J Med.1998;338:784–790. [PubMed]
- Kuc RE, Davenport AP. Comparison of endothelin-A and endothelin-B receptor distribution visualized by radioligand binding versus immunocytochemical localization using subtype selective antisera. J Cardiovasc Pharmacol. 2004;44:S224–S226. [PubMed]
- Kukkola PJ, Savage P, Sakane Y, Berry JC, Bilci NA, Ghai RD, et al. Differential structure–activity relationships of phosphoramidon analogs for inhibition of 3 metalloproteases—endothelin-converting enzyme, neutral endopeptidase, and angiotensin-converting enzyme. J Cardiovasc Pharmacol. 1995;26:S65–S68. [PubMed]
- Kumar C, Mwangi V, Nuthulaganti P, Wu HL, Pullen M, Brun K, et al. Cloning and characterization of a novel endothelin receptor from Xenopus heart. J Biol Chem. 1994;269:13414–13420.[PubMed]
- Kurihara Y, Kurihara H, Suzuki H, Kodama T, Maemura K, Nagai R, et al. Elevated blood-pressure and craniofacial abnormalities in mice deficient in endothelin-1. Nature. 1994;368:703–710.[PubMed]
- Lahav R. Endothelin receptor B is required for the expansion of melanocyte precursors and malignant melanoma. Int J Dev Biol. 2005;49:173–180. [PubMed]
- Masaki T. Historical review: endothelin. Trends Pharmacol Sci. 2004;25:219–224. [PubMed]
- McKittrick BA, Stamford AW, Weng XY, Ma K, Chackalamannil S, Czarniecki M, et al. Design and synthesis of phosphinic acids that triply inhibit endothelin converting enzyme, angiotensin converting enzyme and neutral endopeptidase 24.11. Bioorg Med Chem Lett. 1996;6:1629–1634.
- Meidan R, Klipper E, Gilboa T, Muller L, Levy N. Endothelin-converting enzyme-1, abundance of isoforms a-d and identification of a novel alternatively spliced variant lacking a transmembrane domain. J Biol Chem. 2005;280:40867–40874. [PubMed]
- Meyer M, Lehnart S, Pieske B, Schlottauer K, Munk S, Holubarsch C, et al. Influence of endothelin 1 on human atrial myocardium—myocardial function and subcellular pathways. Basic Res Cardiol.1996;91:86–93. [PubMed]
- Mickley EJ, Gray GA, Webb DJ. Activation of endothelin ETA receptors masks the constrictor role of endothelin ETB receptors in rat isolated small mesenteric arteries. Br J Pharmacol. 1997;120:1376–1382. [PMC free article] [PubMed]
- Mihara SI, Nakajima S, Matumura S, Kohnoike T, Fujimoto M. Pharmacological characterization of a potent nonpeptide endothelin receptor antagonist, 97–139. J Pharmacol Exp Ther. 1994;268:1122–1128. [PubMed]
- Mizuguchi T, Nishiyama M, Moroi K, Tanaka H, Saito T, Masuda Y, et al. Analysis of two pharmacologically predicted endothelin B receptor subtypes by using the endothelin B receptor gene knockout mouse. Br J Pharmacol. 1997;120:1427–1430. [PMC free article] [PubMed]
- Molenaar P, O’Reilly G, Sharkey A, Kuc RE, Harding DP, Plumpton C, et al. Characterization and localization of endothelin receptor subtypes in the human atrioventricular conducting system and myocardium. Circ Res. 1993;72:526–538. [PubMed]
- Morita S, Kitamura K, Yamamoto Y, Eto T, Osada Y, Sumiyoshi A, et al. Immunoreactive endothelin in human kidney. Ann Clin Biochem. 1991;28:267–271. [PubMed]
- Morris CD, Rose A, Curwen J, Hughes AM, Wilson DJ, Webb DJ. Specific inhibition of the endothelin A receptor with ZD4054: clinical and pre-clinical evidence. Br J Cancer. 2005;92:2148–2152.[PMC free article] [PubMed]
- Motte S, McEntee K, Naeije R. Endothelin receptor antagonists. Br J Cancer. 2005;92:2148–2152.[PubMed]
- Muller DN, Mullally A, Dechend R, Park JK, Fiebeler A, Pilz B, et al. Endothelin-converting enzyme inhibition ameliorates angiotensin II-induced cardiac damage. Hypertension. 2002;40:840–846.[PubMed]
- Murugesan N, Gu ZX, Spergel S, Young M, Chen P, Mathur A, et al. Biphenylsulfonamide endothelin receptor antagonists. 4. Discovery of N-[[2′-[[(4,5-dimethyl-3-isoxazolyl)amino]sulfonyl]-4-(2-oxazolyl)[1,1′-biphenyl]-2-yl]methyl]-N,3,3-trimethylbutanamide (BMS-207940), a highly potent and orally active ETA selective antagonist. J Med Chem. 2003;46:125–137. [PubMed]
- Murugesan N, Gu ZX, Stein PD, Spergel S, Mathur A, Leith L, et al. Biphenylsulfonamide endothelin receptor antagonists. 2. Discovery of 4′-oxazolyl biphenylsulfonamides as a new class of potent, highly selective ETA antagonists. J Med Chem. 2000;43:3111–3117. [PubMed]
- Mylona P, Cleland JGF. Update of REACH-1 and MERIT-HF clinical trials in heart failure. Eur J Heart Fail. 1999;1:197–200. [PubMed]
- Nguyen QT, Cernacek P, Calderoni A, Stewart DJ, Picard P, Sirois P, et al. Endothelin A receptor blockade causes adverse left ventricular remodeling but improves pulmonary artery pressure after infarction in the rat. Circulation. 1998;98:2323–2330. [PubMed]
- Nunez DJR, Brown MJ, Davenport AP, Neylon CB, Schofield JP, Wyse RK. Endothelin-1 messenger-RNA is widely expressed in porcine and human tissues. J Clin Invest. 1990;85:1537–1541.[PMC free article] [PubMed]
- O’Connor CM, Gattis WA, Adams KF, Hasselblad V, Chandler B, Frey A, et al. Tezosentan in patients’ with acute heart failure and acute coronary syndromes—results of the Randomized Intravenous Tezosentan study (RITZ-4) J Am Coll Cardiol. 2003;41:1452–1457. [PubMed]
- O’Connor CM, Gattis WA, Adams KF, Shah MR, Kobrin I, Frey A, et al. Tezosentan in patients with acute heart failure and acute coronary syndromes: design of the Randomized Intravenous Tezosentan study (RITZ-4) Am Heart J. 2002;144:583–588. [PubMed]
- Ohlstein EH, Nambi P, Lago A, Hay DWP, Beck G, Fong KL, et al. Nonpeptide endothelin receptor antagonists. 6. Pharmacological characterization of SE 217242, a potent and highly bioavailable endothelin receptor antagonist. J Pharmacol Exp Ther. 1996;276:609–615. [PubMed]
- Opgenorth TJ, Adler AL, Calzadilla SV, Chiou WJ, Dayton BD, Dixon DB, et al. Pharmacological characterization of A-127722: an orally active and highly potent ET(A)-selective receptor antagonist. J Pharmacol Exp Ther. 1996;276:473–481. [PubMed]
- Opgenorth TJ, Wuwong JR, Shiosaki K. Endothelin-converting enzymes. FASEB J. 1992;6:2653–2659. [PubMed]
- Packer M, Bristow MR, Cohn JN, Colucci WS, Fowler MB, Gilbert EM, et al. The effect of carvedilol on morbidity and mortality in patients with chronic heart failure. US Carvedilol Heart Failure Study Group. N Engl J Med. 1996;334:1349–1355. [PubMed]
- Pernow J, Bohm F, Johansson BL, Hedin U, Ryden L. Enhanced vasoconstrictor response to endothelin-B-receptor stimulation in patients with atherosclerosis. J Cardiovasc Pharmacol.2000;36:S418–S420. [PubMed]
- Properzi G, Terenghi G, Gu XH, Poccia G, Pasqua R, Francavilla S, et al. Early increase precedes a depletion of endothelin-1 but not of von Willebrand factor in cutaneous microvessels of diabetic patients, a quantitative immunohistochemical study. J Pathol. 1995;175:243–252. [PubMed]
- Quaschning T, Rebhan B, Wunderlich C, Wanner C, Richter CM, Pfab T, et al. Endothelin B receptor-deficient mice develop endothelial dysfunction independently of salt loading. J Hypertens.2005;23:979–985. [PubMed]
- Ramos-Casals M, Brito-Zeron P, Nardi N, Claver G, Risco G, Parraga FD, et al. Successful treatment of severe Raynaud’s phenomenon with bosentan in four patients with systemic sclerosis.Rheumatology. 2004;43:1454–1456.
- Reinhart GA, Preusser LC, Burke SE, Wessale JL, Wegner CD, Opgenorth TJ, et al. Hypertension induced by blockade of ETB receptors in conscious nonhuman primates: role of ETA receptors. Am J Physiol Heart Circ Physiol. 2002;283:H1555–H1561. [PubMed]
- Resink TJ, Hahn AWA, Scott-Burden T, Powell J, Weber E, Buhler FR. Inducible endothelin messenger RNA expression and peptide secretion in cultured human vascular smooth muscle cells.Biochem Biophys Res Commun. 1990;168:1303–1310. [PubMed]
- Riechers H, Albrecht HP, Amberg W, Baumann E, Bernard H, Bohm HJ, et al. Discovery and optimization of a novel class of orally active nonpeptidic endothelin-A receptor antagonists. J Med Chem. 1996;39:2123–2128. [PubMed]
- Roux S, Breu V, Giller T, Neidhart W, Ramuz H, Coassolo P, et al. Ro 61-1790, a new hydrosoluble endothelin antagonist: general pharmacology and effects on experimental cerebral vasospasm. J Pharmacol Exp Ther. 1997;283:1110–1118. [PubMed]
- Rubanyi GM, Polokoff MA. Endothelins—molecular biology, biochemistry, pharmacology, physiology, and pathophysiology. Pharmacol Rev. 1994;46:325–415. [PubMed]
- Rubin LJ, Badesch DB, Barst RJ, Galie N, Black CM, Keogh A, et al. Bosentan therapy for pulmonary arterial hypertension. N Engl J Med. 2002;346:896–903. [PubMed]
- Rubinstein I, Gurbanov K, Hoffman A, Better OS, Winaver J. Differential effect of endothelin-1 on renal regional blood-flow—role of nitric-oxide. J Cardiovasc Pharmacol. 1995;26:S208–S210.[PubMed]
- Russell FD, Davenport AP. Secretory pathways in endothelin synthesis. Br J Pharmacol. 1999;126:391–398. [PMC free article] [PubMed]
- Sakamoto A, Yanagisawa M, Sakurai T, Takuwa Y, Yanagisawa H, Masaki T. Cloning and functional expression of human cDNA for the ETB endothelin receptor. Biochem Biophys Res Commun.1991;178:656–663. [PubMed]
- Sakurai T, Yanagisawa M, Takuwa Y, Miyazaki H, Kimura S, Goto K, et al. Cloning of a cDNA-encoding a non-isopeptide-selective subtype of the endothelin receptor. Nature. 1990;348:732–735. [PubMed]
- Schalcher C, Cotter G, Reisin L, Bertel O, Kobrin I, Guyene TT, et al. The dual endothelin receptor antagonist tezosentan acutely improves hemodynamic parameters in patients with advanced heart failure. Am Heart J. 2001;142:340–349. [PubMed]
- Schiffrin EL. Endothelin: potential role in hypertension and vascular hypertrophy. Hypertension.1995;25:1135–1143. [PubMed]
- Schmidt M, Kroger B, Jacob E, Seulberger H, Subkowski T, Otter R, et al. Molecular characterization of human and bovine endothelin-converting enzyme (ECE-1) FEBS Lett. 1994;356:238–243.[PubMed]
- Shetty SS, Savage P, DelGrande D, De Lombaert S, Jeng AY. Characterization of CGS 31447, a potent and non-peptidic endothelin-converting enzyme inhibitor. J Cardiovasc Pharmacol. 1998;31:S68–S70. [PubMed]
- Shi SJ, Rakugi H, Higashimori K, Jiang BB, Higaki J, Mikami H, et al. Augmentation by converting-enzyme inhibition of accelerated endothelin release from rat mesenteric-arteries following nephrectomy. Biochem Biophys Res Commun. 1994;202:246–251. [PubMed]
- Snyder MJ, Jacobs MR, Grau RG, Wilkes DS, Knox KS. Resolution of severe digital ulceration during a course of bosentan therapy. Ann Intern Med. 2005;142:802–803. [PubMed]
- Spatz M, Kawai N, Merkel N, Bembry J, McCarron RM. Functional properties of cultured endothelial cells derived from large microvessels of human brain. Am J Physiol. 1997;272:C231–C239.[PubMed]
- Spieker LE, Mitrovic V, Noll G, Pacher R, Schulze MR, Muntwyler J, et al. Acute hemodynamic and neurohumoral effects of selective ETA receptor blockade in patients with congestive heart failure. J Am Coll Cardiol. 2000;35:1745–1752. [PubMed]
- Stewart DJ, Levy RD, Cernacek P, Langleben D. Increased plasma endothelin-1 in pulmonary-hypertension—marker or mediator of disease. Ann Intern Med. 1991;114:464–469. [PubMed]
- Strachan FE, Spratt JC, Wilkinson IB, Johnston NR, Gray GA, Webb DJ. Systemic blockade of the endothelin-B receptor increases peripheral vascular resistance in healthy men. Hypertension.1999;33:581–585. [PubMed]
- Tabrizchi R. SLV-306. Solvay. Curr Opin Investig Drugs. 2003;4:329–332.
- Takahashi H, Soma S, Muramatsu M, Oka M, Ienaga H, Fukuchi Y. Discrepant distribution of big endothelin (ET)-1 and ET receptors in the pulmonary artery. Curr Opin Investig Drugs. 2003;4:329–332..[PubMed]
- Takaishi S, Tuchiya N, Sato A, Negishi T, Takamatsu Y, Matsushita Y, et al. B-90063, a novel endothelin converting enzyme inhibitor isolated from a new marine bacterium, Blastobacter sp. SANK 71894. J Antibiot. 1998;51:805–815. [PubMed]
- Torre-Amione G, Durand JB, Nagueh S, Vooletich MT, Kobrin I, Pratt C. A pilot safety trial of prolonged (48
h) infusion of the dual endothelin-receptor antagonist tezosentan in patients with advanced heart failure. Chest. 2001a;120:460–466. [PubMed]
- Torre-Amione G, Young JB, Colucci WS, Lewis BS, Pratt C, Cotter G, et al. Hemodynamic and clinical effects of tezosentan, an intravenous dual endothelin receptor antagonist, in patients hospitalized for acute decompensated heart failure. J Am Coll Cardiol. 2003;42:140–147. [PubMed]
- Torre-Amione G, Young JB, Durand JB, Bozkurt B, Mann DL, Kobrin I, et al. Hemodynamic effects of tezosentan, an intravenous dual endothelin receptor antagonist, in patients with class iii–iv congestive heart failure. Circulation. 2001b;103:973–980. [PubMed]
- Trapani AJ, Beil ME, Bruseo CW, De Lombaert S, Jeng AY. Pharmacological properties of CGS 35066, a potent and selective endothelin-converting enzyme inhibitor, in conscious rats. J Cardiovasc Pharmacol. 2000;36:S40–S43. [PubMed]
- Trapani AJ, Beil ME, Bruseo CW, Savage P, Firooznia F, Jeng AY. CGS 35601 and its orally active prodrug CGS 37808 as triple inhibitors of endothelin-converting enzyme-1, neutral endopeptidase 24.11, and angiotensin-converting enzyme. J Cardiovasc Pharmacol. 2004;44:S211–S215.[PubMed]
- Tschudi MR, Luscher TF. Characterization of contractile endothelin and angiotensin receptors in human resistance arteries—evidence for 2 endothelin and one angiotensin receptor. Biochem Biophys Res Commun. 1994;204:685–690. [PubMed]
- Tsurumi Y, Fujie K, Nishikawa M, Kiyoto S, Okuhara M. Biological and pharmacological properties of highly selective new endothelin-converting enzyme-inhibitor WS79089B isolated fromStreptosporangium roseum no-79089. J Antibiot. 1995a;48:169–174. [PubMed]
- Tsurumi Y, Ueda H, Hayashi K, Takase S, Nishikawa M, Kiyoto S, et al. WS75624-A and WS75624-B, new endothelin-converting enzyme-inhibitors isolated from Saccharothrix sp. no-75624. 1. Taxonomy, fermentation, isolation, physicochemical properties and biological-activities. J Antibiot.1995b;48:1066–1072. [PubMed]
- Umekawa K, Hasegawa H, Tsutsumi Y, Sato K, Matsumura Y, Ohashi N. Pharmacological characterization of a novel sulfonylureid-pyrazole derivative, SM-19712, a potent nonpeptidic inhibitor of endothelin converting enzyme. Jpn J Pharmacol. 2000;84:7–15. [PubMed]
- Vajkoczy P, Meyer B, Weidauer S, Raabe A, Thome C, Ringel F, et al. Clazosentan (AXV-034343), a selective endothelin A receptor antagonist, in the prevention of cerebral vasospasm following severe aneurysmal subarachnoid hemorrhage: results of a randomized, double-blind, placebo-controlled, multicenter phase IIa study. J Neurosurg. 2005;103:9–17. [PubMed]
- Valdenaire O, Lepailleur-Enouf D, Egidy G, Thouard A, Barret A, Vranckx R, et al. A fourth isoform of endothelin-converting enzyme (ECE-1) is generated from an additional promoter—molecular cloning and characterization. Eur J Biochem. 1999;264:341–349. [PubMed]
- von Geldern TW, Tasker AS, Sorensen BK, Winn M, Szczepankiewicz BG, Dixon DB, et al. Pyrrolidine-3-carboxylic acids as endothelin antagonists. 4. Side chain conformational restriction leads to ETB selectivity. J Med Chem. 1999;42:3668–3678. [PubMed]
- Watanabe T, Awane Y, Ikeda S, Fujiwara S, Kubo K, Kikuchi T, et al. Pharmacology of a nonselective ET(A) and ET(B) receptor antagonist, TAK-044 and the inhibition of myocardial infarct size in rats. Br J Pharmacol. 1995;114:949–954. [PMC free article] [PubMed]
- Weber C, Schmitt R, Birnboeck H, Hopfgartner G, vanMarle SP, Peeters PAM, et al. Pharmacokinetics and pharmacodynamics of the endothelin-receptor antagonist bosentan in healthy human subjects. Clin Pharmacol Ther. 1996;60:124–137. [PubMed]
- Whitebread S, Mele M, Kamber B, Degasparo M. Preliminary biochemical characterization of two angiotensin-II receptor subtypes. Biochem Biophys Res Commun. 1989;163:284–291. [PubMed]
- Wu CD, Chan MF, Stavros F, Raju B, Okun I, Mong S, et al. Discovery of TBC11251, a potent, long acting, orally active endothelin receptor-A selective antagonist. J Med Chem. 1997;40:1690–1697.[PubMed]
- Wu CD, Decker ER, Blok N, Bui H, You TJ, Wang JM, et al. Discovery, modeling, and human pharmacokinetics of N-(2-acetyl-4,6-dimethylphenyl)-3-(3,4-dimethylisoxazol-5-sulfamoyl)thiophene-2-carboxamide (TBC3711), a second generation, ETA selective, and orally bioavailable endothelin antagonist. J Med Chem. 2004;47:1969–1986. [PubMed]
- Wypij DM, Nichols JS, Novak PJ, Stacy DL, Berman J, Wiseman JS. Role of mast-cell chymase in the extracellular processing of big-endothelin-1 to endothelin-1 in the perfused rat lung. Biochem Pharmacol. 1992;43:845–853. [PubMed]
- Yanagisawa H, Hammer RE, Richardson JA, Emoto N, Williams SC, Takeda S, et al. Disruption of ECE-1 and ECE-2 reveals a role for endothelin-converting enzyme-2 in murine cardiac development. J Clin Invest. 2000;105:1373–1382. [PMC free article] [PubMed]
- Yanagisawa M, Kurihara H, Kimura S, Tomobe Y, Kobayashi M, Mitsui Y, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature. 1988;332:411–415.[PubMed]
Abbreviations
- BP- blood pressure
- EC – endothelial cell
- ECE – endothelin-converting enzyme
- ET – endothelin
- ET-1 – endothelin-1
- ETA/B – ETA/B receptor
- IMCD – inner-medullary collecting duct
- NEP – neutral endopeptidase
- NO – nitric oxide
- NYHA – New York Heart Association
- PAH- pulmonary arterial hypertension
- SAH- subarachnoid haemorrhage
- SSc – systemic sclerosis
- VSMC – vascular smooth muscle cell
Other Research on ET and Cardiovascular Disease on this Scientific Web Site include:
Cardiovascular Disease (CVD) and the Role of agent alternatives in endothelial Nitric Oxide Synthase (eNOS) Activation and Nitric Oxide Production
Curator and Research Study Originator: Aviva Lev-Ari, PhD, RN, 7/19/2012
Macrovascular Disease – Therapeutic Potential of cEPCs: Reduction Methods for CV Risk
An Investigation of the Potential of circulating Endothelial Progenitor Cells (cEPCs) as a Therapeutic Target for Pharmacological Therapy Design for Cardiovascular Risk Reduction: A New Multimarker Biomarker Discovery
Curator: Aviva Lev-Ari, PhD, RN 7/2/2012
Endothelin Receptors in Cardiovascular Diseases: The Role of eNOS Stimulation
Author: Aviva Lev-Ari, PhD, 10/4/2012
Inhibition of ET-1, ETA and ETA-ETB, Induction of NO production, stimulation of eNOS and Treatment Regime with PPAR-gamma agonists (TZD): cEPCs Endogenous Augmentation for Cardiovascular Risk Reduction – A Bibliography
Curator: Aviva Lev-Ari, 10/4/2012.
Coronary Artery Disease – Medical Devices Solutions: From First-In-Man Stent Implantation, via Medical Ethical Dilemmas to Drug Eluting Stents
Author: Aviva Lev-Ari, PhD, RN, 8/13/2012
This is a remarkable work. It is more comprehensive than any I can find, and it will require several rereads.
Dr. Larry,
Thank you for your comment. It is based on two SEMINAL papers in the UK, 2008 and in Japan 2012. I am the curator who wrote about ET in 2006, as per the references on ET on this Scientific Web Site.
[…] Clinical Trials Results for Endothelin System: Pathophysiological role in Chronic Heart Failure, Acu… (pharmaceuticalintelligence.com) […]
[…] http://pharmaceuticalintelligence.com/2012/10/19/clinical-trials-results-for-endothelin-system-patho… […]
[…] Clinical Trials Results for Endothelin System: Pathophysiological role in Chronic Heart Failure, Acu… […]
[…] Clinical Trials Results for Endothelin System: Pathophysiological role in Chronic Heart Failure, Acu… […]
[…] Curator: Aviva Lev-Ari, PhD, RN http://pharmaceuticalintelligence.com/2012/10/19/clinical-trials-results-for-endothelin-system-patho… […]
[…] Clinical Trials Results for Endothelin System: Pathophysiological role in Chronic Heart Failure, Acu… […]
PUT IT IN CONTEXT OF CANCER CELL MOVEMENT
The contraction of skeletal muscle is triggered by nerve impulses, which stimulate the release of Ca2+ from the sarcoplasmic reticuluma specialized network of internal membranes, similar to the endoplasmic reticulum, that stores high concentrations of Ca2+ ions. The release of Ca2+ from the sarcoplasmic reticulum increases the concentration of Ca2+ in the cytosol from approximately 10-7 to 10-5 M. The increased Ca2+ concentration signals muscle contraction via the action of two accessory proteins bound to the actin filaments: tropomyosin and troponin (Figure 11.25). Tropomyosin is a fibrous protein that binds lengthwise along the groove of actin filaments. In striated muscle, each tropomyosin molecule is bound to troponin, which is a complex of three polypeptides: troponin C (Ca2+-binding), troponin I (inhibitory), and troponin T (tropomyosin-binding). When the concentration of Ca2+ is low, the complex of the troponins with tropomyosin blocks the interaction of actin and myosin, so the muscle does not contract. At high concentrations, Ca2+ binding to troponin C shifts the position of the complex, relieving this inhibition and allowing contraction to proceed.
Figure 11.25
Association of tropomyosin and troponins with actin filaments. (A) Tropomyosin binds lengthwise along actin filaments and, in striated muscle, is associated with a complex of three troponins: troponin I (TnI), troponin C (TnC), and troponin T (TnT). In (more ) Contractile Assemblies of Actin and Myosin in Nonmuscle Cells
Contractile assemblies of actin and myosin, resembling small-scale versions of muscle fibers, are present also in nonmuscle cells. As in muscle, the actin filaments in these contractile assemblies are interdigitated with bipolar filaments of myosin II, consisting of 15 to 20 myosin II molecules, which produce contraction by sliding the actin filaments relative to one another (Figure 11.26). The actin filaments in contractile bundles in nonmuscle cells are also associated with tropomyosin, which facilitates their interaction with myosin II, probably by competing with filamin for binding sites on actin.
Figure 11.26
Contractile assemblies in nonmuscle cells. Bipolar filaments of myosin II produce contraction by sliding actin filaments in opposite directions. Two examples of contractile assemblies in nonmuscle cells, stress fibers and adhesion belts, were discussed earlier with respect to attachment of the actin cytoskeleton to regions of cell-substrate and cell-cell contacts (see Figures 11.13 and 11.14). The contraction of stress fibers produces tension across the cell, allowing the cell to pull on a substrate (e.g., the extracellular matrix) to which it is anchored. The contraction of adhesion belts alters the shape of epithelial cell sheets: a process that is particularly important during embryonic development, when sheets of epithelial cells fold into structures such as tubes.
The most dramatic example of actin-myosin contraction in nonmuscle cells, however, is provided by cytokinesisthe division of a cell into two following mitosis (Figure 11.27). Toward the end of mitosis in animal cells, a contractile ring consisting of actin filaments and myosin II assembles just underneath the plasma membrane. Its contraction pulls the plasma membrane progressively inward, constricting the center of the cell and pinching it in two. Interestingly, the thickness of the contractile ring remains constant as it contracts, implying that actin filaments disassemble as contraction proceeds. The ring then disperses completely following cell division.
Figure 11.27
Cytokinesis. Following completion of mitosis (nuclear division), a contractile ring consisting of actin filaments and myosin II divides the cell in two.
http://www.ncbi.nlm.nih.gov/books/NBK9961/
This is good. I don’t recall seeing it in the original comment. I am very aware of the actin myosin troponin connection in heart and in skeletal muscle, and I did know about the nonmuscle work. I won’t deal with it now, and I have been working with Aviral now online for 2 hours.
I have had a considerable background from way back in atomic orbital theory, physical chemistry, organic chemistry, and the equilibrium necessary for cations and anions. Despite the calcium role in contraction, I would not discount hypomagnesemia in having a disease role because of the intracellular-extracellular connection. The description you pasted reminds me also of a lecture given a few years ago by the Nobel Laureate that year on the mechanism of cell division.
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette
I actually consider this amazing blog , âSAME SCIENTIFIC IMPACT: Scientific Publishing –
Open Journals vs. Subscription-based « Pharmaceutical Intelligenceâ, very compelling plus the blog post ended up being a good read.
Many thanks,Annette