Published in final edited form as:
Pharmacol Res. 2017 November ; 125(Pt A): 57–71. doi:10.1016/j.phrs.2017.05.020.
Renin Angiotensin Aldosterone Inhibition in the Treatment of Cardiovascular Disease
Carlos M Ferrario, MD1 and Adam E. Mullick, PhD2
1Department of Surgery, Wake Forest University Health Science, Winston Salem, NC 27157
2Cardiovascular Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010
Abstract
A collective century of discoveries establishes the importance of the renin angiotensin aldosterone system in maintaining blood pressure, fluid volume and electrolyte homeostasis via autocrine, paracrine and endocrine signaling. While research continues to yield new functions of angiotensin II and angiotensin-(1-7), the gap between basic research and clinical application of these new findings is widening. As data accumulates on the efficacy of angiotensin converting enzyme inhibitors and angiotensin II receptor blockers as drugs of fundamental importance in the treatment of cardiovascular and renal disorders, it is becoming apparent that the achieved clinical benefits is suboptimal and surprisingly no different than what can be achieved with other therapeutic interventions. We discuss this issue and summarize new pathways and mechanisms effecting the synthesis and actions of angiotensin II. The presence of renin-independent non- canonical pathways for angiotensin II production are largely unaffected by agents inhibiting renin angiotensin system activity. Hence, new efforts should be directed to develop drugs that can effectively block the synthesis and/or action of intracellular angiotensin II. Improved drug penetration into cardiac or renal sites of disease, inhibiting chymase –the primary angiotensin II forming enzyme in the human heart–, and/or inhibiting angiotensinogen synthesis would all be more effective strategies to inhibit the system. Additionally, given the role of angiotensin II in the maintenance of renal homeostatic mechanisms, any new inhibitor should possess greater selectivity of targeting pathogenic angiotensin II signaling processes and thereby limit inappropriate inhibition.
Graphical abstract
Address for Correspondence: Carlos M Ferrario, MD, Department of Surgery, Wake Forest University Health Science Center, Medical Center Blvd., Winston Salem, NC 27157, cferrari@wakehealth.edu, Phone: 336-918-8442.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
Ferrario and Mullick Page 2
Keywords
angiotensin-(1-12); angiotensin-(1-7); angiotensin converting enzyme inhibitors; angiotensin receptor blockers; hypertension; heart failure
1.0 Introduction
In the face of profound advances in the biomedical sciences, preventive health care approaches, education, and socioeconomic improvements, deaths due to cardiovascular disease (CVD) are globally on the rise when compared with mortality rates 25 years ago [1]. In the USA, rates of middle-aged white non-Hispanic men and women have shown a significant rise in all-cause mortality [2] that may be attributed in part to the impact of societal stresses on cardiovascular health. Roth et al. [1] documented recently that ischemic heart disease remains the leading cause of death worldwide accounting for almost half of the increase in the number of cardiovascular deaths. The 2017 American Heart Association (AHA) update shows heart failure (HF) rates increasing to 800,000 new cases over the last five years [3]. The AHA’s update report specifies that the number of people with heart failure (HF) diagnosis is expected to rise by 46 percent by 2030 [3], which computes to eight million HF patients. Disentangling the numerous factors influencing these statistics is outside the objectives of this review article. These data, however, underline the existence of a real disconnect among scientific advances in cardiovascular disease mechanisms, the development of new medicines to halt disease processes, and the void for a more aggressive implementation of health care resources across all segments of the American population.
An emerging illustration of the disconnect in translating scientific knowledge with continual discovery processes in drug development is applicable to heart medicines where a significant impasse has occurred since the introduction of angiotensin II (Ang II) receptor blockers (ARBs) [4, 5]. While efforts to surpass the benefits achieved with ARBs through the development of the direct renin inhibitor –aliskiren– did not meet expectations [6–8], the combination of valsartan with an inhibitor of endopeptidase 24-11 (neprilysin) has shown promise in reducing HF progression when compared with the standard of care [8, 9].
Nevertheless, even this new angiotensin receptor-neprilysin combination inhibitor) showed limited superiority over conventional angiotensin converting enzyme (ACE) inhibitor therapy [9].
Ferrario and Mullick Page 3
Since the first demonstration of the impact of ACE inhibition on hypertensive patients with the orally active agent teprotide [10], accelerated research efforts by academic investigators and pharmaceutical companies through a 27 years』 period brought about the introduction of eleven drugs targeting ACE, nine compounds acting as selective orally active blockers of the Ang II type 1-receptor (ARBs), and one direct renin inhibitor [11]. Mineralocorticoid receptor antagonists were added to this armamentarium to improve efficacy in situations such as primary aldosteronism and resistant hypertension [12]. These remarkable clinical translation achievements are the corollary of a voluminous research literature documenting the importance of the renin angiotensin aldosterone system (RAAS) as a key contributor to the development and progression of cardiovascular disease [13]. Ang II modulation of cellular inter- and intracellular signaling mechanisms participating in growth, cell-to-cell communication, immunity, lipid peroxidation, and insulin resistance plays a fundamental role in cardiovascular pathology.
It is not the scope of this article to review the literature on Ang II physiological mechanisms. Likewise, this review does not intend to address the clinical literature that justifies the use of RAAS inhibitors as the cornerstone of cardiovascular disease therapies. This review addresses emerging limitations behind the translational disconnect between what has been learned in the laboratory setting regarding Ang II roles in cardiovascular disease progression and the real-world effectiveness of RAAS inhibitors in halting or reversing clinical events. Instead, we will underscore one potential explanation for the fact that clinical outcome studies using a direct renin inhibitor, ACE inhibitors or ARBs have demonstrated benefits that are less than what would have been expected. Those wishing to delve more deeply on the arguments presented in this review can access the enclosed references [14–21].
2.0 Biotransformation Pathways of Angiotensins. Current Concepts
Since the original recognition of renin, angiotensinogen (AGT), and ACE as the critical proteins contributing to Ang II formation, impressive advances in the understanding of the role of this system in both physiology and pathology unmasked that this hormonal system was constituted by a network of proteins, peptides, and receptors more complex than anticipated.
Figure 1 shows the currently accepted multi-pathway processing steps leading to the generation of the main functionally active peptides Ang-(1-9) [22, 23], Ang II, Ang-(1-7) [24, 25], and their respective derivatives Ala1-Ang II [26] and alamandine [Ala1-Ang-(1-7)] [27]. Critical angiotensins forming substrates are AGT and the shorter AGT derived sequences, angiotensin-(1-25) (BigAng 25) [28] and angiotensin-(1-12) [Ang-(1-12)] [29]. Processing of AGT by renal renin may be particularly important within the circulation and the renal medulla [30–32], whereas non-renin pathways may be more actively engaged in forming Ang II in tissues such as the heart [33, 34]. The discovery of the two alternate Ang II-forming substrates, Ang-(1-25) and Ang-(1-12), invigorated the concept that mechanisms of angiotensin peptide formation in organs and cells may not follow the biochemical processing documented in the circulation. In keeping with this interpretation, tissue Ang II formation from either Ang-(1-25) or Ang-(1-12) appears to be primarily due to chymase (EC 3.4.21.39), a member of the serine family of proteinases that are abundantly present in mast
Ferrario and Mullick Page 4
cells [35–38]. There are no definitive studies addressing the enzymatic pathway accounting for Ang-(1-12) and Ang-(1-12) generation from AGT. A preliminary report from our laboratory implicates kallikrein or an aprotinin-sensitive enzyme for the cleavage of AGT into Ang-(1-12) [39]. While the potential contribution of Ang-(1-25) as an Ang II forming substrate is confined to one published paper where the substrate was identified from the urine of normal subjects [28], more extensive research on Ang-(1-12) has led to the conclusion that this extended form of angiotensin I (Ang I) is truly a functionally
endogenous Ang II generating substrate that is predominantly expressed in tissues serving as an intracrine source for direct Ang II production [40]. Ang-(1-12) expression and content is augmented in the heart of spontaneous hypertensive rats (SHR) [41], the left ventricle of rats expressing the human AGT gene [42], and the enlarged left atrial appendage of subjects with left heart disease and left atrial enlargement [43]. Additional studies document Ang-(1-12) as a source for Ang II actions in modulating baroreflexes [44], central sympathetic outflow [45], and augmenting cardiac contractility through altering the electrophysiological properties of the myocytes [46]. Additional studies provide evidence for a tonic role of Ang- (1-12) in the central regulation of blood pressure through the demonstration that cerebrospinal fluid (CSF) delivery of an Ang-(1-12) antibody reduced the magnitude of hypertension in transgenic rats expressing the Ren-2 gene in their genome [47] while Ang- (1-12) injected into the arcuate nuclei of the hypothalamus caused a robust hypertensive response associated with increased splanchnic nerve sympathetic discharges [48]. As illustrated in Figure 1, Ang II formation from Ang-(1-12) represents a non-renin dependent pathway [34, 49]. Ang-(1-12) affinity for chymase is several orders of magnitude higher than for ACE [40]; studies of Ang-(1-12) metabolism in plasma membranes isolated from human left atrial [50] or left ventricular tissues [51] showed chymase as the primary Ang II forming enzyme. A robust but often neglected literature implicates chymase as the primary Ang II forming enzyme from Ang I in humans [35–37, 52]. This is of considerable importance as it explains in part the relatively lesser efficacy of ACE inhibitors in suppressing Ang II levels during chronic treatment with these agents [53, 54].
A third enzymatic pathway is constituted by the mono carboxypeptidase angiotensin converting enzyme 2 (ACE2) which shows less than 50% amino acid homology with ACE and is insensitive to blockade with ACE inhibitors [55–57]. ACE2 hydrolyzes Ang I into Ang-(1-9) and Ang II into Ang-(1-7) [58]. ACE2 role as a pivot point regulating the formation of the cardio-renal protective peptides Ang-(1-9) and Ang-(1-7), in part through lowering the concentrations of Ang I and Ang II, respectively, may represent a critical step in modulating Ang II actions in the etiopathogenesis of cardiovascular disease [59].
Since the original description of Ang-(1-7) biological activity [60], multiple studies now confirm the existence of an intrinsic arm within the RAAS in which the heptapeptide acts to oppose the vasoconstrictor, trophic, proliferative, and pro-thrombotic Ang II actions.
Seminal studies by Ferrario and collaborators on Ang-(1-7) provided the basis for the inclusion of Ang-(1-7) as a counterregulatory arm of the RAAS (reviewed in [61]) and identified neprilysin (E.C. 3.4.24.11) [62], prolyl endopeptidase (E.C. 3.4.21.26), and later ACE2 (EC:3.4.17.23) as Ang-(1-7)-forming enzymes from Ang I [63, 64]. Definitive studies demonstrating reduced Ang-(1-7) in essential hypertension [65], the contribution of Ang-
(1-7) to the antihypertensive action of ACE inhibitors [66], and its participation in reparative
Ferrario and Mullick Page 5
left ventricular remodeling during post-myocardial infarction are reviewed elsewhere [25, 39, 59, 61].
Ang II and Ang-(1-7) biological activity is mediated through the coupling of the peptides to the G-couple receptor proteins -AT1, AT2, mas, and the mas-related G protein-coupled receptor-member D (MrgD) receptors [67–72]. Although AT1 receptors exhibit a wide tissue distribution, expression and function of these proteins have been best studied in cardiovascular, renal, and the peripheral and the central nervous systems. Non-peptide ARBs act as specific AT1 receptor blockers in the vascular system, adrenal cortex, and the kidneys although losartan, its active metabolite EXP3174 [73, 74] and irbesartan [75, 76] also act as partial antagonists of thromboxane A2 receptors (TxA2). Likewise, telmisartan behaves as a partial agonist of the peroxisome proliferator-activated receptor gamma (PPAR-γ) [77] while the uricosuric effects of losartan through blockade of the renal tubular uric acid transporter [78–80] are credited with broadening the therapeutic effectiveness of this agent and compensating for the mild hyperuricemia associated with chronic diuretic therapy. Ang II binding to AT1 receptors stimulates Gq/11 and Gi/o proteins leading to Ca2+ signal [81]. Signaling mechanisms mediated by Ang II binding to AT2 receptors are less defined.
Carey’s review [82] reports that binding of Ang II to AT2 receptors results in the activation
of phosphotyrosine phosphatase and consequent inhibition of kinase p 42 and p 44 mitogen- activated protein (MAP) kinases. Ang-(1-7) binding to the mas receptor leads to inhibition of serum-stimulated ERK1/2 MAP kinase activity [83, 84]. In an experimental HF model, Ang-(1-7) improved intracellular Ca++ mobilization through activation of the mas-related
nitric oxide-bradykinin pathway [85]. More recently, elegant experiments by Tezner et al.
[72] demonstrate that Ang-(1-7) binding to the MrgD receptor activates the Gαs/AC/cAMP pathway leading to an increase in PKA activity and CREB phosphorylation. Importantly, this study provided definitive evidence that PD123319 is not a selective AT2 receptor antagonist [72].
3.0 Benefits and Pitfalls of Renin Angiotensin Aldosterone System Inhibition
With more than half a century of experience in the use of therapies that are directed to inhibit Ang II actions, ACE inhibitors and ARBs have become an indispensable prescription for the treatment of essential hypertension, progression of chronic renal disease, post- myocardial infarction, congestive heart failure, and type 2 diabetes mellitus. The inclusion of aldosterone receptor antagonists to this therapeutic armamentarium is generally restricted to the management of resistant hypertension [86–88]. A summary of adverse side effects associated with these agents are documented in Table 1. Given that Ang II and Ang-(1-7) should be viewed as pleiotropic hormones, it is not surprising that pharmacological approaches designed to suppress the ACE/Ang II/AT1 axis or activate the ACE2/Ang-
(1-7)/mas axis are being explored in a variety of other human diseases [89].
The wide use of these medications has provided the opportunity to evaluate these agents』 true benefits in reducing coronary heart disease, stroke, HF, cardiovascular and all-cause mortality in randomized controlled outcome trials and post-surveillance studies. An
Ferrario and Mullick Page 6
emerging literature questions, however, whether the benefit of target organ protection and reduction of cardiovascular risk associated with blockade of RAAS components is achieving what would have been predicted from the voluminous literature that establishes that dysregulation of this system is an obligatory fundamental factor of the mechanisms accounting for target organ damage and cardiovascular-mediated clinical events. The idea that excessive RAAS hyperactivity is optimally inhibited by combining different RAAS inhibitors [90–93] has instead led to worsening rather than improvement of clinical outcomes despite apparent benefits in reduction of surrogate measures like hypertension and proteinuria [94–96]. Preclinical studies also demonstrate a critical role of Ang II in the pathogenesis of atherosclerosis [97], however, limited clinical evidence of the antiatherogenic effects of Ang II blockade is available [15, 98–100]. Likewise, a report that included 24 trials with 198,275 patient years of follow-up found that RAAS inhibition showed superiority only when compared to placebo but not with active controls in patients with stable coronary heart disease [15]. This conclusion is reminiscent of the report made by the Blood Pressure Lowering Treatment Trialists』 Collaboration group who reported no evidence for differences among drug classes for major cardiovascular events [101]. While many arguments may be posited as to the reasons for this disconnect, we have proposed that incomplete blockade of Ang II pathological actions is due to the failure of RAAS inhibitors to access intracellular proteins accounting for canonical and non-canonical mechanisms of Ang II formation [19–21, 39].
4.0 Direct Renin Inhibitors: An incomplete story
AGT cleavage by renin, considered the rate-limiting step in Ang II generation, is a logical step to inhibit RAAS. Pepstatin was the first synthetic renin inhibitor to be considered for potential therapeutic actions [102]. These early studies led to the development of a series of first-generation agents that required parenteral administration and showed limited activity in healthy volunteers [103, 104]. Although orally active compounds like enalkiren, remikiren and zankiren were developed their clinical potential were not pursued as they had poor bioavailability, short half-life, and weak antihypertensive activity [105]. A more sophisticated structure-based design approach led to the synthesis of aliskiren fumarate, the first non-peptide direct renin inhibitor with favorable pharmacokinetic properties [106].
Aliskiren was approved in 2007 by the FDA and the Europe Middle East and Africa (EMEA) regulatory bodies for the treatment of hypertension, either as a monotherapy or in combination with other antihypertensive agents [107].
A key therapeutic rationale for aliskiren was to achieve complete RAAS inhibition in patients receiving ACE inhibitors or ARBs. It is well documented that following ACE inhibitor treatment there can be reactive increases in compensatory pathways leading to plasma Ang II returning to pretreatment levels or higher [108]. Likewise, aldosterone breakthrough, observed with ACE or ARB therapy [109–111], may be in part due to adrenal AT2 receptor activation [112, 113] or Ang III non-Ang II receptor stimulation [114].
Increases in plasma renin concentration following therapy with ACE inhibitors or ARBs
may in itself be a cardiovascular risk factor [115–117].
Ferrario and Mullick Page 7
Indeed, with aliskiren added to the antihypertensive standard of care, the compensatory rise in plasma renin activity was neutralized suggesting that better RAAS suppression could be now realized [118] and further reductions of blood pressure were observed when combined with other antihypertensives such as ACE inhibitors or ARBs [91, 118, 119]. These data corroborated and extended earlier studies of Menard and colleagues [90, 120] who demonstrated incomplete RAAS suppression with a single agent. Additionally, such data highlight the complexity of RAAS and the existence of non-canonical pathways that contribute to RAAS.
Better end organ protection in patients with kidney disease, HF, and atherosclerosis, independent of blood pressure reductions, is a significant unmet medical need. Hence, ineffective and/or insufficient inhibition of tissue Ang II activity at cardiovascular and renal sites was the biologic rationale used to evaluate aliskiren as an add-on therapy in patients with HF or diabetic kidney disease [121]. The concept that tissue or local Ang II production exerts pathophysiologic effects independent of systemic RAAS had been well established before the development of aliskiren [122]. Implicit in this view was that ACE inhibitors (or ARBs) exert some of their protective effects in tissue independent of blood pressure lowering, however, such effects are incomplete [123].
Initial clinical studies evaluating aliskiren as an add-on to ACE inhibitor or ARB therapy demonstrated beneficial effects on surrogate endpoints, such as reductions in blood pressure and proteinuria. Additionally, adverse effects such as hyperkalemia, renal impairment or hypotension were sufficiently low as to not cause concern. Despite these early positive results, larger trials with longer treatment duration and hard cardiovascular or renal outcomes showed no clinical utility, in fact, adverse effects were observed [124–126]. An experimental study suggested that the antihypertensive response produced by concomitant blockade of both renin and Ang II triggers significant renal injury due to loss of autoregulatory capacity [127]. Currently the use of aliskiren in combination to an ACE inhibitor or ARB is proscribed with no further plans of development.
An understanding of the clinical failures of aliskiren has remained elusive. It is tempting to speculate that combination treatment of aliskiren and ARB revealed a unique and deleterious profile of renin inhibition [128]. Preclinical studies have demonstrated renal accumulation of aliskiren [129, 130], which could promote a discordance between renal tissue versus circulating RAAS suppression [131]. In support of this concept, VTP-27999, a more potent renin inhibitor, produced greater renin inhibition that paradoxically resulted in extrarenal RAAS stimulation in healthy volunteers [132]. Consistent with these data are the observations that aliskiren results in a ~2-fold greater increase in plasma renin levels compared to equivalent blood pressure lowering doses of an ACE inhibitor or an ARB [107].
It may be concluded that the therapeutic index of renin inhibition is no better compared to an ACE inhibitor or ARB, and that all therapies have the propensity to inappropriately suppress RAAS when used in combination. Given the strength of the evidence of synergy when combining RAAS blockers to beneficially impact hypertension, HF and kidney disease [90– 93], a hypothesis can be formulated that the therapeutic goal of RAAS blockade should be to maximally limit pathogenic Ang II signaling without disrupting renal homeostatic processes.
Ferrario and Mullick Page 8
Hence, that aggressive RAAS inhibition has not demonstrated clinical utility may be a failure to maximally inhibit RAAS without provoking renal complications. As previously discussed, non-canonical pathways of pathogenic Ang II generation and signaling underscore the challenges of effective RAAS inhibition as combinations of RAAS inhibitors could in theory offer better efficacy, but also produce a significant renal safety hazard.
Despite the evidence that intrarenal Ang II contributes to hypertension or renal disease [31, 133], renal homeostatic functions required for maintenance of renal blood flow and GFR are in part regulated by Ang II [134, 135]. That all RAAS components have been identified throughout the entire nephron is suggestive of an essential role of intrarenal RAAS for volume and electrolyte homeostasis [136].
It is well established that RAAS blockage presents a particular risk-benefit analysis in regards to renal impairment [137], and that such risks are amplified in the setting of salt- restriction/dehydration [138–140], in the elderly [141], or in patients with diabetes, heart or renal disease [142]. Because Ang II has divergent activities on promoting hypertension and disease as well as maintaining essential renal homeostatic mechanisms, the conflicting results obtained to-date underscore the need to better understand the role of intrarenal RAAS. Such insights could lead the way to the development of a superior inhibitors capable of preserving essential renal Ang II activities while inhibiting pathogenic Ang II signaling.
5.0 Angiotensin Converting Enzyme Inhibitors
ACE inhibitors are the mainstay of cardiovascular disease treatment and are prescribed for the treatment of hypertension, myocardial infarction, left ventricular dysfunction, HF, diabetic mellitus, and renal insufficiency [143]. The relatively benign side effect profile of ACE inhibitors are summarized in Table 1. The potency of ACE inhibition is influenced by the drug’s affinity to interact with the zinc (Zn++) ligand of the ACE [144]. There are three distinct chemical classes of ACE inhibitors. Sulfhydryl containing ACE inhibitors (i.e., captopril) bind strongly with the Zn++ ligand but disulphide formation limits their half-life. Drugs containing a carboxyl group constitute most ACE inhibitors. These agents bind to side chains of the enzyme within the active moiety for improved potency and duration of action [144]. A third group of drugs composed of phosphorus-containing ACE inhibitors is represented by fosinopril [145, 146]. Although direct ACE inhibitors comparisons among the available agents are rare, lisinopril, perindopril, and quinapril may be favored because of their prolonged half-life and greater lipophilicity [147]. As reviewed elsewhere [18], evidence from large clinical trials document a beneficial effect of ACE inhibitors in retarding HF progression and repeat hospitalizations, adverse cardiac remodeling and increased survival post myocardial infarction, vascular disease secondary to atherosclerosis or hypertension, and chronic renal disease including diabetic nephropathy.
While the antihypertensive effects of these drugs are proven and many studies have confirmed their ability to acutely suppress the conversion of Ang I into Ang II, a consistent suppression of tissue or plasma Ang II concentrations has not been demonstrated [53].
While an ACE escape mechanism has been suggested to account for the recovery of Ang II [148], no sufficient attention has been paid to the multiple studies showing that chymase,
Ferrario and Mullick Page 9
rather than ACE, is the main Ang II-forming enzyme from either Ang I or Ang-(1-12) in humans in tissues like the heart and vascular wall [20, 35–39, 52]. Studies by Wei et al.
[149] in ACE knockout mice and those of Ahmad et al. [40, 50, 51, 150] and Ferrario et al.
[42] in rats expressing the human AGT gene demonstrate a critical role of chymase as a tissue Ang II forming enzyme. Alternate enzymatic pathways may also contribute to Ang II generation. According to Husain et al. [151] and Dive et al. [152] characterization of a novel inhibitor of the N-terminal active site of ACE suggested the existence of a Ang II-forming metalloproteinase enzyme distinct from both ACE and chymase. No further data exist as to the potential chemical nature and function of this enzyme.
Differences in expression and Ang II-forming enzymes affinity as a function of tissue compartment and species may explain the finding that increased plasma Ang II levels are present in 50 percent of ACE inhibitor treated subjects [54]. The apparent limited ability of ACE inhibitors to maintain a consistent suppression of plasma and tissue Ang II levels in part explains why re-analysis of clinical benefits associated with ACE inhibition from robust meta-analysis of the currently reported clinical trials documents a higher than expected residual risk of clinical events [18]. As reviewed in Reyes et al. [21], the relative risk reduction (RR) achieved with ACE inhibitors for the treatment of hypertension, post- myocardial infarction, and HF averaged 27% in major clinical trials. In keeping with these findings, Baker’s et al. [14] analysis of 7 clinical studies that included 32,559 participants showed a RR of 0.87 [95% CI, 0.81 to 0.94] and 0.83 [CI, 0.73 to 0.94] for total mortality and nonfatal myocardial infarction, respectively. That means that the absolute benefit of these treatments benefited no more than 17% of treated patients. Similar conclusions were reported by van Vark et al. [153] who found the hazard ratio for ACE inhibitor treated all- cause mortality from hypertensive subjects to average 0.90 (CI, 0.84–0.97). Therefore, the residual risk associated with ACE inhibitors across all examined trials remains unacceptably high. A detailed analysis of the impact of ACE inhibitors on all-cause mortality, cardiovascular mortality, myocardial infarction, stroke, and the composite of myocardial infarction and stroke showed that the number of patients needed to be treated to prevent one event ranged from 67 to 409 [17]. Zanchetti and collaborators have provided a detailed analysis of the significance of the residual risk in treated hypertensive patients [154].
6.0 Angiotensin Receptor Blockers
The theoretical rationale for a more specific blockade of Ang II pathological actions through the binding of non-peptide antagonists to the AT1 receptor accounted for the introduction of ARBs to the antihypertensive prescription armamentarium in 1995 [67]. These drugs ability to achieve their clinical goals bypassing the limitations of an ACE escape phenomena and non-ACE sources of Ang II formation was viewed as a definitive advantage. This is despite their effect to dramatically increase blood Ang II levels due to blockade of AT1-mediated receptor internalization [13]. The proven ability of ARBs to elicit minimal or essentially non-clinically relevant side effects has been an added advantage of these agents (Table 1).
All eight approved ARBs are selective ligands of AT1 receptors and in pharmacological studies showed significant potency in their ability to cause a rightward shift of the dose- response curve to Ang II [67]. Differences in the ability of ARBs to reduce the maximal response in-vitro has led to their subclassification as possessing surmountable or
Ferrario and Mullick Page 10
insurmountable antagonism [155]. The clinical impact of these pharmacological ligand- interactions in terms of the drugs ability to achieve lasting antihypertensive effects remains unproven. Large clinical trials utilizing losartan [156–158], valsartan [159–163], candesartan [164–167], irbesartan [168, 169], telmisartan [94, 96] and olmesartan [170] have proven their ability to control blood pressure in hypertensive patients, reduce stroke-risk, decrease HF hospitalizations, and improve the prognosis of diabetes nephropathy. A composite of key clinical trials RR and confidence intervals is documented in Figure 2. From the analysis of the 26 trials presented in Figure 2, the pooled RR reduction averaged 0.93 (C.I. 0.84 – 1.01). These data demonstrate a relatively small benefit of ARB in the prevention or treatment of clinical events or superiority over either ACE inhibitors or other therapies. On the other hand, only the Losartan Intervention For Endpoint Reduction in Hypertension (LIFE) trial suggests a potential for superiority over other treatments. The extensive data gathered from the investigation of 9,124 hypertensive patients with electrocardiographic evidence of left ventricular hypertrophy in the LIFE trial documented that for the comparable antihypertensive actions of the two active treatment arms, those randomized to the losartan- based therapy showed a 13% lower RR of primary cardiovascular events and 25% smaller RR of fatal and non-fatal strokes [157]. Similarly, superior outcomes over conventional therapy were documented in the Reduction of Endpoints in NIDDM with the Angiotensin II Antagonist Losartan (RENAAL) Study [156] and the Irbesartan Diabetic Nephropathy Trial (IDNT) [168] in subjects with type 2 diabetic nephropathy (Figure 2). As concluded by Düsing [18, 171], improved safety and enhanced tolerability over other therapies may be the greatest clinical advantage of this drug class. However, some have questioned whether ARBs show equivalent efficacy when compared with ACE inhibitors [172]. In our minds, such lackluster and/or nonexistent efficacy improvements beyond ACE inhibitors underscores the role of the RAAS in the etiopathogenesis of cardiovascular disease. The small effect of ARBs is suggestive of intracellular sites of Ang II activity that would be largely unopposed [19, 20, 173–175]. That ARBs induce compensatory pathways that increase circulating Ang II as well as increased expression of downstream metabolites like Ang-(1-7) [13, 59] underscore the complexity of understanding the mechanisms that limit their efficacy.
7.0 Mineralocorticoid Receptor Antagonists (MRA)
The introduction of spironolactone, the first mineralocorticoid receptor antagonist (MRA), culminated efforts of multiple investigators who in the 1950’s were preoccupied with exploring the relationship between aldosterone and sodium metabolism [176, 177]. Cranston et al. [178] first report of the modest antihypertensive actions of spironolactone initiated further interest in exploring the role of aldosterone as a causative mechanism of essential hypertension leading Conn to propose that primary aldosteronism could explain between 15 and 25% of the cause of essential hypertension [179]. Comprehensive updates on the mechanism of action and indications of MRA are published [12, 119]. The side effects associated with the use of these MRAs are shown in Table 1. Both spironolactone and the newer MRA eplerenone are frequently prescribed in combination with other antihypertensive agents for the treatment of resistant hypertension or HF [180, 181]. The increased selectivity for mineralocorticoid receptors (MR) of a new nonsteroidal MRA, finerenone (BAY 94-8662), is the current focus of an aggressive program seeking to assess
Ferrario and Mullick Page 11
the efficacy of this third generation MRA on HF, chronic kidney disease, and diabetic nephropathy [182–185]. Finerenone acts as a full antagonist of the MR and shows an even accumulation of the drug in the heart and kidney contrasting with the much larger kidney accumulation of spironolactone and eplerenone.
Emerging strategies to counteract the hypertensive and profibrotic actions of aldosterone are being explored through the development of drugs that inhibit the activity of aldosterone synthase, the enzyme that represents the rate limiting step of aldosterone production [119, 186, 187]. The advantage of aldosterone synthase inhibitors (ASI) over MRA is that MRA do not block non-genomic actions of aldosterone [119]. In addition, blockade of aldosterone synthesis obviates the reactive increase in MRA-induced aldosterone [119]. The aldosterone synthase inhibitor, LCI699 has been reported to decrease plasma and urine aldosterone concentrations, increases PRA, and prevents target organ damage [188, 189]. As reviewed by Oparil and Schmieder [119], disappointing results obtained in phase II clinical trials, including minimal blood pressure reductions to an escape phenomena, resulted in discontinuing the first orally active ASI.
8.0 Angiotensin Receptor Neprilysin Inhibitor (ARNI)
The combination of the neprilysin inhibitor sacubitril with valsartan, although a combination therapy receiving much attention due to its effects on natriuretic peptides, is included in this review by virtue of its AT1 receptor blocking component and the potential effects of sacubitril on angiotensin metabolism. While LCZ696 (sacubitril/valsartan) has been approved for the treatment of HF, the vasodilator effects of this combination therapy as an antihypertensive agent remain unappreciated due to its sponsor’s focus on HF. Nevertheless, following the first report of the pharmacological actions of LCZ696 by Gu and colleagues [190], a PubMed search reveals 260 publications on this drug to-date.
Neprilysin, or neutral endopeptidase (NEP), catalyzes the degradation of key peptide hormones that regulate cardiovascular and renal homeostasis. NEP actions in angiotensin metabolism include degradation of Ang II and Ang-(1-7), activities that would be expected to produce opposing effects on vasomotor tone and blood pressure [63, 191]. However, it is the proteolysis of natriuretic peptides (NPs) the mechanism that has garnered much interest. The three members of this family are atrial (ANP), brain or B-type (BNP) and C-type (CNP). ANP and BNP bind to the atrial natriuretic peptide receptor (NPRA), a guanylyl cyclase-coupled receptor located in the vasculature and kidneys. Activation of this receptor increases cGMP leading to vasodilation, natriuresis and diuresis [192, 193]. CNP is specific for NPRB, also a guanylyl cyclase-coupled receptor; however, the significance of NPRB activation in the context of the cardiovascular system is less certain. Expression and release of ANP and BNP from cardiomyocytes is increased during heart wall stress and hypertrophy, common HF features. Their activation promotes blood pressure and volume reductions that act to counteract the increased cardiac wall stress. Such effects represent a counterregulatory system to oppose the actions of Ang II. Therefore, maintaining elevated levels of ANP and BNP via inhibition of NEP are an attractive strategy for combating the undesirable effects of inappropriate RAAS activation.
Ferrario and Mullick Page 12
Candoxatril, the first NEP inhibitor evaluated in the clinic, produced dose-dependent increases in ANP and natriuresis in healthy volunteers [194]. Since these encouraging results did not translate to any benefit in patients with hypertension or heart failure, further drug development was halted [195]. An additional failure of another NEP inhibitor, ecadotril, demonstrated the challenges of targeting NEP [196]. In this study, ecadotril was given at four dose levels in HF patients with systolic dysfunction receiving an ACE inhibitor. In support of target engagement, plasma and urinary cGMP increased in a dose-dependent manner. However, ANP levels were not significantly changed [196]. Surrogates of HF suggested no beneficial effects of treatment; in fact, there was a discordance in mortality of more deaths with ecadotril treatment compared to placebo. Additionally, there was a suggestion of drug-induced aplastic anemia which was hypothesized to be due to toxicity of the thioester group of ecadotril.
NEP has broad proteolytic activity beyond natriuretic peptide regulation. NEP also degrades Ang II, renal Ang-(1-7), bradykinin, substance P, adrenomedullin, glucagon, vasoactive intestinal peptide, and amyloid-β [191]. Therefore, NEP inhibition would be predicted to increase some peptides that would counteract the protective effects of increasing NPs or elicit responses that compromise safety. Hence the rationale of combining a NEP inhibitor with either a ACE inhibitor or an ARB is an appropriate strategy. The first therapy tested with such properties was omapatrilat, which had dual actions to inhibit both ACE and NEP [197–199]. Unlike the previous clinical results with NEP inhibition alone, omapatrilat had potent anti-hypertensive effects that were superior relative to ACE inhibitor treatment [197] and that were accompanied by increased Ang-(1-7) urinary excretion [199]. Additional encouraging signs of improved efficacy compared to the standard of care in HF [200] provided the rationale to test omapatrilat in a large HF outcome trial which demonstrated no benefit compared to enalapril [201]. Worryingly, a reported increase in the occurrence and severity of angioedema halted further drug development. This was considered a consequence of increased bradykinin due to omapatrilat’s inhibition of ACE, NEP and aminopeptidase.
Considering the poor risk to benefit ratio, further clinical development of omapatrilat was discontinued [202].
Combining a NEP inhibitor with an ARB, instead of an ACE inhibitor, had the appeal of having less of an effect to exacerbate the untoward effects associated with ACE inhibitors and led to the development of LCZ696 (Entresto®, sacubitril valsartan), a first-in-class combined NEP inhibitor and ARB. Clinical studies demonstrated LCZ696 to be superior to valsartan for blood pressure lowering [203] and HF amelioration [204, 205] without evidence of adverse safety or tolerability [206]. These encouraging results supported the evaluation of LCZ696 compared to enalapril in an 8,000 patient outcome study in HF with reduced ejection fraction. The Prospective Comparison of the Angiotensin Receptor– Neprilysin Inhibitor [ARNI] with ACEI [Angiotensin-Converting –Enzyme Inhibitor] to Determine Impact on Global Mortality and Morbidity in Heart Failure Trial (PARADIGM- HF) was powered to determine an effect on the primary composite endpoint of death from cardiovascular causes or first hospitalization for HF following 34 months of treatments [207]. However, after a mean patient follow-up of 27 months, the PARADIGM-HF data monitoring committee recommended early termination of the trial for efficacy [207].
LCZ696 reduced the primary composite endpoint by 21.8% and all-cause mortality by 16%.
Ferrario and Mullick Page 13
Such improvements were unprecedented, as they were of the same magnitude of protection observed in the initial studies that established ACE inhibitors as the gold standard in HF treatment [208]. Nevertheless, the authors of this study do not elaborate on the fact that the difference in the reduction of the primary event between HF patients assigned to the enalapril and LCZ696 arms of the study was a mere 4.7% [207]. These effects were consistent across all prespecified subgroups. Although LCZ696 was associated with a higher proportion of patients with hypotension and non-serious angioedema there was a reduced incidence of renal impairment, hyperkalemia, and cough with LCZ696 treatment [207].
Importantly, the occurrence of serious angioedema was unchanged between both groups.
Entresto® was approved by the FDA in July 2015 for the treatment of HF with reduced ejection fraction and recently was given Class I recommendation authorizing its use as the standard of care. Its efficacy in HF with preserved ejection fraction is currently underway in a 4,000+ patient outcome trial (PARAGON-HF). Additionally, the drug sponsor recently announced an effort to evaluate its efficacy and safety in over 40 ongoing and planned clinical trials (FortiHFy program), which would make it the largest industry-sponsored HF clinical program to date. One area of particular concern is any effect of Entresto® to contribute to Alzheimer disease (AD), as animal models demonstrate a role of neprilysin to degrade pathogenic amyloid-β [209]. Current clinical assessments of changes in pathogenic amyloid-β [210] or dementia-related events [211] with Entresto® have not revealed a safety concern; however, longer follow-up studies are still warranted to exclude such concerns.
9.0Alternative Approaches to RAAS Blockade
9.1Angiotensinogen Antisense
Linkage and genetic association studies have suggested an association between AGT and hypertension [212, 213]. Common variants within the AGT promoter region and post- translational redox modifications of AGT have been shown to modulate the substrate and be associated with hypertension and hypertensive-related diseases [214–216]. Experimental studies have demonstrated a relationship between plasma AGT levels and hypertension, with seminal work demonstrating that plasma AGT and blood pressure are elevated in transgenic mice with increasing amounts of AGT [217].
Despite the biological rationale of targeting AGT, the substrate has not been a target of traditional small molecule inhibitor approaches, as downstream proteases and/or receptors are much more 『druggable』. Antisense is especially well-suited for such targets, and given the primacy of AGT as the key RAAS substrate, there were early efforts to antisense AGT. Reports of blood pressure lowering following antisense oligonucleotide (ASO) AGT treatments were initially reported from Phillips and co-workers [218–220]. These early reports used unmodified phosphodiester or phosphorothioate linked oligodeoxynucleotides targeting the AUG start codon to inhibit translation initiation, a commonly used early antisense design method. A single dose administration of ASO was administered into the lateral ventricles of the brain and resulted in relatively modest (~30%) but significant AGT reductions within the hypothalamus [220]. Impressive mean arterial pressure reductions, approaching 40 mm Hg, followed AGT ASO administration. Similar doses of the AGT ASO given IV for systemic exposure did not have any effect on blood pressure [221].
Ferrario and Mullick Page 14
With the success of targeting central AGT expression, methods to enable ASO liver delivery to reduce systemic AGT were described. Various strategies were employed, such as liposomal formulation with viral agglutinins [222], liposomal formulation alone [221], asialoglycoprotein-poly(L)lystine-ASO complexes [223–225] and adeno-associated viral (AAV) based [226]. Collectively these reports demonstrate that ASO-mediated systemic reductions of AGT can reduce blood pressure in hypertensive rats. The complexity of these formulations, challenges of AAV-based gene therapy and the requirement of IV administration are significant barriers to translate these methods into clinical approaches for hypertension management. Additionally, the therapeutic effect of these methods was relatively modest, with reports showing only 40 – 50% maximal reductions of plasma AGT. Major limitations inherent in these methods were (1) the identification of active sequences,
(2) the oligonucleotide chemistry and (3) an understanding of the antisense mechanisms.
As stated earlier, early antisense strategies to inhibit translation initiation by targeting the AUG initiation codon were commonly used [227]. Early on this was an attractive strategy as it only required knowledge of the gene sequence at a limited site. However, more potent methods of gene inhibition had been described that utilized RNase H-mediated RNA cleavage of ASO-RNA duplexes [228, 229]. The advantages of RNase H-dependent approaches were both mechanistic and practical; for example, RNA could be measured (e.g. by PCR) to determine ASO activity. With the ability to use higher-throughput methods to screen for active ASOs, RNase H-active ASOs are amenable to much larger screens of ASOs such that hundreds (or more) of prospective sequences can be evaluated. Such screening is critical for the identification of ASOs with acceptable tolerability and potency. Although RNase-H active ASOs were used in some reports, the screen used to identify active sequences was inadequate. For example, only 3 AGT ASOs were evaluated [222, 223] which is insufficient given our experience.
Another important limitation was the oligonucleotide chemistry. The phosphorothioate (PS) backbone modification described in these reports was critical in the early development of ASOs and allowed for improved nuclease resistance and increased binding to plasma proteins [229]. Further improvements in sugar modifications, applied at the opposite ends of the ASO to create 『gapmers』, have provided the most value in improving the pharmacological properties of ASOs. Such sugar modifications improve potency resulting in AGT reductions of up to 90% [230–232]. Finally, oligonucleotide modifications to elicit argonaute 2-mediated RNA cleavage, used in all small interfering RNA (siRNA) approaches, offers another potent mechanism of inhibiting AGT [233]. Thus, ASO gapmer and siRNA methods offer the most potent mechanisms of RNA-based therapeutics currently available. Early development programs have been described using both platforms [234–236]. Such therapeutic approaches offer a unique means of RAAS blockade as compensatory feedback and non-canonical pathways of Ang II generation and signaling may not limit therapeutic efficacy to the same extent that they do with ACE inhibitors and/or ARBs. Safety studies will be required to ensure that the more complete suppression of Ang II via AGT inhibition does not result in dysregulation of renal Ang II homeostatic processes.
Ferrario and Mullick Page 15
9.2Angiotensin II Vaccines
Endogenous immuno-neutralization of circulating Ang I or Ang II has been studied as an alternate single or adjuvant therapy for hypertension. As reviewed by Oparil and Schmieder [119], vaccines directed to block Ang II have been relatively effective clinically and no evidence has been obtained as to their efficacy and ability to produce sustained antihypertensive responses that are accompanied by reduction in clinical events.
9.3Activators of AT2 receptors or the ACE2/Ang-(1-7)/mas receptor axis
The characterization of ACE2 as a pathway for Ang II degradation and the observation that significant cardiac dysfunction was associated with ACE2 gene suppression in mice [55, 237, 238] allowed skeptic investigators to accept the existence of an endogenous system in which Ang-(1-7) represented an internal feedback mechanism for limiting Ang II hyperactivity [24, 25, 59, 61]. These contributions also stimulated the development of AT2 receptor agonists as a potential approach to compensate for Ang II actions.
Compound 21 (C21) is the first non-peptide agonist of the AT2 receptor that is investigated as a potential antihypertensive compound [239–241]. Vasorelaxant and potent natriuretic actions of C21 in experimental models of hypertension failed to be associated with sustained blood pressure reductions [242–244] unless concomitant AT1 receptor blockade is included in the study. The absence of robust and sustained antihypertensive actions may limit the future clinical development of AT2 receptor agonists.
More intensive approaches have been applied to drugs augmenting the activity of the ACE2/ Ang-(1-7)/mas receptor axis. These approaches focus on facilitating the cardiorenal protective actions of Ang-(1-7) through enhancing Ang II metabolism by ACE2 [245], facilitating conversion of Ang I into Ang-(1-9), or both. Clinical experience with human recombinant ACE2 (hrACE2) is limited to studies in normal healthy volunteers [246] and an ongoing clinical trial involving patients with acute respiratory distress syndrome (Clinical Trial Identifier NCT01597635). In healthy volunteers, administration of hrACE2 was associated with no changes in blood pressure and significant decreases in plasma Ang II levels; changes in plasma Ang-(1-7) levels were inconsistent [246].
Protecting or extending the bioavailability and half-life of Ang-(1-7) is another approach that is being investigated clinically as a critical body of experimental research suggest it as a viable avenue for drug development. Strategies include: a)- Ang-(1-7) encapsulation in hydroxy-propyl-β-cyclodextrin (HP-β-CD/Ang1-7); and b)- preventing Ang-(1-7) enzymatic degradation via inclusion of a thioether bridge to the peptide [247]. Clinical evidence for a beneficial effect of augmenting Ang-(1-7) activity has been negative.
10.0 Summary and Conclusions
Björn Folkow, the notable Swedish physiologist, first called attention to the importance of 「adaptive changes of vascular structure」 as a mechanism contributing to the increased vascular resistance of essential hypertension [248, 249]. The increased vascular resistance reflects an adaptive structural change in precapillary resistance vessels to the increased load. Vessel thickening augments their reactivity to neurohormonal stimuli as well as altering the
Ferrario and Mullick Page 16
ratio between the thickness of the vessel wall and its lumen. The adaptive structural response of resistance vessels to elevated blood pressure may limit their maximal vasodilator response, as documented in experimental [248] and clinical studies [250–253]. The combined contributions of vascular hypertrophic remodeling, increased vascular stiffness and vascular endothelial dysfunction over a long time period may explain the less than optimal efficacy of current treatments with drugs that inhibit the expression or action of Ang
II. On the other hand, the common clinical experience that combining RAAS agents with another agent from a different drug class achieves superior antihypertensive effects suggests otherwise [92]. On an aggregate, laboratory research and genome approaches continue to implicate a close to causal role of excess neurohormonal drive in the pathogenesis of CVD. Pleiotropic Ang II actions in terms of cardiac and vascular remodeling, increased neurogenic drive, stimulation of immune adaptive processes, build-up of radical oxygen species, release of thrombogenic factors, and extracellular matrix remodeling, provides compelling evidence to its contribution to the myriad of etiopathogenetic mechanisms of CVD. In accepting this evidence, we are faced with the issue of what is the cause of the disconnect between the science behind the functions of the RAAS and the clinical outcomes of preventing its blockade.
While discussion of these issues may raise as much ire as disturbing a hornet’s nest, there are indeed two major possible explanations that need to be considered. First, tissue mechanisms associated with the cardiac and vascular adaptive response to hypertension, coronary heart disease and HF may be associated with or caused by activation of intracrine mechanisms of Ang II formation and action [20, 21, 173, 174, 254–257]. Biochemical mechanisms for angiotensins generation and receptor signaling are present within the cellular environment including nuclei mitochondria [258–260]. As reviewed by Kumar et al.
[173] direct evidence for intracrine activity is inferred by the demonstration that intracellular dialysis of Ang II, Ang-(1-7), or renin in cardiac myocytes is associated with changes in junctional conductance. In keeping with this interpretation, intracellular delivery of Ang-
(1-12) altered the excitability of WKY cardiac myocytes, an effect that could be prevented by administration of chymostatin or valsartan [46]. Several studies have documented no changes in cardiac Ang II content during chronic inhibition of ACE or AT1 receptor blockade [19, 20, 261, 262]. We have proposed that these data suggest a failure ACE
inhibitors ARBs and renin inhibitors to reach the intracellular site (s) at which Ang II is generated [20, 21]. Second, multiple studies have documented that chymase rather than ACE is the major Ang II forming enzyme from either Ang I [36, 52, 263] or, more recently, Ang- (1-12) [33, 40, 50]. Thus, pathogenic non-canonical paracrine and intracrine mechanism of Ang II are unaffected by current standards of care [19].
While the clinical importance of these alternate Ang II mechanisms remains to be precisely documented, it is evident to us that a more promising drug development program should be based on creating drugs that can neutralize intracellular formation or action of Ang II. Wei et al. [264] reported improved left ventricle function, decreased adverse cardiac remodeling, and improved survival of hamsters treated with combined ACE and chymase inhibition in a model of post myocardial infarction. In addition, a strong body of experimental evidence demonstrates robust antiarrhythmic effects of chymase inhibition [265, 266] while additional studies reported beneficial effects of chymase inhibition in reversing vascular atherosclerosis
Ferrario and Mullick Page 17
and adverse cardiac remodeling [267–271]. The relevance of chymase in CVD will be tested in an ongoing clinical study evaluating the chymase inhibitor BAY1142524 in a post- myocardial infarction setting (CHIARA MIA 2).
Such pathologic actions of Ang II must be weighed against the essential role of Ang II to preserve renal homeostatic functions. As with any therapy, there is an inherent risk-to- benefit ratio defining the therapeutic index of a RAAS inhibitor. Thus, one of the many challenges of RAAS drug discovery will be to find new methods to inhibit this system, which result in therapies with greater efficacy no compromise in safety.
Acknowledgments
12.0 Funding Sources:
CMF research studies were supported by grant 2P01HL051952 from the National Heart, Blood, Lung Institute of the National Institutes of Health. AEM is an employee of Ionis Pharmaceuticals.
Abbreviations
ACE 2 Angiotensin converting enzyme 2
Ang I Angiotensin I
Ang II Angiotensin II
Ang-(1-12) Angiotensin-(1-12)
Ang-(1-7) Angiotensin-(1-7)
Ang-(1-9) Angiotensin-(1-9)
AGT Angiotensinogen
ANRI Angiotensin Receptor Neprilysin Inhibitor
ASO Antisense oligonucleotide
BigAng 25 Big angiotensin 25
CVD Cardiovascular disease (s)
DRI Direct renin inhibitor
LCZ696 Sacubitril-valsartan
HF Heart Failure
MRA Mineralocorticoid receptor antagonist
MrgD Mas-related G-protein coupled receptor member D
NP Natriuretic peptides
NPRA Atrial natriuretic peptide receptor
Ferrario and Mullick Page 18
NEP Neprilysin
RAAS Renin angiotensin aldosterone system
11.0Literature Cited
1.Roth GA, Forouzanfar MH, Moran AE, Barber R, Nguyen G, Feigin VL, Naghavi M, Mensah GA, Murray CJ. Demographic and epidemiologic drivers of global cardiovascular mortality. N Engl J Med. 2015; 372(14):1333–41. [PubMed: 25830423]
2.Case A, Deaton A. Rising morbidity and mortality in midlife among white non-Hispanic Americans in the 21st century. Proc Natl Acad Sci U S A. 2015; 112(49):15078–83. [PubMed: 26575631]
3.Benjamin EJ, Blaha MJ, Chiuve SE, Cushman M, Das SR, Deo R, de Ferranti SD, Floyd J, Fornage M, Gillespie C, Isasi CR, Jimenez MC, Jordan LC, Judd SE, Lackland D, Lichtman JH, Lisabeth L, Liu S, Longenecker CT, Mackey RH, Matsushita K, Mozaffarian D, Mussolino ME, Nasir K, Neumar RW, Palaniappan L, Pandey DK, Thiagarajan RR, Reeves MJ, Ritchey M, Rodriguez CJ, Roth GA, Rosamond WD, Sasson C, Towfighi A, Tsao CW, Turner MB, Virani SS, Voeks JH, Willey JZ, Wilkins JT, Wu JH, Alger HM, Wong SS, Muntner P. American Heart Association Statistics, S. Stroke Statistics C. Heart Disease and Stroke Statistics-2017 Update: A Report From the American Heart Association. Circulation. 2017
4.Timmermans PB. Angiotensin II receptor antagonists: an emerging new class of cardiovascular therapeutics. Hypertens Res. 1999; 22(2):147–53. [PubMed: 10487332]
5.Timmermans PB, Duncia JV, Carini DJ, Chiu AT, Wong PC, Wexler RR, Smith RD. Discovery of losartan, the first angiotensin II receptor antagonist. J Hum Hypertens. 1995; 9(Suppl 5):S3–18.
6.Azizi M, Menard J. Renin inhibitors and cardiovascular and renal protection: an endless quest? Cardiovasc Drugs Ther. 2013; 27(2):145–53. [PubMed: 22392185]
7.Gheorghiade M, Bohm M, Greene SJ, Fonarow GC, Lewis EF, Zannad F, Solomon SD, Baschiera F, Botha J, Hua TA, Gimpelewicz CR, Jaumont X, Lesogor A, Maggioni AP. A Investigators Coordinators. Effect of aliskiren on postdischarge mortality and heart failure readmissions among patients hospitalized for heart failure: the ASTRONAUT randomized trial. JAMA. 2013; 309(11): 1125–35. [PubMed: 23478743]
8.McMurray JJ, Krum H, Abraham WT, Dickstein K, Kober LV, Desai AS, Solomon SD, Greenlaw N, Ali MA, Chiang Y, Shao Q, Tarnesby G, Massie BM. Investigators AC. Aliskiren, Enalapril, or Aliskiren and Enalapril in Heart Failure. N Engl J Med. 2016; 374(16):1521–32. [PubMed: 27043774]
9.McMurray JJ. Neprilysin inhibition to treat heart failure: a tale of science, serendipity, and second chances. Eur J Heart Fail. 2015; 17(3):242–7. [PubMed: 25756942]
10.Gavras H, Brunner HR, Laragh JH, Gavras I, Vukovich RA. The use of angiotensin-converting enzyme inhibitor in the diagnosis and treatment of hypertension. Clin Sci Mol Med Suppl. 1975; 2:57s–60s. [PubMed: 1077792]
11.Nussberger J, Wuerzner G, Jensen C, Brunner HR. Angiotensin II suppression in humans by the orally active renin inhibitor Aliskiren (SPP100): comparison with enalapril. Hypertension. 2002; 39(1):E1–8. [PubMed: 11799102]
12.Ferrario CM, Schiffrin EL. Role of mineralocorticoid receptor antagonists in cardiovascular disease. Circ Res. 2015; 116(1):206–13. [PubMed: 25552697]
13.Dell』Italia LJ. Translational success stories: angiotensin receptor 1 antagonists in heart failure. Circ Res. 2011; 109(4):437–52. [PubMed: 21817164]
14.Baker WL, Coleman CI, Kluger J, Reinhart KM, Talati R, Quercia R, Phung OJ, White CM. Systematic review: comparative effectiveness of angiotensin-converting enzyme inhibitors or angiotensin II-receptor blockers for ischemic heart disease. Ann Intern Med. 2009; 151(12):861– 71. [PubMed: 20008762]
15.Bangalore S, Fakheri R, Wandel S, Toklu B, Wandel J, Messerli FH. Renin angiotensin system inhibitors for patients with stable coronary artery disease without heart failure: systematic review and meta-analysis of randomized trials. BMJ. 2017; 356:j4. [PubMed: 28104622]
Ferrario and Mullick Page 19
16.Blacher J, Evans A, Arveiler D, Amouyel P, Ferrieres J, Bingham A, Yarnell J, Haas B, Montaye M, Ruidavets JB, Ducimetiere P, Group PS. Residual cardiovascular risk in treated hypertension and hyperlipidaemia: the PRIME Study. J Hum Hypertens. 2010; 24(1):19–26. [PubMed: 19474798]
17.Brugts JJ, van Vark L, Akkerhuis M, Bertrand M, Fox K, Mourad JJ, Boersma E. Impact of renin- angiotensin system inhibitors on mortality and major cardiovascular endpoints in hypertension: A number-needed-to-treat analysis. Int J Cardiol. 2015; 181:425–9. [PubMed: 25569271]
18.Dusing R. Mega clinical trials which have shaped the RAS intervention clinical practice. Ther Adv Cardiovasc Dis. 2016; 10(3):133–50. [PubMed: 27271312]
19.Ferrario CM. Cardiac remodelling and RAS inhibition. Ther Adv Cardiovasc Dis. 2016; 10(3): 162–71. [PubMed: 27105891]
20.Ferrario CM, Ahmad S, Varagic J, Cheng CP, Groban L, Wang H, Collawn JF, Dell Italia LJ. Intracrine angiotensin II functions originate from noncanonical pathways in the human heart. Am J Physiol Heart Circ Physiol. 2016; 311(2):H404–14. [PubMed: 27233763]
21.Reyes S, Varagic J, Ahmad S, VonCannon J, Kon N, Wang H, Groban L, Cheng CP, Ferrario CM. Novel Cardiac Intracrine Mechanisms Based on Ang-(1-12)/Chymase Axis Requires a Revision of Therapeutic Approaches in Human Heart Disease. Curr Hypertens Rep. 2017
22.Drummer OH, Kourtis S, Johnson H. Formation of angiotensin II and other angiotensin peptides from des-leu 10-angiotensin I in rat lung and kidney. Biochem Pharmacol. 1988; 37(22):4327–33. [PubMed: 2848526]
23.Drummer OH, Kourtis S, Johnson H. Effect of chronic enalapril treatment on enzymes responsible for the catabolism of angiotensin I and formation of angiotensin II. Biochem Pharmacol. 1990; 39(3):513–8. [PubMed: 2407244]
24.Ferrario CM, Brosnihan KB, Diz DI, Jaiswal N, Khosla MC, Milsted A, Tallant EA. Angiotensin- (1-7): a new hormone of the angiotensin system. Hypertension. 1991; 18(5 Suppl):III126–33. [PubMed: 1937675]
25.Ferrario CM, Chappell MC, Tallant EA, Brosnihan KB, Diz DI. Counterregulatory actions of angiotensin-(1-7). Hypertension. 1997; 30(3 Pt 2):535–41. [PubMed: 9322978]
26.Jankowski V, Vanholder R, van der Giet M, Tolle M, Karadogan S, Gobom J, Furkert J, Oksche A, Krause E, Tran TN, Tepel M, Schuchardt M, Schluter H, Wiedon A, Beyermann M, Bader M, Todiras M, Zidek W, Jankowski J. Mass-spectrometric identification of a novel angiotensin peptide in human plasma. Arterioscler Thromb Vasc Biol. 2007; 27(2):297–302. [PubMed: 17138938]
27.Lautner RQ, Villela DC, Fraga-Silva RA, Silva N, Verano-Braga T, Costa-Fraga F, Jankowski J, Jankowski V, Sousa F, Alzamora A, Soares E, Barbosa C, Kjeldsen F, Oliveira A, Braga J, Savergnini S, Maia G, Peluso AB, Passos-Silva D, Ferreira A, Alves F, Martins A, Raizada M, Paula R, Motta-Santos D, Klempin F, Pimenta A, Alenina N, Sinisterra R, Bader M, Campagnole- Santos MJ, Santos RA. Discovery and characterization of alamandine: a novel component of the renin-angiotensin system. Circ Res. 2013; 112(8):1104–11. [PubMed: 23446738]
28.Nagata S, Hatakeyama K, Asami M, Tokashiki M, Hibino H, Nishiuchi Y, Kuwasako K, Kato J, Asada Y, Kitamura K. Big angiotensin-25: a novel glycosylated angiotensin-related peptide isolated from human urine. Biochem Biophys Res Commun. 2013; 441(4):757–62. [PubMed: 24211583]
29.Nagata S, Kato J, Sasaki K, Minamino N, Eto T, Kitamura K. Isolation and identification of proangiotensin-12, a possible component of the renin-angiotensin system. Biochem Biophys Res Commun. 2006; 350(4):1026–31. [PubMed: 17045572]
30.Gonzalez AA, Prieto MC. Roles of collecting duct renin and (pro)renin receptor in hypertension: mini review. Ther Adv Cardiovasc Dis. 2015; 9(4):191–200. [PubMed: 25780059]
31.Navar LG, Kobori H, Prieto MC, Gonzalez-Villalobos RA. Intratubular renin-angiotensin system in hypertension. Hypertension. 2011; 57(3):355–62. [PubMed: 21282552]
32.Navar LG, Prieto MC, Satou R, Kobori H. Intrarenal angiotensin II and its contribution to the genesis of chronic hypertension. Curr Opin Pharmacol. 2011; 11(2):180–6. [PubMed: 21339086]
33.Ahmad S, Varagic J, Groban L, Dell』Italia LJ, Nagata S, Kon ND, Ferrario CM. Angiotensin- (1-12): a chymase-mediated cellular angiotensin II substrate. Curr Hypertens Rep. 2014; 16(5): 429. [PubMed: 24633843]
Ferrario and Mullick Page 20
34.Trask AJ, Jessup JA, Chappell MC, Ferrario CM. Angiotensin-(1-12) is an alternate substrate for angiotensin peptide production in the heart. Am J Physiol Heart Circ Physiol. 2008; 294(5):H2242–7. [PubMed: 18359898]
35.Balcells E, Meng QC, Johnson WH Jr, Oparil S, Dell』Italia LJ. Angiotensin II formation from ACE and chymase in human and animal hearts: methods and species considerations. Am J Physiol. 1997; 273(4 Pt 2):H1769–74. [PubMed: 9362242]
36.Dell』Italia LJ, Husain A. Dissecting the role of chymase in angiotensin II formation and heart and blood vessel diseases. Curr Opin Cardiol. 2002; 17(4):374–9. [PubMed: 12151872]
37.Urata H, Healy B, Stewart RW, Bumpus FM, Husain A. Angiotensin II-forming pathways in normal and failing human hearts. Circ Res. 1990; 66(4):883–90. [PubMed: 2156635]
38.Urata H, Kinoshita A, Misono KS, Bumpus FM, Husain A. Identification of a highly specific chymase as the major angiotensin II-forming enzyme in the human heart. J Biol Chem. 1990; 265(36):22348–57. [PubMed: 2266130]
39.Ferrario CM, Ahmad S, Nagata S, Simington SW, Varagic J, Kon N, Dell』italia LJ. An evolving story of angiotensin-II-forming pathways in rodents and humans. Clin Sci (Lond). 2014; 126(7): 461–9. [PubMed: 24329563]
40.Ahmad S, Varagic J, VonCannon JL, Groban L, Collawn JF, Dell』Italia LJ, Ferrario CM. Primacy of cardiac chymase over angiotensin converting enzyme as an angiotensin-(1-12) metabolizing enzyme. Biochem Biophys Res Commun. 2016; 478(2):559–64. [PubMed: 27465904]
41.Jessup JA, Trask AJ, Chappell MC, Nagata S, Kato J, Kitamura K, Ferrario CM. Localization of the novel angiotensin peptide, angiotensin-(1-12), in heart and kidney of hypertensive and normotensive rats. Am J Physiol Heart Circ Physiol. 2008; 294(6):H2614–8. [PubMed: 18408132]
42.Ferrario CM, VonCannon J, Jiao Y, Ahmad S, Bader M, Dell』Italia LJ, Groban L, Varagic J. Cardiac angiotensin-(1-12) expression and systemic hypertension in rats expressing the human angiotensinogen gene. Am J Physiol Heart Circ Physiol. 2016; 310(8):H995–1002. [PubMed: 26873967]
43.Nagata S, Varagic J, Kon ND, Wang H, Groban L, Simington SW, Ahmad S, Dell』Italia LJ, VonCannon JL, Deal D, Ferrario CM. Differential expression of the angiotensin-(1-12)/chymase axis in human atrial tissue. Ther Adv Cardiovasc Dis. 2015; 9(4):168–80. [PubMed: 26082339]
44.Arnold AC, Isa K, Shaltout HA, Nautiyal M, Ferrario CM, Chappell MC, Diz DI. Angiotensin-
(1-12) requires angiotensin converting enzyme and AT1 receptors for cardiovascular actions within the solitary tract nucleus. Am J Physiol Heart Circ Physiol. 2010; 299(3):H763–71. [PubMed: 20562338]
45.Arakawa H, Kawabe K, Sapru HN. Angiotensin-(1-12) in the rostral ventrolateral medullary pressor area of the rat elicits sympathoexcitatory responses. Exp Physiol. 2013; 98(1):94–108. [PubMed: 22707504]
46.De Mello WC, Dell』Itallia LJ, Varagic J, Ferrario CM. Intracellular angiotensin-(1-12) changes the electrical properties of intact cardiac muscle. Mol Cell Biochem. 2016; 422(1–2):31–40. [PubMed: 27590241]
47.Isa K, Garcia-Espinosa MA, Arnold AC, Pirro NT, Tommasi EN, Ganten D, Chappell MC, Ferrario CM, Diz DI. Chronic immunoneutralization of brain angiotensin-(1-12) lowers blood pressure in transgenic (mRen2)27 hypertensive rats. Am J Physiol Regul Integr Comp Physiol. 2009; 297(1):R111–5. [PubMed: 19403863]
48.Arakawa H, Chitravanshi VC, Sapru HN. The hypothalamic arcuate nucleus: a new site of cardiovascular action of angiotensin-(1-12) and angiotensin II. Am J Physiol Heart Circ Physiol. 2011; 300(3):H951–60. [PubMed: 21186269]
49.Ferrario CM, Varagic J, Habibi J, Nagata S, Kato J, Chappell MC, Trask AJ, Kitamura K, Whaley- Connell A, Sowers JR. Differential regulation of angiotensin-(1-12) in plasma and cardiac tissue in response to bilateral nephrectomy. Am J Physiol Heart Circ Physiol. 2009; 296(4):H1184–92. [PubMed: 19218503]
50.Ahmad S, Simmons T, Varagic J, Moniwa N, Chappell MC, Ferrario CM. Chymase-dependent generation of angiotensin II from angiotensin-(1-12) in human atrial tissue. PLoS One. 2011; 6(12):e28501. [PubMed: 22180785]
Ferrario and Mullick Page 21
51.Ahmad S, Wei CC, Tallaj J, Dell』Italia LJ, Moniwa N, Varagic J, Ferrario CM. Chymase mediates angiotensin-(1-12) metabolism in normal human hearts. J Am Soc Hypertens. 2013; 7(2):128–36. [PubMed: 23312967]
52.Husain A. The chymase-angiotensin system in humans. J Hypertens. 1993; 11(11):1155–9. [PubMed: 8301095]
53.Biollaz J, Brunner HR, Gavras I, Waeber B, Gavras H. Antihypertensive therapy with MK 421: angiotensin II--renin relationships to evaluate efficacy of converting enzyme blockade. J Cardiovasc Pharmacol. 1982; 4(6):966–72. [PubMed: 6185790]
54.Roig E, Perez-Villa F, Morales M, Jimenez W, Orus J, Heras M, Sanz G. Clinical implications of increased plasma angiotensin II despite ACE inhibitor therapy in patients with congestive heart failure. Eur Heart J. 2000; 21(1):53–7. [PubMed: 10610744]
55.Donoghue M, Hsieh F, Baronas E, Godbout K, Gosselin M, Stagliano N, Donovan M, Woolf B, Robison K, Jeyaseelan R, Breitbart RE, Acton S. A novel angiotensin-converting enzyme-related carboxypeptidase (ACE2) converts angiotensin I to angiotensin 1–9. Circ Res. 2000; 87(5):E1–9. [PubMed: 10969042]
56.Rice GI, Thomas DA, Grant PJ, Turner AJ, Hooper NM. Evaluation of angiotensin-converting enzyme (ACE), its homologue ACE2 and neprilysin in angiotensin peptide metabolism. Biochem J. 2004; 383(Pt 1):45–51. [PubMed: 15283675]
57.Turner AJ, Tipnis SR, Guy JL, Rice G, Hooper NM. ACEH/ACE2 is a novel mammalian metallocarboxypeptidase and a homologue of angiotensin-converting enzyme insensitive to ACE inhibitors. Can J Physiol Pharmacol. 2002; 80(4):346–53. [PubMed: 12025971]
58.Ocaranza MP, Michea L, Chiong M, Lagos CF, Lavandero S, Jalil JE. Recent insights and therapeutic perspectives of angiotensin-(1-9) in the cardiovascular system. Clin Sci (Lond). 2014; 127(9):549–57. [PubMed: 25029123]
59.Ferrario CM. Angiotensin-converting enzyme 2 and angiotensin-(1-7): an evolving story in cardiovascular regulation. Hypertension. 2006; 47(3):515–21. [PubMed: 16365192]
60.Schiavone MT, Santos RA, Brosnihan KB, Khosla MC, Ferrario CM. Release of vasopressin from the rat hypothalamo-neurohypophysial system by angiotensin-(1-7) heptapeptide. Proc Natl Acad Sci U S A. 1988; 85(11):4095–8. [PubMed: 3375255]
61.Ferrario CM. New physiological concepts of the renin-angiotensin system from the investigation of precursors and products of angiotensin I metabolism. Hypertension. 2010; 55(2):445–52. [PubMed: 20026757]
62.Yamamoto K, Chappell MC, Brosnihan KB, Ferrario CM. In vivo metabolism of angiotensin I by neutral endopeptidase (EC 3.4.24.11) in spontaneously hypertensive rats. Hypertension. 1992; 19(6 Pt 2):692–6. [PubMed: 1317352]
63.Welches WR, Brosnihan KB, Ferrario CM. A comparison of the properties and enzymatic activities of three angiotensin processing enzymes: angiotensin converting enzyme, prolyl endopeptidase and neutral endopeptidase 24.11. Life Sci. 1993; 52(18):1461–80. [PubMed: 8387132]
64.Welches WR, Santos RA, Chappell MC, Brosnihan KB, Greene LJ, Ferrario CM. Evidence that prolyl endopeptidase participates in the processing of brain angiotensin. J Hypertens. 1991; 9(7): 631–8. [PubMed: 1653799]
65.Ferrario CM, Martell N, Yunis C, Flack JM, Chappell MC, Brosnihan KB, Dean RH, Fernandez A, Novikov SV, Pinillas C, Luque M. Characterization of angiotensin-(1-7) in the urine of normal and essential hypertensive subjects. Am J Hypertens. 1998; 11(2):137–46. [PubMed: 9524041]
66.Luque M, Martin P, Martell N, Fernandez C, Brosnihan KB, Ferrario CM. Effects of captopril related to increased levels of prostacyclin and angiotensin-(1-7) in essential hypertension. J Hypertens. 1996; 14(6):799–805. [PubMed: 8793704]
67.de Gasparo M, Catt KJ, Inagami T, Wright JW, Unger T. International union of pharmacology.
XXIII. The angiotensin II receptors. Pharmacol Rev. 2000; 52(3):415–72. [PubMed: 10977869]
68.Kambayashi Y, Bardhan S, Takahashi K, Tsuzuki S, Inui H, Hamakubo T, Inagami T. Molecular cloning of a novel angiotensin II receptor isoform involved in phosphotyrosine phosphatase inhibition. J Biol Chem. 1993; 268(33):24543–6. [PubMed: 8227011]
Ferrario and Mullick Page 22
69.Mukoyama M, Nakajima M, Horiuchi M, Sasamura H, Pratt RE, Dzau VJ. Expression cloning of type 2 angiotensin II receptor reveals a unique class of seven-transmembrane receptors. J Biol Chem. 1993; 268(33):24539–42. [PubMed: 8227010]
70.Nakajima M, Mukoyama M, Pratt RE, Horiuchi M, Dzau VJ. Cloning of cDNA and analysis of the gene for mouse angiotensin II type 2 receptor. Biochem Biophys Res Commun. 1993; 197(2):393– 9. [PubMed: 8267573]
71.Santos RA, Simoes e Silva AC, Maric C, Silva DM, Machado RP, de Buhr I, Heringer-Walther S, Pinheiro SV, Lopes MT, Bader M, Mendes EP, Lemos VS, Campagnole-Santos MJ, Schultheiss HP, Speth R, Walther T. Angiotensin-(1-7) is an endogenous ligand for the G protein-coupled receptor Mas. Proc Natl Acad Sci U S A. 2003; 100(14):8258–63. [PubMed: 12829792]
72.Tetzner A, Gebolys K, Meinert C, Klein S, Uhlich A, Trebicka J, Villacanas O, Walther T. G- Protein-Coupled Receptor MrgD Is a Receptor for Angiotensin-(1-7) Involving Adenylyl Cyclase, cAMP, and Phosphokinase A. Hypertension. 2016; 68(1):185–94. [PubMed: 27217404]
73.Li P, Ferrario CM, Brosnihan KB. Nonpeptide angiotensin II antagonist losartan inhibits thromboxane A2-induced contractions in canine coronary arteries. J Pharmacol Exp Ther. 1997; 281(3):1065–70. [PubMed: 9190837]
74.Li P, Ferrario CM, Brosnihan KB. Losartan inhibits thromboxane A2-induced platelet aggregation and vascular constriction in spontaneously hypertensive rats. J Cardiovasc Pharmacol. 1998; 32(2): 198–205. [PubMed: 9700980]
75.Fukuhara M, Neves LA, Li P, Diz DI, Ferrario CM, Brosnihan KB. The angiotensin II AT1 receptor antagonist irbesartan prevents thromboxane A2-induced vasoconstriction in the rat hind- limb vascular bed in vivo. J Hypertens. 2001; 19(3 Pt 2):561–6. [PubMed: 11327630]
76.Li P, Fukuhara M, Diz DI, Ferrario CM, Brosnihan KB. Novel angiotensin II AT(1) receptor antagonist irbesartan prevents thromboxane A(2)-induced vasoconstriction in canine coronary arteries and human platelet aggregation. J Pharmacol Exp Ther. 2000; 292(1):238–46. [PubMed: 10604953]
77.Goyal SN, Bharti S, Bhatia J, Nag TC, Ray R, Arya DS. Telmisartan, a dual ARB/partial PPAR- gamma agonist, protects myocardium from ischaemic reperfusion injury in experimental diabetes. Diabetes Obes Metab. 2011; 13(6):533–41. [PubMed: 21320264]
78.Chida R, Hisauchi I, Toyoda S, Kikuchi M, Komatsu T, Hori Y, Nakahara S, Sakai Y, Inoue T, Taguchi I. Impact of irbesartan, an angiotensin receptor blocker, on uric acid level and oxidative stress in high-risk hypertension patients. Hypertens Res. 2015; 38(11):765–9. [PubMed: 26178150]
79.Ferrario C, Abdelhamed AI, Moore M. AII antagonists in hypertension, heart failure, and diabetic nephropathy: focus on losartan. Curr Med Res Opin. 2004; 20(3):279–93. [PubMed: 15025837]
80.Wolff ML, Cruz JL, Vanderman AJ, Brown JN. The effect of angiotensin II receptor blockers on hyperuricemia. Ther Adv Chronic Dis. 2015; 6(6):339–46. [PubMed: 26568810]
81.Guo DF, Sun YL, Hamet P, Inagami T. The angiotensin II type 1 receptor and receptor-associated proteins. Cell Res. 2001; 11(3):165–80. [PubMed: 11642401]
82.Carey RM, Wang ZQ, Siragy HM. Role of the angiotensin type 2 receptor in the regulation of blood pressure and renal function. Hypertension. 2000; 35(1 Pt 2):155–63. [PubMed: 10642292]
83.Gallagher PE, Ferrario CM, Tallant EA. MAP kinase/phosphatase pathway mediates the regulation of ACE2 by angiotensin peptides. Am J Physiol Cell Physiol. 2008; 295(5):C1169–74. [PubMed: 18768926]
84.Tallant EA, Ferrario CM, Gallagher PE. Angiotensin-(1-7) inhibits growth of cardiac myocytes through activation of the mas receptor. Am J Physiol Heart Circ Physiol. 2005; 289(4):H1560–6. [PubMed: 15951342]
85.Zhang X, Cheng HJ, Zhou P, Kitzman DW, Ferrario CM, Li WM, Cheng CP. Cellular basis of angiotensin-(1-7)-induced augmentation of left ventricular functional performance in heart failure. Int J Cardiol. 2017
86.Calhoun D. Sy 14-3 Primary Aldosteronism in Resistant Hypertension. J Hypertens. 2016; 34(Suppl 1- ISH 2016 Abstract Book):e369.
87.Calhoun DA. Use of aldosterone antagonists in resistant hypertension. Prog Cardiovasc Dis. 2006; 48(6):387–96. [PubMed: 16714158]
Ferrario and Mullick Page 23
88.Clark D 3rd, Guichard JL, Calhoun DA, Ahmed MI. Recent advancements in the treatment of resistant hypertension. Postgrad Med. 2012; 124(1):67–73. [PubMed: 22314116]
89.Passos-Silva DG, Brandan E, Santos RA. Angiotensins as therapeutic targets beyond heart disease. Trends Pharmacol Sci. 2015; 36(5):310–20. [PubMed: 25847571]
90.Azizi M, Chatellier G, Guyene TT, Murieta-Geoffroy D, Menard J. Additive effects of combined angiotensin-converting enzyme inhibition and angiotensin II antagonism on blood pressure and renin release in sodium-depleted normotensives. Circulation. 1995; 92(4):825–34. [PubMed: 7641363]
91.Azizi M, Menard J, Bissery A, Guyenne TT, Bura-Riviere A, Vaidyanathan S, Camisasca RP. Pharmacologic demonstration of the synergistic effects of a combination of the renin inhibitor aliskiren and the AT1 receptor antagonist valsartan on the angiotensin II-renin feedback interruption. Journal of the American Society of Nephrology: JASN. 2004; 15(12):3126–33. [PubMed: 15579516]
92.Ferrario CM. Addressing the theoretical and clinical advantages of combination therapy with inhibitors of the renin-angiotensin-aldosterone system: antihypertensive effects and benefits beyond BP control. Life Sci. 2010; 86(9–10):289–99. [PubMed: 19958778]
93.Kim S, Yoshiyama M, Izumi Y, Kawano H, Kimoto M, Zhan Y, Iwao H. Effects of combination of ACE inhibitor and angiotensin receptor blocker on cardiac remodeling, cardiac function, and survival in rat heart failure. Circulation. 2001; 103(1):148–54. [PubMed: 11136700]
94.Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P, Anderson C. Investigators O. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008; 358(15):1547–59. [PubMed: 18378520]
95.Mancia G, Schumacher H. Incidence of adverse events with telmisartan compared with ACE inhibitors: evidence from a pooled analysis of clinical trials. Patient Prefer Adherence. 2012; 6:1– 9. [PubMed: 22272064]
96.Yusuf S, Teo KK, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P, Anderson C. for the Ontarget Investigators. Telmisartan, ramipril, or both in patients at high risk for vascular events. N Engl J Med. 2008; 358(15):1547–59. [PubMed: 18378520]
97.Doran DE, Weiss D, Zhang Y, Griendling KK, Taylor WR. Differential effects of AT1 receptor and Ca2+ channel blockade on atherosclerosis, inflammatory gene expression, and production of reactive oxygen species. Atherosclerosis. 2007; 195(1):39–47. [PubMed: 17224157]
98.Stumpe KO, Agabiti-Rosei E, Zielinski T, Schremmer D, Scholze J, Laeis P, Schwandt P, Ludwig
M. Ms investigators. Carotid intima-media thickness and plaque volume changes following 2-year angiotensin II-receptor blockade. The Multicentre Olmesartan atherosclerosis Regression Evaluation (MORE) study. Ther Adv Cardiovasc Dis. 2007; 1(2):97–106. [PubMed: 19124398]
99.Ishii H, Kobayashi M, Kurebayashi N, Yoshikawa D, Suzuki S, Ichimiya S, Kanashiro M, Sone T, Tsuboi H, Amano T, Uetani T, Harada K, Marui N, Murohara T. Impact of angiotensin II receptor blocker therapy (olmesartan or valsartan) on coronary atherosclerotic plaque volume measured by intravascular ultrasound in patients with stable angina pectoris. Am J Cardiol. 2013; 112(3):363–8. [PubMed: 23623047]
100.Ferrario CM, Varagic J. The ANG-(1-7)/ACE2/mas axis in the regulation of nephron function. Am J Physiol Renal Physiol. 2010; 298(6):F1297–305. [PubMed: 20375118]
101.Turnbull F, Neal B, Ninomiya T, Algert C, Arima H, Barzi F, Bulpitt C, Chalmers J, Fagard R, Gleason A, Heritier S, Li N, Perkovic V, Woodward M, MacMahon S. Blood Pressure Lowering Treatment Trialists C. Effects of different regimens to lower blood pressure on major cardiovascular events in older and younger adults: meta-analysis of randomised trials. BMJ. 2008; 336(7653):1121–3. [PubMed: 18480116]
102.Aoyagi T, Kunimoto S, Morishima H, Takeuchi T, Umezawa H. Effect of pepstatin on acid proteases. The Journal of antibiotics. 1971; 24(10):687–94. [PubMed: 4945810]
103.Staessen JA, Li Y, Richart T. Oral renin inhibitors. Lancet. 2006; 368(9545):1449–56. [PubMed: 17055947]
104.Webb DJ, Manhem PJ, Ball SG, Inglis G, Leckie BJ, Lever AF, Morton JJ, Robertson JI, Murray GD, Menard J, et al. A study of the renin inhibitor H142 in man. Journal of hypertension. 1985; 3(6):653–8. [PubMed: 3910726]
Ferrario and Mullick Page 24
105.Pool JL. Direct renin inhibition: focus on aliskiren. Journal of managed care pharmacy: JMCP. 2007; 13(8 Suppl B):21–33.
106.Wood JM, Maibaum J, Rahuel J, Grutter MG, Cohen NC, Rasetti V, Ruger H, Goschke R, Stutz S, Fuhrer W, Schilling W, Rigollier P, Yamaguchi Y, Cumin F, Baum HP, Schnell CR, Herold P, Mah R, Jensen C, O』Brien E, Stanton A, Bedigian MP. Structure-based design of aliskiren, a novel orally effective renin inhibitor. Biochemical and biophysical research communications. 2003; 308(4):698–705. [PubMed: 12927775]
107.Brown MJ. Aliskiren. Circulation. 2008; 118(7):773–84. [PubMed: 18695203]
108.Nobakht N, Kamgar M, Rastogi A, Schrier RW. Limitations of angiotensin inhibition. Nature reviews. Nephrology. 2011; 7(6):356–9. [PubMed: 21502972]
109.Bomback AS, Klemmer PJ. The incidence and implications of aldosterone breakthrough. Nature clinical practice. Nephrology. 2007; 3(9):486–92.
110.Cicoira M, Zanolla L, Rossi A, Golia G, Franceschini L, Cabrini G, Bonizzato A, Graziani M, Anker SD, Coats AJ, Zardini P. Failure of aldosterone suppression despite angiotensin-converting enzyme (ACE) inhibitor administration in chronic heart failure is associated with ACE DD genotype. J Am Coll Cardiol. 2001; 37(7):1808–12. [PubMed: 11401115]
111.Moranne O, Bakris G, Fafin C, Favre G, Pradier C, Esnault VL. Determinants and changes associated with aldosterone breakthrough after angiotensin II receptor blockade in patients with type 2 diabetes with overt nephropathy. Clin J Am Soc Nephrol. 2013; 8(10):1694–701. [PubMed: 23929924]
112.Mazzocchi G, Gottardo G, Macchi V, Malendowicz LK, Nussdorfer GG. The AT2 receptor- mediated stimulation of adrenal catecholamine release may potentiate the AT1 receptor-mediated aldosterone secretagogue action of angiotensin-II in rats. Endocr Res. 1998; 24(1):17–28. [PubMed: 9553752]
113.Naruse M, Tanabe A, Sato A, Takagi S, Tsuchiya K, Imaki T, Takano K. Aldosterone breakthrough during angiotensin II receptor antagonist therapy in stroke-prone spontaneously hypertensive rats. Hypertension. 2002; 40(1):28–33. [PubMed: 12105134]
114.Yatabe J, Yoneda M, Yatabe MS, Watanabe T, Felder RA, Jose PA, Sanada H. Angiotensin III stimulates aldosterone secretion from adrenal gland partially via angiotensin II type 2 receptor but not angiotensin II type 1 receptor. Endocrinology. 2011; 152(4):1582–8. [PubMed: 21303953]
115.Kehoe B, Keeton GR, Hill C. Elevated plasma renin activity associated with renal dysfunction. Nephron. 1986; 44(1):51–7.
116.Malmqvist K, Ohman KP, Lind L, Nystrom F, Kahan T. Relationships between left ventricular mass and the renin-angiotensin system, catecholamines, insulin and leptin. Journal of internal medicine. 2002; 252(5):430–9. [PubMed: 12528761]
117.Alderman MH, Ooi WL, Cohen H, Madhavan S, Sealey JE, Laragh JH. Plasma renin activity: a risk factor for myocardial infarction in hypertensive patients. American journal of hypertension. 1997; 10(1):1–8. [PubMed: 9008242]
118.O』Brien E, Barton J, Nussberger J, Mulcahy D, Jensen C, Dicker P, Stanton A. Aliskiren reduces blood pressure and suppresses plasma renin activity in combination with a thiazide diuretic, an angiotensin-converting enzyme inhibitor, or an angiotensin receptor blocker. Hypertension. 2007; 49(2):276–84. [PubMed: 17159081]
119.Oparil S, Schmieder RE. New approaches in the treatment of hypertension. Circulation research. 2015; 116(6):1074–95. [PubMed: 25767291]
120.Menard J, Campbell DJ, Azizi M, Gonzales MF. Synergistic effects of ACE inhibition and Ang II antagonism on blood pressure, cardiac weight, and renin in spontaneously hypertensive rats. Circulation. 1997; 96(9):3072–8. [PubMed: 9386177]
121.Stanton A. Therapeutic potential of renin inhibitors in the management of cardiovascular disorders. American journal of cardiovascular drugs: drugs, devices, and other interventions. 2003; 3(6):389–94.
122.Urata H, Healy B, Stewart RW, Bumpus FM, Husain A. Angiotensin II-forming pathways in normal and failing human hearts. Circulation research. 1990; 66(4):883–90. [PubMed: 2156635]
Ferrario and Mullick Page 25
123.Dzau VJ, Bernstein K, Celermajer D, Cohen J, Dahlof B, Deanfield J, Diez J, Drexler H, Ferrari R, Van Gilst W, Hansson L, Hornig B, Husain A, Johnston C, Lazar H, Lonn E, Luscher T, Mancini J, Mimran A, Pepine C, Rabelink T, Remme W, Ruilope L, Ruzicka M, Schunkert H, Swedberg K, Unger T, Vaughan D, Weber M. Pathophysiologic and therapeutic importance of tissue ACE: a consensus report. Cardiovascular drugs and therapy. 2002; 16(2):149–60. [PubMed: 12090908]
124.McMurray JJ, Krum H, Abraham WT, Dickstein K, Kober LV, Desai AS, Solomon SD, Greenlaw N, Ali MA, Chiang Y, Shao Q, Tarnesby G, Massie BM. Aliskiren, Enalapril, or Aliskiren and Enalapril in Heart Failure. The New England journal of medicine. 2016; 374(16):1521–32. [PubMed: 27043774]
125.Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner SM, Solomon SD, Chaturvedi N, Persson F, Desai AS, Nicolaides M, Richard A, Xiang Z, Brunel P, Pfeffer MA. Cardiorenal end points in a trial of aliskiren for type 2 diabetes. The New England journal of medicine. 2012; 367(23):2204–13. [PubMed: 23121378]
126.Gheorghiade M, Bohm M, Greene SJ, Fonarow GC, Lewis EF, Zannad F, Solomon SD, Baschiera F, Botha J, Hua TA, Gimpelewicz CR, Jaumont X, Lesogor A, Maggioni AP. Effect of aliskiren on postdischarge mortality and heart failure readmissions among patients hospitalized for heart failure: the ASTRONAUT randomized trial. JAMA. 2013; 309(11):1125–35. [PubMed: 23478743]
127.Moniwa N, Varagic J, Ahmad S, VonCannon JL, Simington SW, Wang H, Groban L, Brosnihan KB, Nagata S, Kato J, Kitamura K, Gomez RA, Lopez ML, Ferrario CM. Hemodynamic and hormonal changes to dual renin-angiotensin system inhibition in experimental hypertension. Hypertension. 2013; 61(2):417–24. [PubMed: 23232645]
128.Rajagopalan S, Bakris GL, Abraham WT, Pitt B, Brook RD. Complete renin-angiotensin- aldosterone system (RAAS) blockade in high-risk patients: recent insights from renin blockade studies. Hypertension. 2013; 62(3):444–9. [PubMed: 23876474]
129.Feldman DL, Jin L, Xuan H, Contrepas A, Zhou Y, Webb RL, Mueller DN, Feldt S, Cumin F, Maniara W, Persohn E, Schuetz H, Jan Danser AH, Nguyen G. Effects of aliskiren on blood pressure, albuminuria, and (pro)renin receptor expression in diabetic TG(mRen-2)27 rats. Hypertension. 2008; 52(1):130–6. [PubMed: 18490518]
130.Lange S, Fraune C, Alenina N, Bader M, Danser AH, Frenay AR, van Goor H, Stahl R, Nguyen G, Schwedhelm E, Wenzel UO. Aliskiren accumulation in the kidney: no major role for binding to renin or prorenin. Journal of hypertension. 2013; 31(4):713–9. [PubMed: 23407438]
131.Te Riet L, van Esch JH, Roks AJ, van den Meiracker AH, Danser AH. Hypertension: renin- angiotensin-aldosterone system alterations. Circulation research. 2015; 116(6):960–75. [PubMed: 25767283]
132.Balcarek J, Seva Pessoa B, Bryson C, Azizi M, Menard J, Garrelds IM, McGeehan G, Reeves RA, Griffith SG, Danser AH, Gregg R. Multiple ascending dose study with the new renin inhibitor VTP-27999: nephrocentric consequences of too much renin inhibition. Hypertension. 2014; 63(5):942–50. [PubMed: 24470465]
133.Ramkumar N, Kohan DE. Proximal tubule angiotensinogen modulation of arterial pressure. Current opinion in nephrology and hypertension. 2013; 22(1):32–6. [PubMed: 23010762]
134.Schnermann J, Levine DZ. Paracrine factors in tubuloglomerular feedback: adenosine, ATP, and nitric oxide. Annual review of physiology. 2003; 65:501–29.
135.Burke M, Pabbidi MR, Farley J, Roman RJ. Molecular mechanisms of renal blood flow autoregulation. Current vascular pharmacology. 2014; 12(6):845–58. [PubMed: 24066938]
136.Rohrwasser A, Morgan T, Dillon HF, Zhao L, Callaway CW, Hillas E, Zhang S, Cheng T, Inagami T, Ward K, Terreros DA, Lalouel JM. Elements of a paracrine tubular renin-angiotensin system along the entire nephron. Hypertension. 1999; 34(6):1265–74. [PubMed: 10601129]
137.Navis G, Faber HJ, de Zeeuw D, de Jong PE. ACE inhibitors and the kidney. A risk-benefit assessment. Drug safety. 1996; 15(3):200–11. [PubMed: 8879974]
138.Griffiths CD, Morgan TO, Delbridge LM. Effects of combined administration of ACE inhibitor and angiotensin II receptor antagonist are prevented by a high NaCl intake. Journal of hypertension. 2001; 19(11):2087–95. [PubMed: 11677376]
Ferrario and Mullick Page 26
139.Richer-Giudicelli C, Domergue V, Gonzalez MF, Messadi E, Azizi M, Giudicelli JF, Menard J. Haemodynamic effects of dual blockade of the renin-angiotensin system in spontaneously hypertensive rats: influence of salt. Journal of hypertension. 2004; 22(3):619–27. [PubMed: 15076169]
140.Sealey JE, Alderman MH, Furberg CD, Laragh JH. Renin-angiotensin system blockers may create more risk than reward for sodium-depleted cardiovascular patients with high plasma renin levels. American journal of hypertension. 2013; 26(6):727–38. [PubMed: 23548411]
141.Turgut F, Balogun RA, Abdel-Rahman EM. Renin-angiotensin-aldosterone system blockade effects on the kidney in the elderly: benefits and limitations. Clinical journal of the American Society of Nephrology: CJASN. 2010; 5(7):1330–9. [PubMed: 20498247]
142.Schoolwerth AC, Sica DA, Ballermann BJ, Wilcox CS. Renal considerations in angiotensin converting enzyme inhibitor therapy: a statement for healthcare professionals from the Council on the Kidney in Cardiovascular Disease and the Council for High Blood Pressure Research of the American Heart Association. Circulation. 2001; 104(16):1985–91. [PubMed: 11602506]
143.Bicket DP. Using ACE inhibitors appropriately. Am Fam Physician. 2002; 66(3):461–8. [PubMed: 12182524]
144.Gohlke, P., Scholkens, BA. ACE Inhibitors: Pharmacology. In: Unger, T., Scholkens, BA., editors. Angiotensin. Springer - Verlag; Berlin: 2004. p. 375-413.
145.Davis R, Coukell A, McTavish D. Fosinopril. A review of its pharmacology and clinical efficacy in the management of heart failure. Drugs. 1997; 54(1):103–16. [PubMed: 9211084]
146.White CM. Pharmacologic, pharmacokinetic, and therapeutic differences among ACE inhibitors. Pharmacotherapy. 1998; 18(3):588–99. [PubMed: 9620109]
147.Wong MC, Chan DK, Wang HH, Tam WW, Cheung CS, Yan BP, Coats AJ. The incidence of all- cause, cardiovascular and respiratory disease admission among 20,252 users of lisinopril vs. perindopril: A cohort study. Int J Cardiol. 2016; 219:410–6. [PubMed: 27362832]
148.Juillerat L, Nussberger J, Menard J, Mooser V, Christen Y, Waeber B, Graf P, Brunner HR. Determinants of angiotensin II generation during converting enzyme inhibition. Hypertension. 1990; 16(5):564–72. [PubMed: 2172161]
149.Wei CC, Tian B, Perry G, Meng QC, Chen YF, Oparil S, Dell』Italia LJ. Differential ANG II generation in plasma and tissue of mice with decreased expression of the ACE gene. Am J Physiol Heart Circ Physiol. 2002; 282(6):H2254–8. [PubMed: 12003835]
150.Ahmad S, Varagic J, Westwood BM, Chappell MC, Ferrario CM. Uptake and metabolism of the novel peptide angiotensin-(1-12) by neonatal cardiac myocytes. PLoS One. 2011; 6(1):e15759. [PubMed: 21249217]
151.Husain A, Li M, Graham RM. Do studies with ACE N- and C-domain-selective inhibitors provide evidence for a non-ACE, non-chymase angiotensin II-forming pathway? Circ Res. 2003; 93(2): 91–3. [PubMed: 12881473]
152.Dive V, Cotton J, Yiotakis A, Michaud A, Vassiliou S, Jiracek J, Vazeux G, Chauvet MT, Cuniasse P, Corvol P. RXP 407, a phosphinic peptide, is a potent inhibitor of angiotensin I converting enzyme able to differentiate between its two active sites. Proc Natl Acad Sci U S A. 1999; 96(8):4330–5. [PubMed: 10200262]
153.van Vark LC, Bertrand M, Akkerhuis KM, Brugts JJ, Fox K, Mourad JJ, Boersma E. Angiotensin- converting enzyme inhibitors reduce mortality in hypertension: a meta-analysis of randomized clinical trials of renin-angiotensin-aldosterone system inhibitors involving 158,998 patients. Eur Heart J. 2012; 33(16):2088–97. [PubMed: 22511654]
154.Zanchetti A, Thomopoulos C, Parati G. Randomized controlled trials of blood pressure lowering in hypertension: a critical reappraisal. Circ Res. 2015; 116(6):1058–73. [PubMed: 25767290]
155.Miura S, Fujino M, Hanzawa H, Kiya Y, Imaizumi S, Matsuo Y, Tomita S, Uehara Y, Karnik SS, Yanagisawa H, Koike H, Komuro I, Saku K. Molecular mechanism underlying inverse agonist of angiotensin II type 1 receptor. J Biol Chem. 2006; 281(28):19288–95. [PubMed: 16690611]
156.Brenner BM, Cooper ME, de Zeeuw D, Keane WF, Mitch WE, Parving HH, Remuzzi G, Snapinn SM, Zhang Z, Shahinfar S. Investigators RS. Effects of losartan on renal and cardiovascular outcomes in patients with type 2 diabetes and nephropathy. N Engl J Med. 2001; 345(12):861–9. [PubMed: 11565518]
Ferrario and Mullick Page 27
157.Dahlof B, Devereux RB, Kjeldsen SE, Julius S, Beevers G, de Faire U, Fyhrquist F, Ibsen H, Kristiansson K, Lederballe-Pedersen O, Lindholm LH, Nieminen MS, Omvik P, Oparil S, Wedel
H. for the LIFE Study Group. Cardiovascular morbidity and mortality in the Losartan Intervention For Endpoint reduction in hypertension study (LIFE): a randomised trial against atenolol. Lancet. 2002; 359(9311):995–1003. [PubMed: 11937178]
158.Konstam MA, Neaton JD, Dickstein K, Drexler H, Komajda M, Martinez FA, Riegger GA, Malbecq W, Smith RD, Guptha S, Poole-Wilson PA. for the Heaal Investigators. Effects of high- dose versus low-dose losartan on clinical outcomes in patients with heart failure (HEAAL study): a randomised, double-blind trial. Lancet. 2009; 374(9704):1840–8. [PubMed: 19922995]
159.Cohn JN, Tognoni G. for the Valsartan Heart Failure Trial Investigators. A randomized trial of the angiotensin-receptor blocker valsartan in chronic heart failure. N Engl J Med. 2001; 345(23): 1667–75. [PubMed: 11759645]
160.Group TNS, McMurray JJ, Holman RR, Haffner SM, Bethel MA, Holzhauer B, Hua TA, Belenkov Y, Boolell M, Buse JB, Buckley BM, Chacra AR, Chiang FT, Charbonnel B, Chow CC, Davies MJ, Deedwania P, Diem P, Einhorn D, Fonseca V, Fulcher GR, Gaciong Z, Gaztambide S, Giles T, Horton E, Ilkova H, Jenssen T, Kahn SE, Krum H, Laakso M, Leiter LA, Levitt NS, Mareev V, Martinez F, Masson C, Mazzone T, Meaney E, Nesto R, Pan C, Prager R, Raptis SA, Rutten GE, Sandstroem H, Schaper F, Scheen A, Schmitz O, Sinay I, Soska V, Stender S, Tamas G, Tognoni G, Tuomilehto J, Villamil AS, Vozar J, Califf RM. Effect of valsartan on the incidence of diabetes and cardiovascular events. N Engl J Med. 2010; 362(16): 1477–90. [PubMed: 20228403]
161.Julius S, Kjeldsen SE, Brunner H, Hansson L, Platt F, Ekman S, Laragh JH, McInnes G, Schork AM, Smith B, Weber M, Zanchetti A. VALUE trial: Long-term blood pressure trends in 13,449 patients with hypertension and high cardiovascular risk. Am J Hypertens. 2003; 16(7):544–8. [PubMed: 12850387]
162.Julius S, Nesbitt SD, Egan BM, Weber MA, Michelson EL, Kaciroti N, Black HR, Grimm RH Jr, Messerli FH, Oparil S, Schork MA. for the Trial of Preventing Hypertension Study Investigators. Feasibility of treating prehypertension with an angiotensin-receptor blocker. N Engl J Med. 2006; 354(16):1685–97. [PubMed: 16537662]
163.Pfeffer MA, McMurray JJ, Velazquez EJ, Rouleau JL, Kober L, Maggioni AP, Solomon SD, Swedberg K, Van de Werf F, White H, Leimberger JD, Henis M, Edwards S, Zelenkofske S, Sellers MA, Califf RM. for the Valsartan in Acute Myocardial Infarction Trial Investigators. Valsartan, captopril, or both in myocardial infarction complicated by heart failure, left ventricular dysfunction, or both. N Engl J Med. 2003; 349(20):1893–906. [PubMed: 14610160]
164.Granger CB, McMurray JJ, Yusuf S, Held P, Michelson EL, Olofsson B, Ostergren J, Pfeffer MA, Swedberg K. for the CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function intolerant to angiotensin- converting-enzyme inhibitors: the CHARM-Alternative trial. Lancet. 2003; 362(9386):772–6. [PubMed: 13678870]
165.McMurray JJ, Ostergren J, Swedberg K, Granger CB, Held P, Michelson EL, Olofsson B, Yusuf S, Pfeffer MA. for the CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin- converting-enzyme inhibitors: the CHARM-Added trial. Lancet. 2003; 362(9386):767–71. [PubMed: 13678869]
166.Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJ, Michelson EL, Olofsson B, Ostergren J, Yusuf S, Pocock S. C Investigators Committees. Effects of candesartan on mortality and morbidity in patients with chronic heart failure: the CHARM-Overall programme. Lancet. 2003; 362(9386):759–66. [PubMed: 13678868]
167.Yusuf S, Pfeffer MA, Swedberg K, Granger CB, Held P, McMurray JJ, Michelson EL, Olofsson B, Ostergren J. C Investigators Committees. Effects of candesartan in patients with chronic heart failure and preserved left-ventricular ejection fraction: the CHARM-Preserved Trial. Lancet. 2003; 362(9386):777–81. [PubMed: 13678871]
168.Lewis EJ, Hunsicker LG, Clarke WR, Berl T, Pohl MA, Lewis JB, Ritz E, Atkins RC, Rohde R, Raz I. Collaborative Study G. Renoprotective effect of the angiotensin-receptor antagonist
Ferrario and Mullick Page 28
irbesartan in patients with nephropathy due to type 2 diabetes. N Engl J Med. 2001; 345(12): 851–60. [PubMed: 11565517]
169.Massie BM, Carson PE, McMurray JJ, Komajda M, McKelvie R, Zile MR, Anderson S, Donovan M, Iverson E, Staiger C, Ptaszynska A. for the for the I-PRESERVE Investigators. Irbesartan in patients with heart failure and preserved ejection fraction. N Engl J Med. 2008; 359(23):2456– 67. [PubMed: 19001508]
170.Ritz E, Viberti GC, Ruilope LM, Rabelink AJ, Izzo JL Jr, Katayama S, Ito S, Mimran A, Menne J, Rump LC, Januszewicz A, Haller H. Determinants of urinary albumin excretion within the normal range in patients with type 2 diabetes: the Randomised Olmesartan and Diabetes Microalbuminuria Prevention (ROADMAP) study. Diabetologia. 2010; 53(1):49–57. [PubMed: 19876613]
171.Dusing R. Pharmacological interventions into the renin-angiotensin system with ACE inhibitors and angiotensin II receptor antagonists: effects beyond blood pressure lowering. Ther Adv Cardiovasc Dis. 2016; 10(3):151–61. [PubMed: 27122491]
172.Ruschitzka F, Taddei S. Angiotensin-converting enzyme inhibitors: first-line agents in cardiovascular protection? Eur Heart J. 2012; 33(16):1996–8. [PubMed: 22659198]
173.Kumar R, Thomas CM, Yong QC, Chen W, Baker KM. The intracrine renin-angiotensin system. Clin Sci (Lond). 2012; 123(5):273–84. [PubMed: 22590974]
174.Re RN. Cardiac angiotensin II: an intracrine hormone? Am J Hypertens. 2003; 16(5 Pt 1):426–7.
175.Re RN, Cook JL. Noncanonical intracrine action. J Am Soc Hypertens. 2011; 5(6):435–48. [PubMed: 21890449]
176.Grundy HM, Simpson SA, Tait JF. Isolation of a highly active mineralocorticoid from beef adrenal extract. Nature. 1952; 169(4306):795–6. [PubMed: 14941045]
177.Tait JF, Simpson SA, Grundy HM. The effect of adrenal extract on mineral metabolism. Lancet. 1952; 1(6699):122–4. [PubMed: 14889780]
178.Cranston WI, Juel-Jensen BE. The effects of spironolactone and chlorthalidone on arterial pressure. Lancet. 1962; 1(7240):1161–4. [PubMed: 13882033]
179.Conn JW. Primary aldosteronism. J Lab Clin Med. 1955; 45(4):661–4. [PubMed: 14368032]
180.Judd E, Calhoun DA. Management of Resistant Hypertension: Do Not Give Up on Medication. Nephrol Self Assess Program. 2014; 13(2):57–63. [PubMed: 25642153]
181.Gomez-Sanchez CE. Non renal effects of aldosterone. Steroids. 2014; 91:1–2. [PubMed: 25440413]
182.Pitt B, Kober L, Ponikowski P, Gheorghiade M, Filippatos G, Krum H, Nowack C, Kolkhof P, Kim SY, Zannad F. Safety and tolerability of the novel non-steroidal mineralocorticoid receptor antagonist BAY 94-8862 in patients with chronic heart failure and mild or moderate chronic kidney disease: a randomized, double-blind trial. Eur Heart J. 2013; 34(31):2453–63. [PubMed: 23713082]
183.Liu LC, Schutte E, Gansevoort RT, van der Meer P, Voors AA. Finerenone: third-generation mineralocorticoid receptor antagonist for the treatment of heart failure and diabetic kidney disease. Expert Opin Investig Drugs. 2015; 24(8):1123–35.
184.Filippatos G, Anker SD, Bohm M, Gheorghiade M, Kober L, Krum H, Maggioni AP, Ponikowski P, Voors AA, Zannad F, Kim SY, Nowack C, Palombo G, Kolkhof P, Kimmeskamp-Kirschbaum N, Pieper A, Pitt B. A randomized controlled study of finerenone vs. eplerenone in patients with worsening chronic heart failure and diabetes mellitus and/or chronic kidney disease. Eur Heart J. 2016; 37(27):2105–14. [PubMed: 27130705]
185.Barfacker L, Kuhl A, Hillisch A, Grosser R, Figueroa-Perez S, Heckroth H, Nitsche A, Erguden JK, Gielen-Haertwig H, Schlemmer KH, Mittendorf J, Paulsen H, Platzek J, Kolkhof P. Discovery of BAY 94-8862: a nonsteroidal antagonist of the mineralocorticoid receptor for the treatment of cardiorenal diseases. ChemMedChem. 2012; 7(8):1385–403. [PubMed: 22791416]
186.Fleseriu M, Castinetti F. Updates on the role of adrenal steroidogenesis inhibitors in Cushing’s syndrome: a focus on novel therapies. Pituitary. 2016; 19(6):643–653. [PubMed: 27600150]
187.Fleseriu M, Pivonello R, Young J, Hamrahian AH, Molitch ME, Shimizu C, Tanaka T, Shimatsu A, White T, Hilliard A, Tian C, Sauter N, Biller BM, Bertagna X. Osilodrostat, a potent oral
Ferrario and Mullick Page 29
11beta-hydroxylase inhibitor: 22-week, prospective, Phase II study in Cushing’s disease. Pituitary. 2016; 19(2):138–48. [PubMed: 26542280]
188.Lea WB, Kwak ES, Luther JM, Fowler SM, Wang Z, Ma J, Fogo AB, Brown NJ. Aldosterone antagonism or synthase inhibition reduces end-organ damage induced by treatment with angiotensin and high salt. Kidney Int. 2009; 75(9):936–44. [PubMed: 19225557]
189.Menard J, Gonzalez MF, Guyene TT, Bissery A. Investigation of aldosterone-synthase inhibition in rats. J Hypertens. 2006; 24(6):1147–55. [PubMed: 16685215]
190.Gu J, Noe A, Chandra P, Al-Fayoumi S, Ligueros-Saylan M, Sarangapani R, Maahs S, Ksander G, Rigel DF, Jeng AY, Lin TH, Zheng W, Dole WP. Pharmacokinetics and pharmacodynamics of LCZ696, a novel dual-acting angiotensin receptor-neprilysin inhibitor (ARNi). J Clin Pharmacol. 2010; 50(4):401–14. [PubMed: 19934029]
191.Chappell MC. Biochemical evaluation of the renin-angiotensin system: the good, bad, and absolute? Am J Physiol Heart Circ Physiol. 2016; 310(2):H137–52. [PubMed: 26475588]
192.Uijl E, Roksnoer LC, Hoorn EJ, Danser AH. From ARB to ARNI in Cardiovascular Control. Current hypertension reports. 2016; 18(12):86. [PubMed: 27837397]
193.Vardeny O, Miller R, Solomon SD. Combined neprilysin and renin-angiotensin system inhibition for the treatment of heart failure. JACC. Heart failure. 2014; 2(6):663–70. [PubMed: 25306450]
194.Ando S, Rahman MA, Butler GC, Senn BL, Floras JS. Comparison of candoxatril and atrial natriuretic factor in healthy men. Effects on hemodynamics, sympathetic activity, heart rate variability, and endothelin. Hypertension. 1995; 26(6 Pt 2):1160–6. [PubMed: 7498988]
195.McDowell G, Nicholls DP. The endopeptidase inhibitor, candoxatril, and its therapeutic potential in the treatment of chronic cardiac failure in man. Expert opinion on investigational drugs. 1999; 8(1):79–84. [PubMed: 15992061]
196.Cleland JG, Swedberg K. Lack of efficacy of neutral endopeptidase inhibitor ecadotril in heart failure. The International Ecadotril Multi-centre Dose-ranging Study Investigators. Lancet. 1998; 351(9116):1657–8. [PubMed: 9620738]
197.Campese VM, Lasseter KC, Ferrario CM, Smith WB, Ruddy MC, Grim CE, Smith RD, Vargas R, Habashy MF, Vesterqvist O, Delaney CL, Liao WC. Omapatrilat versus lisinopril: efficacy and neurohormonal profile in salt-sensitive hypertensive patients. Hypertension. 2001; 38(6):1342–8. [PubMed: 11751715]
198.Ferrario CM, Averill DB, Brosnihan KB, Chappell MC, Iskandar SS, Dean RH, Diz DI. Vasopeptidase inhibition and Ang-(1-7) in the spontaneously hypertensive rat. Kidney Int. 2002; 62(4):1349–57. [PubMed: 12234305]
199.Ferrario CM, Smith RD, Brosnihan B, Chappell MC, Campese VM, Vesterqvist O, Liao WC, Ruddy MC, Grim CE. Effects of omapatrilat on the renin-angiotensin system in salt-sensitive hypertension. Am J Hypertens. 2002; 15(6):557–64. [PubMed: 12074359]
200.Eisenstein EL, Nelson CL, Simon TA, Smitten AL, Lapuerta P, Mark DB. Vasopeptidase inhibitor reduces inhospital costs for patients with congestive heart failure: results from the IMPRESS trial. Inhibition of Metallo Protease by BMS-186716 in a Randomized Exercise and Symptoms Study in Subjects With Heart Failure. American heart journal. 2002; 143(6):1112–7. [PubMed: 12075271]
201.Packer M, Califf RM, Konstam MA, Krum H, McMurray JJ, Rouleau JL, Swedberg K. Comparison of omapatrilat and enalapril in patients with chronic heart failure: the Omapatrilat Versus Enalapril Randomized Trial of Utility in Reducing Events (OVERTURE). Circulation. 2002; 106(8):920–6. [PubMed: 12186794]
202.Pickering TG. Effects of stress and behavioral interventions in hypertension: the rise and fall of omapatrilat. Journal of clinical hypertension. 2002; 4(5):371–3. [PubMed: 12368583]
203.Ruilope LM, Dukat A, Bohm M, Lacourciere Y, Gong J, Lefkowitz MP. Blood-pressure reduction with LCZ696, a novel dual-acting inhibitor of the angiotensin II receptor and neprilysin: a randomised, double-blind, placebo-controlled, active comparator study. Lancet. 2010; 375(9722): 1255–66. [PubMed: 20236700]
204.Solomon SD, Claggett B, McMurray JJ, Hernandez AF, Fonarow GC. Combined neprilysin and renin-angiotensin system inhibition in heart failure with reduced ejection fraction: a meta- analysis. Eur J Heart Fail. 2016; 18(10):1238–1243. [PubMed: 27364182]
Ferrario and Mullick Page 30
205.Solomon SD, Zile M, Pieske B, Voors A, Shah A, Kraigher-Krainer E, Shi V, Bransford T, Takeuchi M, Gong J, Lefkowitz M, Packer M, McMurray JJ. A.w.A.R.B.o.M.O.h.f.w.p.e.f.I. Prospective comparison of. The angiotensin receptor neprilysin inhibitor LCZ696 in heart failure with preserved ejection fraction: a phase 2 double-blind randomised controlled trial. Lancet. 2012; 380(9851):1387–95. [PubMed: 22932717]
206.Kario K, Sun N, Chiang FT, Supasyndh O, Baek SH, Inubushi-Molessa A, Zhang Y, Gotou H, Lefkowitz M, Zhang J. Efficacy and safety of LCZ696, a first-in-class angiotensin receptor neprilysin inhibitor, in Asian patients with hypertension: a randomized, double-blind, placebo- controlled study. Hypertension. 2014; 63(4):698–705. [PubMed: 24446062]
207.McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, Zile MR. P-H Investigators Committees. Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med. 2014; 371(11):993–1004. [PubMed: 25176015]
208.Yusuf S, Pitt B, Davis CE, Hood WB, Cohn JN. Effect of enalapril on survival in patients with reduced left ventricular ejection fractions and congestive heart failure. The New England journal of medicine. 1991; 325(5):293–302. [PubMed: 2057034]
209.Feldman AM, Haller JA, DeKosky ST. Valsartan/Sacubitril for Heart Failure: Reconciling Disparities Between Preclinical and Clinical Investigations. JAMA. 2016; 315(1):25–6. [PubMed: 26641736]
210.Langenickel TH, Tsubouchi C, Ayalasomayajula S, Pal P, Valentin MA, Hinder M, Jhee S, Gevorkyan H, Rajman I. The effect of LCZ696 (sacubitril/valsartan) on amyloid-beta concentrations in cerebrospinal fluid in healthy subjects. British journal of clinical pharmacology. 2016; 81(5):878–90. [PubMed: 26663387]
211.Cannon JA, Shen L, Jhund PS, Kristensen SL, Kober L, Chen F, Gong J, Lefkowitz MP, Rouleau JL, Shi VC, Swedberg K, Zile MR, Solomon SD, Packer M, McMurray JJ. Dementia-related adverse events in PARADIGM-HF and other trials in heart failure with reduced ejection fraction. European journal of heart failure. 2017; 19(1):129–137. [PubMed: 27868321]
212.Lalouel JM, Rohrwasser A. Genetic susceptibility to essential hypertension: insight from angiotensinogen. Hypertension. 2007; 49(3):597–603. [PubMed: 17242300]
213.Dickson ME, Sigmund CD. Genetic basis of hypertension: revisiting angiotensinogen. Hypertension. 2006; 48(1):14–20. [PubMed: 16754793]
214.Zhou A, Carrell RW, Murphy MP, Wei Z, Yan Y, Stanley PL, Stein PE, Broughton Pipkin F, Read RJ. A redox switch in angiotensinogen modulates angiotensin release. Nature. 2010; 468(7320): 108–11. [PubMed: 20927107]
215.Ward K, Hata A, Jeunemaitre X, Helin C, Nelson L, Namikawa C, Farrington PF, Ogasawara M, Suzumori K, Tomoda S, et al. A molecular variant of angiotensinogen associated with preeclampsia. Nature genetics. 1993; 4(1):59–61. [PubMed: 8513325]
216.Corvol P, Jeunemaitre X. Molecular genetics of human hypertension: role of angiotensinogen. Endocrine reviews. 1997; 18(5):662–77. [PubMed: 9331547]
217.Kim HS, Krege JH, Kluckman KD, Hagaman JR, Hodgin JB, Best CF, Jennette JC, Coffman TM, Maeda N, Smithies O. Genetic control of blood pressure and the angiotensinogen locus. Proceedings of the National Academy of Sciences of the United States of America. 1995; 92(7): 2735–9. [PubMed: 7708716]
218.Phillips MI, Wielbo D, Gyurko R. Antisense inhibition of hypertension: a new strategy for renin- angiotensin candidate genes. Kidney international. 1994; 46(6):1554–6. [PubMed: 7700004]
219.Gyurko R, Wielbo D, Phillips MI. Antisense inhibition of AT1 receptor mRNA and angiotensinogen mRNA in the brain of spontaneously hypertensive rats reduces hypertension of neurogenic origin. Regulatory peptides. 1993; 49(2):167–74. [PubMed: 8134617]
220.Wielbo D, Sernia C, Gyurko R, Phillips MI. Antisense inhibition of hypertension in the spontaneously hypertensive rat. Hypertension. 1995; 25(3):314–9. [PubMed: 7875755]
221.Wielbo D, Simon A, Phillips MI, Toffolo S. Inhibition of hypertension by peripheral administration of antisense oligodeoxynucleotides. Hypertension. 1996; 28(1):147–51. [PubMed: 8675256]
Ferrario and Mullick Page 31
222.Tomita N, Morishita R, Higaki J, Aoki M, Nakamura Y, Mikami H, Fukamizu A, Murakami K, Kaneda Y, Ogihara T. Transient decrease in high blood pressure by in vivo transfer of antisense oligodeoxynucleotides against rat angiotensinogen. Hypertension. 1995; 26(1):131–6. [PubMed: 7541778]
223.Sugano M, Tsuchida K, Sawada S, Makino N. Reduction of plasma angiotensin II to normal levels by antisense oligodeoxynucleotides against liver angiotensinogen cannot completely attenuate vascular remodeling in spontaneously hypertensive rats. Journal of hypertension. 2000; 18(6):725–31. [PubMed: 10872557]
224.Makino N, Sugano M, Ohtsuka S, Sawada S, Hata T. Chronic antisense therapy for angiotensinogen on cardiac hypertrophy in spontaneously hypertensive rats. Cardiovascular research. 1999; 44(3):543–8. [PubMed: 10690286]
225.Makino N, Sugano M, Ohtsuka S, Sawada S. Intravenous injection with antisense oligodeoxynucleotides against angiotensinogen decreases blood pressure in spontaneously hypertensive rats. Hypertension. 1998; 31(5):1166–70. [PubMed: 9576130]
226.Tang X, Mohuczy D, Zhang YC, Kimura B, Galli SM, Phillips MI. Intravenous angiotensinogen antisense in AAV-based vector decreases hypertension. The American journal of physiology. 1999; 277(6 Pt 2):H2392–9. [PubMed: 10600860]
227.Boiziau C, Kurfurst R, Cazenave C, Roig V, Thuong NT, Toulme JJ. Inhibition of translation initiation by antisense oligonucleotides via an RNase-H independent mechanism. Nucleic acids research. 1991; 19(5):1113–9. [PubMed: 1850511]
228.Kole R, Krainer AR, Altman S. RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nature reviews. Drug discovery. 2012; 11(2):125–40. [PubMed: 22262036]
229.Bennett CF, Swayze EE. RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annual review of pharmacology and toxicology. 2010; 50:259–93.
230.Lu H, Wu C, Howatt DA, Balakrishnan A, Moorleghen JJ, Chen X, Zhao M, Graham MJ, Mullick AE, Crooke RM, Feldman DL, Cassis LA, Vander Kooi CW, Daugherty A. Angiotensinogen Exerts Effects Independent of Angiotensin II. Arteriosclerosis, thrombosis, and vascular biology. 2016; 36(2):256–65.
231.Ravichandran K, Ozkok A, Wang Q, Mullick AE, Edelstein CL. Antisense-mediated angiotensinogen inhibition slows polycystic kidney disease in mice with a targeted mutation in Pkd2. American journal of physiology. Renal physiology. 2015; 308(4):F349–57. [PubMed: 25537744]
232.Saigusa T, Dang Y, Mullick AE, Yeh ST, Zile MR, Baicu CF, Bell PD. Suppressing angiotensinogen synthesis attenuates kidney cyst formation in a Pkd1 mouse model. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2016; 30(1):370–9. [PubMed: 26391272]
233.Olearczyk J, Gao S, Eybye M, Yendluri S, Andrews L, Bartz S, Cully D, Tadin-Strapps M. Targeting of hepatic angiotensinogen using chemically modified siRNAs results in significant and sustained blood pressure lowering in a rat model of hypertension. Hypertension research: official journal of the Japanese Society of Hypertension. 2014; 37(5):405–12. [PubMed: 24335718]
234.Haase N, Foster D, Bercher J, Milstein S, Golic M, Charisse K, Rugor J, Kuchimanchi S, Przybyl L, Bettencourt B, Müller DN, Hinkle G, Dechend R. RNAi Therapeutics Targeting Human Angiotensinogen (hAGT) Ameliorate Preeclamptic Sequelae in an Established Transgenic Rodent Model for Preeclampsia. Hypertension. 2014; 64 Abstract 666.
235.Mullick AE, Yeh ST, Graham MJ, Crooke RM. Liver-specific antisense inhibition of angiotensinogen reduced BP in a hypertensive rat model resistant to standard RAS inhibition. Hypertension. 2015; 66(Suppl 1) Abstract P605.
236.Mullick AE, Brown HL, Graham MJ, Crooke RM. Antisense Inhibition Of Angiotensinogen Reduces BP In Normotensive Sprague-Dawley Rats And Effectively Eliminates RAS-dependent BP Control In Hypertensive SHR Rats. Hypertension. 2012; 60 Abstract 627.
237.Crackower MA, Sarao R, Oudit GY, Yagil C, Kozieradzki I, Scanga SE, Oliveira-dos-Santos AJ, da Costa J, Zhang L, Pei Y, Scholey J, Ferrario CM, Manoukian AS, Chappell MC, Backx PH,
Ferrario and Mullick Page 32
Yagil Y, Penninger JM. Angiotensin-converting enzyme 2 is an essential regulator of heart function. Nature. 2002; 417(6891):822–8. [PubMed: 12075344]
238.Turner AJ, Hiscox JA, Hooper NM. ACE2: from vasopeptidase to SARS virus receptor. Trends Pharmacol Sci. 2004; 25(6):291–4. [PubMed: 15165741]
239.Carey RM, Padia SH. Role of angiotensin AT(2) receptors in natriuresis: Intrarenal mechanisms and therapeutic potential. Clin Exp Pharmacol Physiol. 2013; 40(8):527–34. [PubMed: 23336117]
240.Chow BS, Allen TJ. Angiotensin II type 2 receptor (AT2R) in renal and cardiovascular disease. Clin Sci (Lond). 2016; 130(15):1307–26. [PubMed: 27358027]
241.Menk M, Graw JA, von Haefen C, Sifringer M, Schwaiberger D, Unger T, Steckelings U, Spies CD. Stimulation of the Angiotensin II AT2 Receptor is Anti-inflammatory in Human Lipopolysaccharide-Activated Monocytic Cells. Inflammation. 2015; 38(4):1690–9. [PubMed: 25758542]
242.Kemp BA, Howell NL, Gildea JJ, Keller SR, Padia SH, Carey RM. AT(2) receptor activation induces natriuresis and lowers blood pressure. Circ Res. 2014; 115(3):388–99. [PubMed: 24903104]
243.Sumners C, de Kloet AD, Krause EG, Unger T, Steckelings UM. Angiotensin type 2 receptors: blood pressure regulation and end organ damage. Curr Opin Pharmacol. 2015; 21:115–21. [PubMed: 25677800]
244.Tamargo J, Duarte J, Ruilope LM. New antihypertensive drugs under development. Curr Med Chem. 2015; 22(3):305–42. [PubMed: 25386825]
245.Wysocki J, Ye M, Rodriguez E, Gonzalez-Pacheco FR, Barrios C, Evora K, Schuster M, Loibner H, Brosnihan KB, Ferrario CM, Penninger JM, Batlle D. Targeting the degradation of angiotensin II with recombinant angiotensin-converting enzyme 2: prevention of angiotensin II-dependent hypertension. Hypertension. 2010; 55(1):90–8. [PubMed: 19948988]
246.Haschke M, Schuster M, Poglitsch M, Loibner H, Salzberg M, Bruggisser M, Penninger J, Krahenbuhl S. Pharmacokinetics and pharmacodynamics of recombinant human angiotensin- converting enzyme 2 in healthy human subjects. Clin Pharmacokinet. 2013; 52(9):783–92. [PubMed: 23681967]
247.Machado-Silva A, Passos-Silva D, Santos RA, Sinisterra RD. Therapeutic uses for Angiotensin- (1-7). Expert Opin Ther Pat. 2016; 26(6):669–78. [PubMed: 27121991]
248.Folkow B. The haemodynamic consequences of adaptive structural changes of the resistance vessels in hypertension. Clin Sci. 1971; 41(1):1–12. [PubMed: 5559111]
249.Folkow B, Sivertsson R. Hemodynamic consequences of adaptive reconstruction of resistance vessels. Lakartidningen. 1971; 68(45):5165–73. [PubMed: 5137385]
250.Smith RD, Yokoyama H, Averill DB, Schiffrin EL, Ferrario CM. Reversal of vascular hypertrophy in hypertensive patients through blockade of angiotensin II receptors. J Am Soc Hypertens. 2008; 2(3):165–72. [PubMed: 20409899]
251.Schiffrin EL. Vascular changes in hypertension in response to drug treatment: Effects of angiotensin receptor blockers. Can J Cardiol. 2002; 18(Suppl A):15A–18A.
252.Schiffrin EL. Effects of antihypertensive drugs on vascular remodeling: do they predict outcome in response to antihypertensive therapy? Curr Opin Nephrol Hypertens. 2001; 10(5):617–24. [PubMed: 11496055]
253.Schiffrin EL. Small artery remodeling in hypertension: can it be corrected? Am J Med Sci. 2001; 322(1):7–11. [PubMed: 11465250]
254.Abadir PM, Walston JD, Carey RM. Subcellular characteristics of functional intracellular renin- angiotensin systems. Peptides. 2012; 38(2):437–45. [PubMed: 23032352]
255.Baker KM, Chernin MI, Schreiber T, Sanghi S, Haiderzaidi S, Booz GW, Dostal DE, Kumar R. Evidence of a novel intracrine mechanism in angiotensin II-induced cardiac hypertrophy. Regul Pept. 2004; 120(1–3):5–13. [PubMed: 15177915]
256.Kumar R, Singh VP, Baker KM. The intracellular renin-angiotensin system: a new paradigm. Trends Endocrinol Metab. 2007; 18(5):208–14. [PubMed: 17509892]
Ferrario and Mullick Page 33
257.Re RN. The cellular biology of angiotensin: Paracrine, autocrine and intracrine actions in cardiovascular tissues. Journal of Molecular and Cellular Cardiology. 1989; 21:63–69. [PubMed: 2697759]
258.Wilson BA, Cruz-Diaz N, Su Y, Rose JC, Gwathmey TM, Chappell MC. Angiotensinogen Import in Isolated Proximal Tubules: Evidence for Mitochondrial Trafficking and Uptake. Am J Physiol Renal Physiol. 2016 ajprenal 00246 2016.
259.Wang H, Jessup JA, Zhao Z, Da Silva J, Lin M, MacNamara LM, Ahmad S, Chappell MC, Ferrario CM, Groban L. Characterization of the cardiac renin angiotensin system in oophorectomized and estrogen-replete mRen2.Lewis rats. PLoS One. 2013; 8(10):e76992. [PubMed: 24204720]
260.Alzayadneh EM, Chappell MC. Nuclear expression of renin-angiotensin system components in NRK-52E renal epithelial cells. J Renin Angiotensin Aldosterone Syst. 2015; 16(4):1135–48. [PubMed: 24961503]
261.Ferrario CM, Jessup J, Chappell MC, Averill DB, Brosnihan KB, Tallant EA, Diz DI, Gallagher PE. Effect of angiotensin-converting enzyme inhibition and angiotensin II receptor blockers on cardiac angiotensin-converting enzyme 2. Circulation. 2005; 111(20):2605–10. [PubMed: 15897343]
262.Varagic J, Ahmad S, VonCannon JL, Moniwa N, Brosnihan KB, Wysocki J, Batlle D, Ferrario CM. Predominance of AT(1) blockade over mas-mediated angiotensin-(1-7) mechanisms in the regulation of blood pressure and renin-angiotensin system in mRen2.Lewis rats. Am J Hypertens. 2013; 26(5):583–90. [PubMed: 23459599]
263.Urata H, Kinoshita A, Perez DM, Misono KS, Bumpus FM, Graham RM, Husain A. Cloning of the gene and cDNA for human heart chymase. J Biol Chem. 1991; 266(26):17173–9. [PubMed: 1894611]
264.Wei CC, Hase N, Inoue Y, Bradley EW, Yahiro E, Li M, Naqvi N, Powell PC, Shi K, Takahashi Y, Saku K, Urata H, Dell』italia LJ, Husain A. Mast cell chymase limits the cardiac efficacy of Ang
I-converting enzyme inhibitor therapy in rodents. J Clin Invest. 2010; 120(4):1229–39. [PubMed: 20335663]
265.Jin D, Takai S, Okamoto Y, Muramatsu M, Miyazaki M. Chymase-derived angiotensin II and arrhythmias after myocardial infarction. Nihon Yakurigaku Zasshi. 2004; 124(2):77–82. [PubMed: 15277725]
266.Jin D, Takai S, Sakaguchi M, Okamoto Y, Muramatsu M, Miyazaki M. An antiarrhythmic effect of a chymase inhibitor after myocardial infarction. J Pharmacol Exp Ther. 2004; 309(2):490–7. [PubMed: 14730006]
267.Jin D, Takai S, Yamada M, Sakaguchi M, Kamoshita K, Ishida K, Sukenaga Y, Miyazaki M. Impact of chymase inhibitor on cardiac function and survival after myocardial infarction. Cardiovasc Res. 2003; 60(2):413–20. [PubMed: 14613871]
268.Takai S, Jin D. Improvement of cardiovascular remodelling by chymase inhibitor. Clin Exp Pharmacol Physiol. 2016; 43(4):387–93. [PubMed: 26798995]
269.Takai S, Jin D, Miyazaki M. Targets of chymase inhibitors. Expert Opin Ther Targets. 2011; 15(4):519–27. [PubMed: 21291347]
270.Takai S, Jin D, Muramatsu M, Miyazaki M. Chymase as a novel target for the prevention of vascular diseases. Trends Pharmacol Sci. 2004; 25(10):518–22. [PubMed: 15380935]
271.Takai S, Jin D, Muramatsu M, Okamoto Y, Miyazaki M. Therapeutic applications of chymase inhibitors in cardiovascular diseases and fibrosis. Eur J Pharmacol. 2004; 501(1–3):1–8. [PubMed: 15464056]
272.Pitt B, Segal R, Martinez FA, Meurers G, Cowley AJ, Thomas I, Deedwania PC, Ney DE, Snavely DB, Chang PI. Randomised trial of losartan versus captopril in patients over 65 with heart failure (Evaluation of Losartan in the Elderly Study ELITE). Lancet. 1997; 349(9054):747– 52. [PubMed: 9074572]
273.Pitt B, Poole-Wilson PA, Segal R, Martinez FA, Dickstein K, Camm AJ, Konstam MA, Riegger G, Klinger GH, Neaton J, Sharma D, Thiyagarajan B. Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomised trial--the Losartan Heart Failure Survival Study ELITE II. Lancet. 2000; 355(9215):1582–7. [PubMed: 10821361]
Ferrario and Mullick Page 34
274.Yusuf S, Teo K, Anderson C, Pogue J, Dyal L, Copland I, Schumacher H, Dagenais G, Sleight P. for the Telmisartan Randomised AssessmeNt Study in A. C. E. iNtolerant subjects with cardiovascular Disease Investigators. Effects of the angiotensin-receptor blocker telmisartan on cardiovascular events in high-risk patients intolerant to angiotensin-converting enzyme inhibitors: a randomised controlled trial. Lancet. 2008; 372(9644):1174–83. [PubMed: 18757085]
275.Dickstein K, Kjekshus J. the Optimaal Steering Committee, for the OPTIMAAL Study Group. Effects of losartan and captopril on mortality and morbidity in high-risk patients after acute myocardial infarction: the OPTIMAAL randomised trial. Optimal Trial in Myocardial Infarction with Angiotensin II Antagonist Losartan. Lancet. 2002; 360(9335):752–60. [PubMed: 12241832]
Ferrario and Mullick Page 35
Figure 1.
Schematic diagram of currently characterized biotransformation pathways accounting for the formation of the biological active peptides Ang-(1-9), Ang II, and Ang-(1-7). Peptides with substitution of aspartic to alanine in position 1 of the Ang II and Ang-(1-7) molecules should be viewed as secondary processing pathways.
Ferrario and Mullick Page 36
Figure 2.
Relative risk and 95 % confidence intervals of the effect of Ang II receptor blockers on primary cardiac end points of large randomized clinical trials. Acronyms are: CHARM- Alternative, Candesartan in Heart failure: Assessment of Reduction in Mortality and morbidity [164]; CHARM-Added, Candesartan in Heart failure: Assessment of Reduction in Mortality and morbidity [165]; ELITE, Evaluation of Losartan in the Elderly Study [272]; ELITE II, the Losartan Heart Failure Survival Study (Evaluation of Losartan in the Elderly Study) [273]; HEAAL, Heart failure Endpoint evaluation of Ang II Antagonist Losartan [158]; I-PRESERVE, Irbesartan in Heart Failure with Preserved Ejection Fraction Study [169]; LIFE, Losartan Intervention For Endpoint reduction Study [157]; ONTARGET, The Ongoing Telmisartan Alone and in Combination with Ramipril Global Endpoint Trial [274]; OPTIMAAL, Optimal Trial in Myocardial Infarction with the Ang II Antagonist Losartan [275]; TRASCEND, Telmisartan Randomized Assessment Study in ACE Intolerant subjects with cardiovascular Disease [274]; TROPHY, Trial of Preventing Hypertension [162]; VAL- HEFT, Valsartan Heart Failure Trial [159]; VALIANT, Valsartan in Acute Myocardial Infarction trial [163]; VALUE, Valsartan Antihypertensive Long-term Use Evaluation study [161].
Ferrario and Mullick Page 37
Table 1
Reported Side Effects of FDA Approved Renin Angiotensin Aldosterone System Inhibitors
DRUG CLASS
REPORTED ADVERSE EFFECTS (1)
Angiotensin Converting Enzyme Inhibitors
[Available in the US are: benazepril (Lotensin™), captopril (Capoten™), enalapril (Vasotec™, Epaned™), fosinopril (Monopril™), lisinopril (Prinivil™, Zestril™), moexipril (Univasc™), perindopril (Aceon™), quinapril (Accupril™), ramipril (Altace™), trandolapril (Mavik™)]
Dried cough (5%–25% of subjects), hyperkalemia, dizziness (lightheadedness or faintness upon rising), headache, drowsiness, diarrhea, low blood pressure, weakness, loss of taste (salty or metallic taste), and rash. Chest pain, increased uric acid levels, sun sensitivity, and increased BUN and creatinine levels have been reported. More serious but rare side effects include kidney failure, allergic reactions (angioedema), a decrease in white blood cells, pancreatitis, and liver dysfunction.
Aldosterone Receptor Antagonists
[Available in the US are: eplerenone (Inspra™) and spironolactone (Aldactone™)]
Acne, alopecia, edema, gender dysphoria (amenorrhea, breast enlargement in men), heart failure, hypertension, hirsutism, hypokalemia. Side effects are less severe with eplerenone.
Angiotensin Receptor Blockers
[Available in the US are: candesartan (Atacand™), eprosartan (Teveten™), irbesartan (Avapro™), losartan (Cozaar™), olmesartan (Benicar™), telmisartan (Micardis™) and, valsartan (Diovan™), azilsartan (Edarbi™).
Headache, fainting, dizziness, nasal congestion, diarrhea, back pain, leg pain. Angioedema and hyperkalemia reported rarely.
Direct Renin Inhibitors
[Available in the US is: aliskiren (Tekturna™)
Side effects comparable to those encountered with ARBs. Diarrhea, however, reported more frequently.
Angiotensin receptor neprilysin inhibitor (ARNI)
Hypotension, hyperkalemia, cough, dizziness, and rarely kidney failure.
[Available in the US is sacubitril/valsartan (Entresto™)
(1)Prescribing information may be consulted for detailed explanation of side-effects.