key: cord-0006243-hynkb0a8 authors: Acharya, K. Ravi; Sturrock, Edward D.; Riordan, James F.; Ehlers, Mario R. W. title: Ace revisited: A new target for structure-based drug design date: 2003 journal: Nat Rev Drug Discov DOI: 10.1038/nrd1227 sha: a570ae9a59df3c57e076252949e6036bdfb5e0e6 doc_id: 6243 cord_uid: hynkb0a8 Current-generation angiotensin-converting enzyme (ACE) inhibitors are widely used for cardiovascular diseases, including high blood pressure, heart failure, heart attack and kidney failure, and have combined annual sales in excess of US $6 billion. However, the use of these ACE inhibitors, which were developed in the late 1970s and early 1980s, is hampered by common side effects. Moreover, we now know that ACE actually consists of two parts (called the N- and C-domains) that have different functions. Therefore, the design of specific domain-selective ACE inhibitors is expected to produce next-generation drugs that might be safer and more effective. Here we discuss the structural features of current inhibitors and outline how next-generation ACE inhibitors could be designed by using the three-dimensional molecular structure of human testis ACE. The ACE structure provides a unique opportunity for rational drug design, based on a combination of in silico modelling using existing inhibitors as scaffolds and iterative lead optimization to drive the synthetic chemistry. The neural activity of the sympathetic nervous system, regulating (through adrenergic receptors) cardiac and vascular function. The resistance to blood flow, which is directly proportional to the extent of vasoconstriction, is one of the primary determinants of blood pressure. angiotensin I (Ang I) and an octapeptide called angiotensin II (Ang II) 15 . Two years later a chloride-dependent, metalloenzyme that could convert Ang I to Ang II was purified from horse plasma 16 , and was later referred to as ACE. ACE, also known as peptidyl-dipeptidase A (EC 3.4.15.1), belongs to the M2 family of the MA clan (a protein clan contains all the modern-day polypeptides that have arisen from a single evolutionary progenitor) of zinc metallopeptidases 17 . It is a dipeptidyl carboxypeptidase that catalyses the hydrolytic cleavage of dipeptides from the carboxyl terminus of a wide variety of oligopeptides in vitro. Its best known function is the in vivo conversion of Ang I (DRVYIHPFHL), which circulates in plasma, into the potent vasopressor Ang II by removal of the C-terminal His-Leu (FIG. 1) . Ang I is generated primarily by the renin-catalysed hydrolysis of the Leu10-Val11 peptide bond of angiotensinogen, a liver-derived 55-kDa plasma protein. ACE also affects blood pressure by cleaving bradykinin (BK, RPPGFSPFR), thereby abolishing its vasodilating activity (because of this, ACE is sometimes referred to as kininase II (kininase I is carboxypeptidase N)). BK is generated from a kininogen precursor by the action of plasma kallikrein (a serine proteinase) in a process analogous to that of Ang I formation (FIG. 1) . Human ACE is a monomeric zinc metalloenzyme that is synthesized as a 1,306-amino-acid polypeptide, is processed to a 1,277-residue mature form, is heavily glycosylated (30% by weight), and is localized to the plasma membrane of endothelial and absorptive epithelial and neuroepithelial cells 18 . A sequence of 22 hydrophobic amino acids located near the carboxyl terminus of the protein serves as a transmembrane domain that anchors ACE to the cell surface. This creates a 28-residue cytosolic domain and a 1,227-residue glycosylated extracellular domain. Its cellular orientation defines ACE as a type I transmembrane ectoprotein and, in the case of endothelial cells, positions it optimally for interaction with its circulating substrates. The amino acid sequence of human endothelial ACE provides clear evidence for a gene duplication event in its evolutionary history. It consists of an N-terminal domain of about 612 amino acids, a 15-residue interdomain sequence and a 650-residue C-terminal domain. A 357-aminoacid segment of the N-domain has more than 60% sequence identity to the corresponding segment of the C-domain (FIG. 2) . Each domain contains a five-residue sequence of amino acids, HEMGH, which is characteristic of catalytic zinc sites found in a large family of neutral endoproteinases. Detailed kinetic and mutational analyses have established that both of the zinc sites have catalytic activity 19, 20 . The physiological consequence of such tandem active sites in an enzyme is unknown. Some differences in catalytic properties have been observed for these two sites: the N-domain site is notably 50-times more active toward the haemoregulatory peptide N-acetyl-Ser-Asp-Lys-Pro (AcSDKP) 21 , 1000-times Here, we provide an overview of ACE and the RAS, current ACE inhibitors and their clinical utility, insights from the tACE crystal structure, and the rationale and prospects for developing second-generation, domainselective inhibitors by structure-guided design. The recognition that an elevated basal blood pressure can lead to a shortened life expectancy and higher morbidity (due to cardiovascular complications such as kidney failure, heart failure and stroke) only evolved during the first half of the twentieth century, when numerous clinical surveys demonstrated a normal distribution of blood pressures and a 10-20% prevalence of hypertension in an apparently healthy population 9 . Hypertension occurs in millions of people worldwide, the large majority of whom are unaware of their condition; a recent report indicated that prevalence rates are even higher than previously suspected, at 28% in North America and 44% in Europe 10 . Many long-term studies were required before the increased morbidity and mortality associated with hypertension became accepted and it was classified as a disease in need of treatment 9 . The underlying cause of hypertension in most cases was, and still is, unknown. Therapy was initially directed at reducing blood volume by the use of DIURETICS, SYMPATHETIC TONE by the use of adrenergic-blocking agents, or VASCULAR RESISTANCE by the use of vasodilators. A major breakthrough in our understanding of blood pressure regulation came with the discovery of the RAS and ACE (reviewed in REF. 11 ). On the basis of a suspected relationship between kidney and blood pressure, Goldblatt, Houssay, Page and others identified the renal enzyme renin and the pressor substance angiotensin in the 1930s and 1940s [12] [13] [14] . In the mid-1950s, Skeggs and co-workers purified angiotensin and found that it existed in two forms: a decapeptide called is much less so 19, 24 . However, given the relative constancy of plasma chloride concentration, the significance of this chloride dependency is not obvious. Human endothelial ACE, also known as somatic ACE, is encoded by a single gene that consists of 26 exons, more sensitive to inhibition by the phosphinic peptide RXP407 (REF. 22) , and more than 3000-times less sensitive to inhibition by RXPA380 (REF. 23 ACE is released from the endothelial cell surface by the action of a so-called ACE-secretase, or sheddase, that cleaves the Arg1203-Ser1204 peptide bond near the transmembrane region 32, 33 . Soluble ACE is a minor component of total ACE activity, and its physiological function is unknown. Activators of protein kinase C stimulate the release of ACE from endothelial and other cells in culture, and markedly elevated concentrations of plasma ACE are observed in certain diseases, particularly sarcoidosis. Besides its role in blood pressure regulation, ACE also has been postulated to participate in 'local' or 'tissue' RASs. The Ang II arising from such systems is thought to act as a paracrine growth factor 4 . This activity has been implicated in the development of left ventricular dysfunction that can occur after a major heart attack, and seems to account for the beneficial effects of ACE inhibitor therapy in such situations. The structural and functional conservation of the gene encoding ACE are indications of its widespread evolutionary importance as a key metallopeptidase involved in the metabolism of regulatory peptides 34 . The Drosophila melanogaster genome contains six genes (Acer, Ance, Ance-2, Ance-3, Ance-4 and Ance-5) that encode ACE-like proteins. Two of these, angiotensinconverting enzyme (Ance) and angiotensin-converting enzyme-related (Acer), are enzymatically active and share 36% sequence identity with human ACE 35 . Ance displays properties very similar to those of the human C-domain, whereas Acer is inhibited by the N-domainselective inhibitor RXP407 (REF. 36 ). ACE2 (also known as ACEH) is an ACE homologue found in humans and rodents that functions as a carboxypeptidase with a preference for C-terminal hydrophobic or basic residues (reviewed in REFS 3, 37) . It is expressed mainly in the heart, kidney and testis and is important for the regulation of blood pressure and cardiac function. Interestingly, ACE2 is not inhibited by ACE inhibitors such as lisinopril, captopril and enalaprilat. One of the main impediments to determining the structure of ACE by X-ray crystallography was the production of diffraction-quality crystals. The C-domain of ACE has seven potential N-linked glycosylation sites, six of which are located on the surface of the protein and the seventh in the juxtamembrane region. Two concurrent approaches succeeded in paving the way for the crystallization 38 and successful X-ray structure determination of tACE 5 . Five of the N-linked sites were disrupted by substituting glutamines for each of the asparagine residues in the glycosylation sequences and a truncated form was expressed in the presence of a glucosidase inhibitor yielding crystals suitable for X-ray diffraction. The latter protein was used for the subsequent threedimensional structure determination of tACE. Highresolution crystal structures of the human tACE and its complex with the widely used inhibitor lisinopril at 2.0 Å resolution were recently reported 5 . all but one of which, exon 13, is transcribed into the corresponding messenger RNA 25 . A form of ACE found only in adult testis (tACE) is encoded by the same gene but its mRNA begins before exon 13 and continues through exon 26. It is transcribed by the interaction of a promoter with a site present in intron 12 that is active only in adult male germinal cells 25 . Translation of this mRNA results in a 701-amino-acid version of ACE that, except for the first 36 residues, is identical to the C-terminal domain of somatic ACE 26 . Testis or germinal ACE is found in developing sperm cells and mature sperm. Sperm lacking ACE are deficient in transport within the oviduct and in binding to the zonae pellucidae, and male ACE -/mice have markedly diminished fertility 27 . A polymorphism involving a 287-base-pair insertion corresponding to an Alu repetitive sequence has been found in intron 16 of the human ACE gene 28 . The insertion is all or none: in one study the (I)/(D) frequency ratio was 44:56. The deletion (D) allele of the ACE gene lacks the sequence that is present in the insertion (I) allele. The D allele is associated with a higher plasma and tissue ACE activity. The DD genotype is positively correlated with plasma ACE activity and numerous attempts have been made to search for associations between the DD genotype and specific cardiovascular diseases, but the conclusions remain controversial 29 . The association of allele I with athletic performance is equally controversial. Some studies have shown that this polymorphism is associated with endurance performance, whereas others have shown the D allele to be associated with that of elite shortdistance athletes 30,31 . notable differences in hydrophobicity and charge are observed in the lid-like structure comprising helices α1, α2 and α3 (using tACE nomenclature; FIG. 2 ). This part of the structure seems to affect the substrate specificity of the N-or C-domain, as tACE mutants which had this region replaced by the corresponding N-domain sequence showed a preference for N-domain substrates 43 (Z. L. Woodman et al., unpublished data). Second, the Zn 2+ -binding site with the canonical HEXXH motif is conserved. Third, it has been shown that the C-domain has greater chloride dependence than the N-domain, both in terms of substrate hydrolysis and inhibitor binding 19, 24 . From the model we can predict that Arg186 (a key residue for binding one of the two chloride ions in tACE) is replaced by His164 in the N-domain. So, in the N-domain only one chloride-ion-binding pocket is plausible, involving Arg500 and Tyr202. Fourth, positioning of the lisinopril molecule in the active site of the N-domain model revealed that the full complement of structurally conserved residues was found as observed in the tACE structure, confirming that the N-domain of somatic ACE could also bind lisinopril with similar affinity, as previously reported 24, 44 . Fifth, the S 2 sub-site is formed by Asn494 and Thr496, which replace a serine and a valine, respectively, in the C-domain. Furthermore, the asparagine occurs in an N-GLYCOSYLATION SEQUON (NVT) that is unique to the N-domain; the attachment of any glycan to this residue would occlude the S 2 pocket. This, in part, might explain how the bulky aromatic ring of the benzamido group in keto-ACE (an ACE inhibitor; see FIG. 5a) is accommodated by the S 2 site in the C-domain (tACE), making keto-ACE more C-domain selective. Sixth, the ACE inhibitor RXP407 has an N-acetyl group that confers a certain degree of N-domain specificity 22 . The model of RXP407 and the N-domain reveals two residues, Arg381 and Tyr369, that form van der Waals interactions with the N-acetyl carbonyl/methyl groups (FIG. 5d) . However, a glutamate and phenylalanine are substituted for these two residues in the C-domain and might exert repulsive forces in the binding of the inhibitor to the C-domain active site. Homology modelling of ACE2 using the atomic coordinates of the tACE structure revealed differences in the ligand-binding pockets of the two homologues that account for their substrate and inhibitor selectivity 105 . First, the accommodation of the S 2 ′ sub-site in tACE increases the substrate-binding cavity and permits the binding of an additional amino acid to the obligatory binding site; second, Gln281, which interacts with the carboxyl terminus of lisinopril in tACE, is replaced by an arginine in ACE2. This represents an elongation of the side chain of residue 281, which causes steric conflict with the P 2 ′ residue of lisinopril when it is docked in the active site of the ACE2 model. In addition, the substitution of the leucine and phenylalanine for the hydrogen-bonding Lys511 and Tyr520, respectively, and replacement of Thr282 in tACE by the more bulky Phe274 residue, probably account for the changes in substrate specificity. There are no striking differences in the large S 1 ′ sub-sites of tACE and ACE2 except for a proline in ACE2 which corresponds to Ala354 in tACE. Drosophila ACE (Ance) contains a single domain similar to tACE. However, it has only three potential N-linked glycosylation sites, which are not required for secretion and enzymatic activity 39 . Recently, the crystal structures of this homologue (which has considerable similarity to the tACE structure), bound to captopril and lisinopril, were reported 40 . The wild-type protein was expressed in a baculovirus expression system and the oligosaccharides did not hamper crystallization. However, it is unlikely that this will be the case with the ACE2 homologue, the ACE N-domain or the full-length somatic ACE structures -ACE structural milestones that are still eagerly awaited. Prominent features of the tACE structure include an abundance of α-helices and a deep central cavity that divides the molecule into two halves, which pack together into an overall ellipsoid shape (FIG. 3) . The active site, identified by the catalytic Zn 2+ ion bound to the HEXXH sequence (and to lisinopril in its complex with the enzyme), is located in the deep cavity, some 10 Å from its entrance. The amino-terminal helices (α1-3) form a lid-like extension that partially covers the activesite channel, which limits the access of substrates and inhibitors. In fact, the aperture of the channel opening is approximately 3 Å in diameter, which is too small for most peptide substrates, indicating that tACE must undergo some conformational change, possibly associated with its unique chloride ion activation, for the substrate to enter the channel. From the structure of the lisinopril-tACE complex (FIG. 4) it is evident that the phenyl ring of the inhibitor interacts with the S 1 sub-site in the active site, the lysine with the S 1 ′ sub-site, and the proline occupies the S 2 ′ sub-site. The carboxyl group, located between the phenylpropyl group and the lysine, binds to the Zn 2+ in the active site and also forms a hydrogen bond with the side-chain carboxylate of Glu384. Other key interactions occur between the sidechain amino group of the inhibitor lysine and Glu162 of tACE, and between the C-terminal proline carboxyl group with Lys511 and Tyr520. The binding of the inhibitor to the S 1 , S 1 ′ and S 2 ′ pockets and its zinc coordination form the basis for the structure-guided design of improved domain-selective ACE inhibitors. In addition, two buried chloride ions that are important for the activation of the enzyme were identified in the crystal structure (outside the active site), both distant (20.7 Å and 10.4 Å, respectively) from the catalytic Zn 2+ ion. The first is bound to Arg186, Arg489 and Trp485, whereas the second is bound to Arg522 and Tyr224. The structure is indicative of an indirect mechanism for chloride activation, possibly through effects on active-site structure. This study also revealed that the structure of tACE (MA sub-clan of M clan of peptidases (M clan peptidases are metalloenzymes and the metal is involved in catalysis)) resembles that of rat neurolysin 41 and a newly identified carboxypeptidase from the hyperthermophilic archaeon Pyrococcus furiosus 42 -both of which are members of the MA clan -despite low sequence similarity. Structure-based modelling of the N-domain of somatic ACE (using the three-dimensional structure of tACE 5 , which has ~60% amino-acid sequence identity to somatic ACE) reveals some interesting features. First, GLYCOSYLATION SEQUON Consensus sequence Asn-X-Ser/Thr whose core glycosylation generally occurs at the Asn residue. Nevertheless, the use of CPA as a model led to key conceptual insights in inhibitor design, because CPA was much better understood and its crystal structure was known. Cushman and Ondetti reasoned that ACE was an exopeptidase like CPA, with the difference that ACE cleaved the penultimate peptide bond to release a dipeptide product. The second major breakthrough in inhibitor design derived from an earlier observation that an extract from the South American pit viper Bothrops jararaca, known as bradykinin potentiating factor (BPF), could inhibit ACE [56] [57] [58] . BPF was a mixture of peptides 59 , which were shown to be potent and specific inhibitors of ACE (TABLE 1), and structure-activity studies indicated that the optimal C-terminal inhibitory sequence was Phe-Ala-Pro 60 . This work led to the proposal that the venom peptides were substrate analogues that bound competitively to the obligatory substrate-binding sites in the ACE active site (FIG. 5a) . What was needed were orally active, non-peptide analogues of BPF. The third key insight derived from work by Byers and Wolfenden 61 describing a new design concept for inhibitors of CPA based on benzylsuccinic acid, referred to as by-product analogues. Cushman and Ondetti recognized that part of the binding affinity of benzylsuccinic acid derived from coordination of the active-site zinc by the carboxyl group, and predicted that a similar succinylamino acid derivative would inhibit ACE if its structure was analogous to the dipeptide product of ACE activity. On the basis of the Phe-Ala-Pro sequence derived from the BPF peptides, they synthesized methylsuccinyl-Pro (analogous to carboxy-Ala-Pro) and indeed found it to be a specific inhibitor, with an IC 50 of 22 µM 53 . Cushman and Ondetti then searched for a superior zinc-binding group and the potency breakthrough was achieved by replacing the carboxyl with a sulphydryl group, yielding captopril with an IC 50 of 23 nM 53, 54 (FIG. 5a) . Captopril became the first ACE inhibitor in clinical use (first approved in 1981) and rapidly established itself as a powerful new therapeutic agent in the treatment of hypertension and heart failure. Reports of captoprilrelated side effects, such as loss of taste and skin rash, prompted Patchett and colleagues to focus on the design of non-sulphydryl ACE inhibitors, by reverting to carboxyl compounds and introducing additional functionalities that would complete the by-product design. Captopril did not make use of at least two potential binding sites: the S 1 binding site and a hydrogen-bonding site for the amide nitrogen of the (substrate) scissile bond (FIG. 5a) . So, structure-activity studies were performed on a series of N-carboxyalkyl dipeptides of the general formula R-CHCO 2 H-A 1 -A 2 . The R group, occupying the S 1 pocket, was best served by benzylmethylene, whereas A 1 -A 2 was either Ala-Pro (as in the BPF peptides) or, surprisingly, Lys-Pro, leading to enalaprilat and lisinopril, respectively (FIG. 5a) 62 , which show nanomolar inhibition constants. Enalaprilat and lisinopril are essentially tripeptide analogues with a Zn 2+ -coordinating carboxyl group substituting for the substrate scissile amide carbonyl. Enalaprilat closely resembles the Phe-Ala-Pro sequence The story of the design and synthesis of the first orally active, potent inhibitors of ACE is one of the great success stories of modern medicinal chemistry. It has been described as one of the first examples of true 'rational drug design' 3 , and although this might not be true in the sense that we understand that term today, the design of the ACE inhibitors in the late 1970s and early 1980s was certainly based on a series of brilliant insights that, together with a sprinkling of serendipity, constituted what might now be called 'rational intuition' . It is not our intention to provide a comprehensive review of the events leading to the design and synthesis of captopril, enalaprilat and lisinopril, the original group of potent ACE inhibitors that formed the basis for all subsequent compounds of what can now be termed first-generation ACE inhibitors. Several excellent reviews of this history have been published during the past two decades, especially by the inventors themselves [45] [46] [47] [48] [49] [50] . However, it is instructive to consider some of the key insights that led to these drugs, especially in the context of the crystal structure that is now available and the structure-guided drug design of second-generation ACE inhibitors that can now be undertaken. A role for a metal in the catalytic mechanism had been suspected since the discovery of the enzyme 16 and was confirmed in the 1970s 51,52 , leading to the proposal that ACE was mechanistically similar to carboxypeptidase A (CPA) 53, 54 . We now know that ACE is unrelated to the CPA class of enzymes, but instead falls into the group of metallo-endopeptidases characterized by the HEXXH zinc-binding motif 55 (CPA possesses HXXE) . has been attributed to a direct vascular protective and anti-atherogenic effect of ACE inhibitors, because it has been observed even in normotensive individuals 69 . A major debate concerns the mechanism by which cardiovascular benefits are conferred by ACE inhibitors. It is generally accepted that Ang II not only has direct pressor effects and stimulates salt and water retention (via aldosterone release), but also stimulates myocyte proliferation and exerts pro-atherogenic effects via the induction of oxidative stress, endothelial dysfunction and vascular inflammation 71, 72 . However, there is also considerable evidence that some of the benefits of ACE inhibition derive from potentiation of BK signalling, which stimulates release of the vasodilator nitric oxide and of the fibrinolytic protein tissue plasminogen activator 72, 73 . Indeed, co-administration of the specific BK-receptor antagonist icatibant significantly attenuated the hypotensive effect of captopril in both normotensive and hypertensive subjects 74 . Further complicating this debate is the recent appreciation that the RAS is more complex than originally thought. There are multiple Ang peptides and at least three or four Ang receptors, some of which have opposing activities (FIG. 1) . For instance, the principal Ang II receptor, the AT 1 receptor, mediates the well-known effects of Ang II described above, but the AT 2 receptor, which has a more limited tissue distribution, mediates largely opposing effects. Similarly, Ang 1-7 , which is derived from both Ang I and Ang II by the action of various peptidases including ACE and ACE2, opposes the actions of Ang II 3, 37 . Moreover, as already discussed, ACE also acts on non-Ang peptides, including BK, substance P, luteinizing hormone-releasing hormone (LH-RH) and the haemoregulator AcSDKP; with the exception of BK, the importance of these peptides for the cardiovascular effects of ACE inhibitors is unknown. Some insights might derive from comparisons of the efficacy and side-effect profiles of ACE inhibitors and AT 1 -receptor blockers (ARBs). ACE inhibitors are generally well tolerated, but certain class-specific side effects have emerged, in particular cough and angioedema 70 . The incidence of cough has been estimated at 5-20% of patients and might result in the discontinuation of treatment 70, 75 . Angioedema affects 0.1-0.5% of patients and can be life-threatening 76 . Both cough and angioedema have been attributed to alterations in levels of non-Ang peptides, especially raised BK concentration. Recently, an association has been found between ACEinhibitor-related angioedema and low plasma levels of aminopeptidase P, an enzyme that is also involved in the metabolism of BK, indicating that these individuals are at risk for developing angioedema when treated with ACE inhibitors 76 . The potentiation of BK signalling by ACE inhibitors (FIG. 1) seems to result not only from reduced BK degradation but also from inhibition of desensitization of the B 2 BK receptor, possibly by inducing crosstalk between ACE and the B 2 receptor [77] [78] [79] . Therefore, both the benefits and side effects arising from increased BK signalling by ACE inhibitors seem to be mechanistically complex and have implications for the design of N-or C-selective inhibitors (see below). that was found to be the optimal C-terminal sequence among the venom peptides. The structure of the ACE-lisinopril complex confirms that lisinopril makes extensive contacts with the active site, including occupation of the S 1 , S 1 ′ and S 2 ′pockets, binding of a lysine by the C-terminal carboxylate, a hydrogen bond involving the (substrate) scissile amide nitrogen, and coordination of the active-site Zn 2+ by the carboxyalkyl carboxylate 5 (FIG. 4) . Lisinopril binds with twofold greater affinity than enalaprilat 63 , probably because the lysine side chain makes better contacts with the deep S 1 ′ pocket, including a weak hydrogen bond between the lysyl ε-amine and Glu162 (REF. 5 ). The design of captopril, enalaprilat and lisinopril was later extended by others, and a total of 17 ACE inhibitors have been approved for clinical use 4 . The later compounds are all variations on the original theme, with most of the differences residing in the functionalities that bind the active-site zinc and the S 2 ′ pocket 48 . The different types of zinc-coordinating groups are of interest, because these have also found application in the design of inhibitors for other metalloproteases, such as the matrix metallopeptidases. On the basis of work first described by Holmquist and Vallee 64 , phosphonates have proven useful 22, 23 , as have hydroxamates 65 , ketones 66 and silanediols 67 . Since their introduction in 1981, ACE inhibitors have been studied extensively (for recent reviews, see REFS 4, 68, 69) . ACE inhibitors are first-line therapy for hypertension, congestive heart failure, left ventricular systolic dysfunction and myocardial infarction, and are recommended for slowing the progression of diabetic and non-diabetic nephropathy 68, 70 . More recently, ACE inhibitors have also been shown to slow the progression of atherosclerotic vascular disease. This vascular benefit conditions 4, 75, 81, 82 . This difference might be due to the additional effect of ACE inhibitors on BK, although this comes at the cost of increased side effects 75, 81 . Interestingly, and contrary to earlier assumptions, angioedema has also been reported with ARB therapy 75, 83 , which might be related to unopposed activation of the AT 2 receptor leading to increased BK concentrations 73 . In light of the enormous importance of the RAS in cardiovascular pathophysiology, there is continued interest in novel compounds that target this system 4 . Eplerenone, the first selective aldosterone antagonist, The ARBs were introduced more recently but have also been studied extensively 4, 80 . As is the case for ACE inhibitors, ARBs have been shown to be effective in the treatment of hypertension and heart failure, in reducing cardiovascular morbidity and mortality, and in slowing the progression of nephropathy. Studies in myocardial infarction and heart failure have indicated a trend towards lower mortality in patients treated with ACE inhibitors versus ARBs, leading to the recommendation that ACE inhibitors remain the first-line agents in these C-domain site will allow some level of BK degradation to continue, catalysed by the N-domain. This could be sufficient to prevent the excessive BK accumulation that has been observed during attacks of angioedema 86 . Second, BK potentiation by B 2 receptor resensitization is maximal when both the N-and C-domains are inhibited 95 , indicating that a pure C-selective inhibitor will have a lower propensity for excessive BK stimulation. Third, the multiple Ang and non-Ang peptides known to be vasoactive are not hydrolysed equally by the two domains 3, 96 , making it likely that the ratio of vasopressor to vasodilator peptides will differ between C-selective and mixed inhibitors. So, a highly selective C-domain inhibitor has the potential for effective blood pressure control with reduced vasodilator-related side effects. In contrast to a C-selective inhibitor, an N-selective inhibitor might open up novel therapeutic areas. As discussed, the N-domain seems to play a minor role in blood pressure control in vivo. At least three physiologically important peptides are hydrolysed preferentially or exclusively by the N-domain: LH-RH, Ang 1-7 and AcSDKP 21, 44, 96 . The contribution of ACE to the metabolism of LH-RH and Ang 1-7 in vivo is unclear, but there is increasing evidence that ACE is the principal metabolizing enzyme for AcSDKP, a natural haemoregulatory hormone 97 . AcSDKP has antiproliferative and antifibrotic activities, and might have utility in protecting haematopoietic stem cells against chemotherapyinduced injury 97 and in limiting cardiac fibrosis 98 . Administration of ACE inhibitors results in a four to sixfold elevation of AcSDKP plasma levels 94, 97 . This might be the basis for the observed association between ACE inhibitors and anaemia, and the effective treatment of altitude polycythaemia by the ACE inhibitor enalaprilat 99 . Current-generation ACE inhibitors in clinical use are essentially mixed N-and C-domain inhibitors 48 . Although a modest degree of domain selectivity can be observed in some cases, this is not likely to be clinically significant. Nevertheless, these differences might be instructive and can guide future attempts to develop highly domain-selective inhibitors. Captopril has been noted to be modestly N-selective, depending on Clconcentration, whereas lisinopril and enalaprilat are more C-selective 24, 44 . More recently, keto-ACE, originally described in 1980 (REF. 66 ), was found to exhibit a 40-50-fold C-selectivity 96 ; one of the BPP peptides, BPPb, was shown to be 300-fold more C-selective 100 ; and the phosphinic tetrapeptide RXPA380 is 3,000-fold more C-selective 23 . By contrast, BPP-12b is 30-fold, and the phosphinic tetrapeptide RXP407 1,000-fold, more N-selective 22, 101 (TABLE 1) . Examination of these compounds reveals a number of features (TABLE 1 and FIG. 5a) . Captopril is the smallest inhibitor and can be viewed as an N-thioalkyl derivative of the dipeptide Ala-Pro, whereas enalaprilat, lisinopril and keto-ACE are tripeptide analogues of Phe-Ala-Pro, Phe-Lys-Pro and Phe-Gly-Pro, respectively. This might indicate that a bulky P 1 group -present in enalaprilat, lisinopril and keto-ACE, but absent in was approved for the treatment of hypertension in September 2002 and will probably also find use in the treatment of severe heart failure 7 . An orally active renin inhibitor, aliskiren, is now in clinical development for hypertension 84 . Of particular interest are the vasopeptidase inhibitors, which are dual ACE-neutral endopeptidase (NEP) inhibitors, of which omapatrilat is the most advanced in clinical development. NEP, also a zinc-metallopeptidase, is the principal enzyme responsible for the degradation of natriuretic peptides, which are vasodilatory and diuretic peptides that reduce volume loading and are therefore beneficial in both hypertension and heart failure 85 . As expected, omapatrilat was equivalent or superior to ACE inhibitors in clinical trials, but was found to be associated with a significantly higher incidence of angioedema, which has delayed its approval 86 . The higher incidence of angioedema is probably related to the fact that NEP also inactivates BK (FIG. 1) , and its inhibition might result in even higher plasma BK levels, which together with raised concentrations of natriuretic peptides (and potentially other vasodilatory peptides) might promote vasodilation-related adverse events 4, 86 . NEP might also be required for the removal of the neurotoxic amyloid β-peptide, and so chronic NEP inhibition could lead to neurodegeneration 87 . The clinical results with omapatrilat have had a sobering effect on the field 88 and have indicated that broad inhibition of vasoregulatory peptides should be approached with caution 89 . Indeed, overly vigorous inhibition of neuro-hormonal activation during heart failure, by simultaneous treatment with an ACE inhibitor, an ARB and a β-blocker was associated with excess mortality in the Valsartan Heart Failure Trial (Val-HeFT) 90 . So, efforts to develop triple inhibitors of ACE, NEP and endothelin-converting enzyme-1 (ECE-1) 91 might need to be re-evaluated. ECE-1, which is structurally and functionally related to NEP, is the principal activating enzyme for the potent vasopressor peptides endothelin-1 and -3 (REF. 92 ), and so triple inhibitors might show even greater efficacy than single ACE or dual ACE/NEP inhibitors. However, there is clearly an even greater potential for adverse events such as angioedema. In light of these developments, highly specific, singledomain inhibitors of ACE offer an attractive alternative. As we have attempted to show in this review, our understanding of the RAS (and related vasoregulatory systems) has come a long way since the introduction of the first ACE inhibitors. The N-and C-domain sites of ACE hydrolyse Ang I and BK at comparable rates in vitro, but in vivo it seems that the C-domain is primarily responsible for regulating blood pressure 93, 94 . This might indicate that a C-selective inhibitor would have a profile comparable to current mixed inhibitors, but this is not necessarily the case. First, whereas Ang I is hydrolysed predominantly by the C-domain in vivo 94 , BK is hydrolysed by both domains 23 and therefore selective inhibition of the be predicted that this sub-site is significantly different in the N-domain active site. Modelling of the S 2 sub-site in the N-domain has also revealed differences, as expected (see earlier), confirming its potential utility for conferring domain selectivity (FIG. 5c,d) . These considerations can form the starting point for the structureguided design of domain-selective inhibitors, which will be refined further once the N-domain structure becomes available. An important caveat in considering the design and pharmacological utility of domain-selective ACE inhibitors is the potential for conformational effects that have not yet been observed in the tACE crystal structure. For instance, it is unknown whether chloride binding and dissociation result in significant movement of the N-terminal 'lid' formed by helices α1, α2 and α3, and thereby restrict substrate access 5 . Even more importantly, the physical orientation of the N-and C-domains in somatic ACE is unknown, as is whether there is any significant degree of domain interaction or cooperativity. Inhibitor titrations in vitro 44 and studies with domainselective inhibitors in vivo 23 have provided indirect evidence for some form of domain interaction, which could have significant effects on the pharmacological profile of domain-selective inhibitors. The past decade has seen major advances in structurebased drug design approaches, including technologies such as mass spectrometry, X-ray crystallography and nuclear magnetic resonance. These are important tools in structural proteomics and to some extent have eliminated the scepticism about the feasibility and value of the structure-based approach. In particular, high-throughput structure-based drug design using protein crystallography has become a very attractive proposition for the pharmaceutical industry. Many examples exist today in which a combination of the three-dimensional structure of the target protein, computer-aided drug design (in silico or virtual screening), and a rational approach using highthroughput screening have produced important lead compounds that are now being evaluated in clinical trials (for recent reviews see REFS 1, 2, 103, 104) . In addition to mining the untapped riches of the human proteome, the application of modern structure-based drug design methods to existing drug targets will generate more selective compounds for known disease targets, such as ACE. We expect that next-generation, domain-selective ACE inhibitors will be a result of such endeavours. captopril -confers C-selectivity, and that a larger P 1 ′ side chain also promotes C-selectivity (FIG. 5b) , because lisinopril is more C-selective than enalaprilat. Interestingly, trandolaprilat, although a potent inhibitor for both domains, was tenfold more C-selective 24 (TABLE 1) . Trandolaprilat contains a C-terminal hexahydroindoline group, which also indicates that a bulky P 2 ′ group confers C-selectivity. This is confirmed by results from radioligand-binding studies, which indicated that both perindoprilat and quinaprilat, which contain hexahydroindoline and tetrahydroisoquinoline groups, respectively, in the P 2 ′ position, were 45-180-fold more C-selective 102 . Similarly, the highly C-selective tetrapeptide RXPA380 also contains a bulky methylindole group in the P 2 ′ position. These studies also indicated that lisinopril and 351A (a hydroxybenzamidine analogue of lisinopril) were 110-146-fold more C-selective, reinforcing the importance of a bulky P 1 ′ group 103 . On the other hand, structure-activity studies performed with a series of phosphinic tetrapeptides indicated that a phenylalanine in the P 1 position did not confer C-selectivity. Instead, the single most important determinant for N-selectivity was an amidated C-terminal carboxyl in the P 2 ′ position, followed by an acidic group in the P 2 position (FIG. 5d) 22 . The bradykinin potentiating peptides (BPPs) confirm the conclusion regarding the P 2 group: BPPa, BPPb, BPPc and BPP2 all end with the sequence Ile-Pro-Pro, yet only BPPc is unselective. BPPc has a proline in the P 2 position versus a lysine in the most C-selective peptide, BPPb 100 . Similarly, the C-selectivity of keto-ACE can probably be ascribed to a bulky P 2 benzyl group (FIG. 5c) ; the selectivity of RXPA380 is also consistent with the proposed importance of a phenyl P 2 group (FIG. 5a) . Taken together, these data indicate that C-selectivity is conferred by bulky P 1 ′ and P 2 ′ groups and a large, neutral or basic P 2 group, whereas N-selectivity is conferred by a blocked C-terminal carboxyl and an acidic P 2 group. The ACE C-domain crystal structure revealed that the S 1 ′ pocket was surprisingly deep, easily accommodating the lysyl group of lisinopril, with a hydrogen bond between Glu162 and the ε-amine (FIGS 4 and 5b) 5 . Additional modifications to the P 1 ′ group will potentially further enhance C-selectivity. Moreover, the C-terminal carboxylate of lisinopril was found to bind to Lys511, explaining the importance of a free C-terminal (P 2 ′) carboxyl for binding to the C-domain active site 22 . Binding to Lys511, instead of to an arginine (as originally predicted), might prompt investigation of functionalities other than carboxylates in this position. Since N-selectivity is conferred by a blocked P 2 ′ carboxylate, it can also High-throughput crystallography and lead discovery in drug design The genesis of high-throughput structure-based drug discovery The angiotensin-converting enzyme gene family: genomics and pharmacology Outstanding review of the RAS and of the landmark studies that established the clinical utility of ACE inhibitors Crystal structure of the human angiotensinconverting enzyme-lisinopril complex First report on the three-dimensional structure of human testis ACE and its interactions with the potent ACE inhibitor lisinopril at the molecular level The development of COX2 inhibitors Comparative pharmacology of H 1 antihistamines: clinical relevance Hypertension: Pathophysiology, Diagnosis, and Management Hypertension prevalence and blood pressure levels in 6 European countries, Canada, and the United States Hypertension: Pathophysiology, Diagnosis, and Management Studies on experimental hypertension. I. The production of persistent elevation of systolic blood pressure by means of renal ischemia Secretion hipertensora del rinon isquemaido A crystalline pressor substance (angiotonin) resulting from the interaction between renin and renin activator The purification of hypertensin I References 14-16 are a classic series of papers describing the isolation and characterization of Ang I Handbook of Proteolytic Enzymes First report of the molecular cloning of somatic ACE and the surprising finding of a two-domain structure The two homologous domains of human angiotensin I-converting enzyme are both catalytically active Differences in properties and enzymatic specificities between the two active sites of angiotensin 1-converting enzyme The hemoregulatory peptide N-acetyl-Ser-Asp-Lys-Pro is a natural and specific substrate of the N-terminal active site of human angiotensin-converting enzyme RXP 407, a phosphinic peptide, is a potent inhibitor of angiotensin I converting enzyme able to differentiate between its two active sites Roles of the two active sites of somatic angiotensin-converting enzyme in the cleavage of angiotensin I and bradykinin The two homologous domains of human angiotensin I-converting enzyme interact differently with competitive inhibitors Structure of the angiotensin I-converting enzyme gene. Two alternate promoters correspond to evolutionary steps of a duplicated gene Molecular cloning of human testicular angiotensinconverting enzyme: the testis isozyme is identical to the C-terminal half of endothelial angiotensin-converting enzyme Angiotensin-converting enzyme and male fertility An insertion/deletion polymorphism in the angiotensin 1-converting enzyme gene accounting for half of the variance of serum enzyme levels Angiotensin converting enzyme gene insertion/deletion polymorphism and cardiovascular disease: therapeutic implications Human performance: a role for the ACE genotype? Endurance and the ACE I/D polymorphism Proteolytic release of membranebound angiotensin-converting enzyme: role of the juxtamembrane stalk sequence Shedding of somatic angiotensinconverting enzyme (ACE) is inefficient compared with testis ACE despite cleavage at identical stalk sites Insect angiotensin-converting enzyme. A processing enzyme with broad substrate specificity and a role in reproduction The Acer gene of Drosophila codes for an angiotensin-converting enzyme homologue Functional conservation of the active sites of human and Drosophila angiotensin I-converting enzyme Just the beginning: novel functions for angiotensin-converting enzymes Deglycosylation, processing and crystallization of human testis angiotensin converting enzyme A major breakthrough using protein-engineering tools on testis ACE, making it amenable for structural study Drosophila melanogaster angiotensin Iconverting enzyme expressed in Pichia pastoris resembles the C domain of the mammalian homologue and does not require glycosylation for secretion and enzymic activity Crystal structure of Drosophila angiotensin I-converting enzyme bound to captopril and lisinopril Structure of neurolysin reveals a deep channel that limits substrate access Crystal structure of a novel carboxypeptidase from the hyperthermophilic archaeon Pyrococcus furiosus Effects of the N-terminal sequence of ACE on the properties of its C-domain First biophysical demonstration that somatic ACE indeed contains two zinc-binding active sites and that LH-RH Enzymes of the renin-angiotensin system and their inhibitors The design and properties of N-carboxyalkyldipeptide inhibitors of angiotensin-converting enzyme Design of angiotensin converting enzyme inhibitors Angiotensin-converting enzyme inhibitors Natural products and design: interrelated approaches in drug discovery The discovery of captopril Spectrophotometric assay and properties of the angiotensin-converting enzyme of rabbit lung Pulmonary angiotensin-converting enzyme. Structural and catalytic properties Design of potent competitive inhibitors of angiotensinconverting enzyme. Carboxyalkanoyl and mercaptoalkanoyl amino acids Design of specific inhibitors of angiotensin-converting enzyme: new class of orally active antihypertensive agents Landmark paper describing the brilliant series of insights leading to the first clinically useful ACE inhibitor, captopril Zinc coordination, function, and structure of zinc enzymes and other proteins Conversion of angiotensin I to angiotensin II Fate of angiotensin I in the circulation Conversion of angiotensin I to angiotensin II by cell-free extracts of dog lung Activity of various fractions of bradykinin potentiating factor against angiotensin I converting enzyme Angiotensin-converting enzyme inhibitors from the venom of Bothrops jararaca. Isolation, elucidation of structure, and synthesis Binding of the by-product analog benzylsuccinic acid by carboxypeptidase A Important extension of the Cushman-Ondetti model, laying the groundwork for a series of additional ACE inhibitors Inhibition of rabbit lung angiotensinconverting enzyme by N α (S)-1-carboxy-3-phenylpropyl]Llysyl-L-proline Metal-coordinating substrate analogs as inhibitors of metalloenzymes Dipeptidehydroxamates are good inhibitors of the angiotensin Iconverting enzyme Synthesis and biological activity of a ketomethylene analogue of a tripeptide inhibitor of angiotensin converting enzyme Siliconbased metalloprotease inhibitors: synthesis and evaluation of silanol and silanediol peptide analogues as inhibitors of angiotensin-converting enzyme The renin-angiotensin system: a review of trials with angiotensin-converting enzyme inhibitors and angiotensin receptor blocking agents Mechanisms of cardiovascular risk reductions with ramipril: insights from HOPE and HOPE substudies Using ACE inhibitors appropriately The AT 1 -type angiotensin receptor in oxidative stress and atherogenesis. Part I: oxidative stress and atherogenesis Inflammation in early atherogenesis: impact of ACE inhibition Contribution of bradykinin to the cardioprotective effects of ACE inhibitors Effect of bradykinin-receptor blockade on the response to angiotensin-converting-enzyme inhibitor in normotensive and hypertensive subjects Effects of losartan and captopril on mortality and morbidity in high-risk patients after acute myocardial infarction: the OPTIMAAL randomized trial Aminopeptidase P in individuals with a history of angio-oedema on ACE inhibitors Potentiation of the actions of bradykinin by angiotensin I-converting enzyme inhibitors. The role of expressed human bradykinin B 2 receptors and angiotensin-converting enzyme in CHO cells Enhancement of bradykinin and resensitization of its B 2 receptor Angiotensin-converting enzyme inhibitor ramiprilat interferes with the sequestration of the B 2 kinin receptor within the plasma membrane of native endothelial cells Angiotensin blockade for hypertension: a promise fulfilled Effect of losartan compared with captopril on mortality in patients with symptomatic heart failure: randomized trial -the Losartan Heart Failure Survival Study ELITE II Comparison of candesartan, enalapril, and their combination in congestive heart failure. Randomized evaluation of strategies for left ventricular dysfunction (RESOLVD) pilot study Can angiotensin receptor antagonists be used safely in patients with previous ACE inhibitor-induced angioedema? Angiotensin II suppression in humans by the orally active renin inhibitor aliskiren (SPP100): comparison with enalapril Vasopeptidase inhibition and angio-oedema β-amyloid catabolism: roles for neprilysin (NEP) and other metallopeptidases? Omapatrilat -the story of Overture and Octave Vasopeptidase inhibition: a double-edged sword? A randomized trial of the angiotensinreceptor blocker valsartan in chronic heart failure N-[2-(Indan-1-yl)-3-mercapto-propionyl] amino acids as highly potent inhibitors of the three vasopeptidases (NEP, ACE, ECE): in vitro and in vivo activities Hydrolysis of peptide hormones by endothelin-converting enzyme-1. A comparison with neprilysin The critical role of tissue angiotensinconverting enzyme as revealed by gene targeting in mice RXP 407, a selective inhibitor of the N-domain of angiotensin I-converting enzyme, blocks in vivo the degradation of hemoregulatory peptide acetyl-Ser-Asp-Lys-Pro with no effect on angiotensin I hydrolysis Bradykinin potentiation by angiotensin-(1-7) and ACE inhibitors correlates with ACE C-and N-domain blockade N-domain-specific substrate and C-domain inhibitors of angiotensin-converting enzyme. Angiotensin-(1-7) and keto-ACE Angiotensin I-converting enzyme and metabolism of the haematological peptide N-acetyl-seryl-aspartyl-lysyl-proline Effect of N-acetyl-seryl-aspartyl-lysylproline on DNA and collagen synthesis in rat cardiac fibroblasts Angiotensin-converting-enzyme inhibition therapy in altitude polycythaemia: a prospective randomised trial Selective inhibition of the C-domain of angiotensin I converting enzyme by bradykinin potentiating peptides The C-type natriuretic peptide precursor of snake brain contains highly specific inhibitors of the angiotensin-converting enzyme Structural constraints of inhibitors for binding at two active sites on somatic angiotensin converting enzyme Integration of virtual and high-throughput screening Structure-based screening and design in drug discovery Angiotensin-converting enzyme-2 (ACE-2): comparative modelling of the active site, specificity requirements and chloride dependence The ACE research in K.R.A.'s and E.D.S.'s laboratories is supported by the Wellcome Trust, UK. We would like to acknowledge the help of R. Natesh and S. Iyer in the preparation of this manuscript. The following terms in this article are linked online to: LocusLink: http://www.ncbi.nlm.nih.gov/LocusLink/ ACE | ACE2 | Acer | angiotensinogen | AT 1 receptor | AT 2 receptor | BK | ECE-1 | endothelin-1 | endothelin-3 | LH-RH | renin Access to this interactive links box is free online.