Discovery and development of ACE inhibitors

Last updated

The discovery of an orally inactive peptide from snake venom established the unimportant role of angiotensin converting enzyme (ACE) inhibitors in regulating blood pressure. This led to the development of captopril, the first ACE inhibitor. When the adverse effects of captopril became apparent new derivates were designed. Then after the discovery of two active sites of ACE: N-domain and C-domain, the development of domain-specific ACE inhibitors began. [1] [2]

Contents

Development of first generation ACE inhibitors

The development of the nonapeptide teprotide (Glu-Trp-Pro-Arg-Pro-Gln-Ile-Pro-Pro), which was originally isolated from the venom of the Brazilian pit viper Bothrops jararaca , greatly clarified the importance of ACE in hypertension. However, its lack of oral activity limited its therapeutic utility. [3] [4]

L-benzyl succinic acid (2(R)-benzyl-3-carboxypropionic acid) was described to be the most potent inhibitor of carboxypeptidase A in the early 1980s. The authors referred to it as a by-product analog and it was proposed to bind to the active site of carboxypeptidase A via succinyl carboxyl group and a carbonyl group. Their findings established that L-benzylsuccinic acid is bound at a single locus at the active site of carboxypeptidase A. The authors discussed but dismissed the suggestion that the carboxylate function might bind to the catalytically functional zinc ion present at the active site. Later however this was found to be the case. [3] [5] [6]

Drug design of captopril (sulfhydrils)

Captopril, a sulfhydril ACE inhibitor. Captopril structure.svg
Captopril, a sulfhydril ACE inhibitor.

Over 2000 compounds were tested randomly in a guinea pig ileum test and succinyl-L-proline was found to have the properties of a specific ACE inhibitor. It showed inhibitory effect of angiotensin I and bradykinin without having any effects on angiotensin II. Then researchers started to search for a model that would explain inhibition on the basis of specific interaction [ disambiguation needed ]s of compounds with the active site of ACE. [5] Previous studies with substrates and inhibitors of ACE suggested that it was a zinc-containing metalloprotein and a carboxypeptidase similar to pancreatic carboxypeptidase A. However ACE releases dipeptides rather than single amino acids from the C-terminus of the peptide substrates. And it was assumed that both their mechanism of action and their active site might be similar. A positively charged Arg145 at the active site was thought to bind with the negatively charged C-terminal carboxyl group of the peptide substrate. It was also proposed that ACE binds by hydrogen bonding to the terminal, non scissile, peptide bond of the substrate. [3]

But since ACE is a dipeptide carboxypeptidase, unlike carboxypeptidase A, the distance between the cationic carboxyl-binding site and the zinc atom should be greater, by approximately the length of one amino acid residue. Proline was chosen as the amino acid moiety because of its presence as the carboxy terminal amino acid residue in teprotide and other ACE inhibitors found in snake venoms. 11 other amino acids were tested but none of them were more inhibitory. So it was proposed that succinyl amino acid derivative should be an ACE inhibitor and succinyl-L-proline was found to be such an inhibitor. [3] [5] [7]

It was also known that the nature of penultimate amino acid residue of a peptide substrate for ACE influences binding to the enzyme. The acyl group of the carboxyalkanoyl amino acid binds the zinc ion of the enzyme and occupies the same position at the active site of ACE as the penultimate. Therefore, the substituent of the acyl group might also influence binding to the enzyme. A 2-methyl substituent with D configuration was found to enhance the inhibitory potency by about 15 fold of succinyl-L-proline. Then the search for a better zinc-binding group started. Replacement of the succinyl carboxyl group by nitrogen-containing functionalities (amine, amide or guanidine) did not enhance inhibitory activity. However a potency breakthrough was achieved by the replacement of the carboxyl group with a sulfhydryl function (SH), a group with greater affinity for the enzyme bound zinc ion. This yielded a potent inhibitor that was 1000 times more potent than succinyl-L-proline. [3] [7] The optimal acyl chain length for mercapto alkanoyl derivates of proline was found to be 3-mercaptopropanoyl-L-proline, 5 times greater than that of 2-mercaptoalkanoyl derivates and 50 times greater than that of 4-mercaptoalkanoyl derivates. So the D-3-mercapto-2-methylpropanoyl-L-proline or Captopril was the most potent inhibitor. Later, the researchers compared a few mercaptoacyl amino acid inhibitors and concluded that the binding of the inhibitor to the enzyme involved a hydrogen bond between a donor site on the enzyme and the oxygen of the amide carbonyl, much like predicted for the substrates. [3] [8]

Drug design of other first generation ACE inhibitors

Enalapril. Enalapril structure.svg
Enalapril.
Lisiniopril. Lisinopril structure.svg
Lisiniopril.

The most common adverse effects of Captopril, skin rash and loss of taste, are the same as caused by mercapto-containing penicillamine. Therefore, a group of researchers aimed at finding potent, selective ACE inhibitors that would not contain a mercapto (SH) function and would have a weaker chelating function. They returned to work with carboxyl compounds and started working with substituted N-carboxymethyl-dipeptides as a general structure (R-CHCOOH-A1-A2). According to previous research they assumed that cyclic imino acids would result in good potency if substituted on the carboxyl terminus of the dipeptide. Therefore, substituting A2 with proline gave good results. They also noted that according to the enzyme's specificity imino acids in the position next to the carboxyl terminus would not give a potent compound. By substituting R and A1 groups with hydrophobic and basic residues would give a potent compound. By substituting –NH in the general structure resulted in loss of potency which is consistent to the enzyme's need for a –NH in corresponding position on the substrates. The results were 2 active inhibitors: Enalaprilat and Lisinopril. These compounds both have phenylalanine in R position which occupies the S1 groove in the enzyme. The result was thus these two new, potent tripeptide analogues with zinc-coordinating carboxyl group: Enalaprilat and Lisinopril. [1] [9]

Discovery of 2 active sites: C-domain and N-domain

Most of the ACE inhibitors on the market today are non-selective towards the two active sites of ACE because their binding to the enzyme is based mostly on the strong interaction [ disambiguation needed ] between the zinc atom in the enzyme and the strong chelating group on the inhibitor. The resolution of the 3D structure of germinal ACE, which has only one active site that corresponds with C-domain of the somatic ACE, offers a structural framework for structure-based design approach. Although N- and C-domain have comparable rates in vitro of ACE hydrolyzing, it seems like that in vivo the C-domain is mainly responsible for regulating blood pressure. This indicates that C-domain selective inhibitors could have similar profile to that of a current non-selective inhibitors. Angiotensin I is mainly hydrolyzed by the C-domain in vivo but bradykinin is hydrolyzed by both active sites. Thus, by developing a C-domain selective inhibitor would permit some degradation of bradykinin by the N-domain and this degradation could be enough to prevent accumulation of excess bradykinin which has been observed during attacks of angioedema. C-domain selective inhibition could possibly result in specialized control of blood pressure with less vasodilator-related adverse effects. N-domain selective inhibitors on the other hand give the possibility of opening up novel therapeutic areas. Apparently, the N-domain does not have a big role in controlling blood pressure but it seems to be the principal metabolizing enzyme for AcSDKP, a natural haemoregulatory hormone. [1] [10] [11]

Drug design of Keto-ACE and its ketomethylene derivatives

It was found that other carbonyl-containing groups such as ketones could substitute for the amide bond that links Phe and Gly in ACE inhibitors. Keto-ACE, first described in 1980, has emerged as a potential lead compound for C-domain specific ACE inhibitors. Keto-ACE, a tripeptide analogue of Phe-Gly-Pro, contains a bulky P1 and P2 benzyl ring and was shown to inhibit the hydrolysis of angiotensin I and bradykinin via the C-domain. The synthesis of keto-ACE analogues with Trp or Phe at the P2’ position led to a marked increase in C-domain selectivity, but the introduction of an aliphatic P2 group conferred N-domain selectivity. Inhibitory potency may further be enhanced by the incorporation of hydrophobic substituent, such as phenyl group at the P1’ position. P1’ substituents with S-stereochemistry have also been shown to possess greater inhibitory potency than their R-counterparts. [2] [8] [12] [13]

Keto-ACE was used as the basis for the design of ketomethylene derivates. Its analogues contain a ketomethylene isostere replacement at the scissile bond that is believed to mimic the tetrahedron transition state of the proteolytic reaction at the active site. The focus was on a simple tripeptide Phe-Ala-Pro, which in earlier enzyme assays has shown inhibition activity. Replacement of alanine with glycin gave a tripeptide with 1/14th of the inhibition activity of Phe-Ala-Pro. The benzoylated derivative of Phe-Gly-Pro, Bz-Phe-Gly-Pro, was twice as active. To reduce the peptidic nature of ketomethylene inhibitors the P1’ and P2’ substituent may be cyclized to form a lactam, where there is a correlation between the inhibitory potency and the ring size. In 2001 it was postulated that a substitution α to nitrogen and making of 3-methyl-substituted analog of A58365A, a pyridone acid isolated from the fermentation broth of the bacterium Streptomyces chromofuscus with ACE inhibitory activity, might influence the level of biological activity by steric or hydrophobic effect, and/or by preventing reactions at C3. It was also noticed during the synthetic work on A58365A that potential precursors were sensitive to oxidation of the five-membered ring and so the 3-methyl analogue might be more stable in this respect. [2] [14] [15]

Drug design of silanediol

The fact that carbon and silicone have similar, but also dissimilar, characteristics triggered the interest in substituting carbon with silanediol as a central, zinc chelating group. Silicone forms a dialkylsilanediol compound that is sufficiently hindered so the formation of a siloxane polymer does not occur. Silanediols are more stable than carbon diols so they are expected to have longer half-life. Silanediols are also neutral at physiological pH (do not ionize). Four stereoisomers of Phe-Ala silanediol were compared to ketone-based inhibitors and the silanediol were found to be fourfold less potent than the ketone analogue. This is because silanediols are weaker zinc chelators compared with ketones. Replacement of the silanediol, with a methylsilano group gave little enzyme inhibition. This confirms that the silanediol group interacts with ACE as a transition state analogue and the interaction is in a manner similar to that of ketone. [16] [17] If the benzyl group of silanediol is replaced by an i-butyl group it gives a weaker ACE inhibitor. Introduction of a hydrophobic methyl phenyl gives a little more potency than an analogue with a tert-butyl-group at P1. That suggests that methyl phenyl gives a better S1 recognition than a tert-butyl group. [2]

Phosphinic peptides

Phosphinic peptides are pseudo-peptides where a phosphinic acid bond (PO2-CH-) has replaced a peptide bond in the peptide analogue sequence. To some extent the chemical structure of phosphinic peptides is similar to that of intermediates which are produced in hydrolysis of peptides by proteolytic enzymes. The hypothesis has been made that these pseudo-peptides mimic the structure of the enzyme substrates in their transition state and crystallography of zinc proteases in complex with phosphinic peptides supports that hypothesis. [10]

Drug design of RXP 407

RXP 407 is the first N-domain selective phosphinic peptide and was discovered by screening phosphinic peptides libraries. Before the discovery of RXP 407 it had long been claimed that the free C-terminal carboxylate group in P2’ position was essential to the potency of ACE inhibitor so it can be reasoned that this has postponed the discovery of N-domain selective ACE inhibitors. When RXP 407 was discovered researchers looked into phosphinic peptides with 3 different general formula, each containing 2 unidentified amino acids, only 1 of these general formula showed potent inhibition (Ac-Yaa-Pheψ(PO2-CH2)Ala-Yaa’-NH2). Peptide mixtures were made, substituting Yaa and Yaa’ with different amino acids, trying to establish if there would be a potent inhibitor that could inhibit either the N-domain or the C-domain of the enzyme. The result was that the compound Ac-Asp (L)-Pheψ(PO2-CH2)(L)Ala-Ala-NH2 actively inhibited the N-domain and was given the name RXP 407. Structure-function relationship showed that the C-terminus carboxamide group played a crucial role in the selectivity for the N-domain of ACE. Additionally, the N-acetyl group and the aspartic side chain in the P2 position aides in the N-domain selectivity of the inhibitor. These features make the inhibitor inaccessible to the C-domain but give good potency for the N-domain, this leads to a difference in inhibitory potency of the active sites of three orders of magnitude. These results also indicate that the N-domain possess a broader selectivity than the C-domain. Another difference between the older ACE inhibitors and RXP 407 is the molecular size of the compound. The older ACE inhibitors had mostly been interacting with S1’, S2’ and S1 subsites but RXP 407 interacts in addition with the S2 subsite. This also is important for the selectivity of the inhibitor since the aspartic side chain and N-acetyl group are located in the P2 position. [18]

Drug design of RXPA 380

RXPA380 was the first inhibitor that was highly selective of the C-domain of ACE, it has the formula Phe-Phe-Pro-Trp. [1] The development of this compound was built on researches that showed that some bradykinin-potentiating peptides showed selectivity for the C-domain and all had several prolines in their structure. These observations lead the researchers to synthesize phosphinic peptides containing a proline residue in the P1’ position and evaluating these compounds led to the discovery of RXPA380. [19] To study the roles of the residues on RXPA380 the researchers made 7 analogues of RXPA380. All of the compounds made were obtained as a mixture of either 2 or 4 diastereoisomers but all of them were easily resolved and only one of them was potent. This is consistent with the initial modeling studies of RXPA380 which showed that only one diastereomer could accommodate in the active site of germinal ACE. Analogues where pseudo-proline or tryptophan residues had been substituted showed less selectivity than RXPA380. This is probably because these two analogues have more potency toward the N-domain than RXPA380 does. Substituting both of these residues gives great potency but none selectivity. This shows that pseudo-proline and tryptophan residues accommodate well in the C-domain but not in the N-domain. 2 more analogues with both pseudo-proline and tryptophan but missing the pseudo-phenylalanine residue in P1 position showed low potency for N-domain, similar to RXPA380. This supports the significant role of these two residues in the selectivity for C-domain. These two analogues also have less potency for the C-domain which shows that the C-domain prefers pseudo-phenylalanine group in P1 position. Modeling of RXPA380-ACE complex showed that the pseudo-proline residue of the inhibitor was surrounded by amino acids similar to that of the N-domain thus interactions with S2’ domain might not be responsible for the selectivity of RXPA380. 7 of 12 amino acids surrounding tryptophan are the same in C- and N-domain, the biggest difference is that 2 bulky and hydrophobic amino acids in the C-domain have been replaced with 2 smaller and polar amino acids in the N-domain. This indicates that low potency of RXPA380 for N-domain is not because the S2’ cavity does not accommodate the tryptophan side chain but rather that important interactions are missing between the tryptophan side chain and the amino acids of the C-domain. Based on the proximity between the tryptophan side chain and Asp1029 there is also a possible hydrogen bond between the carboxylate of Asp1029 and the NH indole ring in the C-domain but this interaction is much weaker in the N-domain. [1]

Related Research Articles

<span class="mw-page-title-main">ACE inhibitor</span> Class of medications used primarily to treat high blood pressure

Angiotensin-converting-enzyme inhibitors are a class of medication used primarily for the treatment of high blood pressure and heart failure. This class of medicine works by causing relaxation of blood vessels as well as a decrease in blood volume, which leads to lower blood pressure and decreased oxygen demand from the heart.

<span class="mw-page-title-main">Bradykinin</span> Chemical compound

Bradykinin (BK) (Greek brady-, slow; -kinin, kīn(eîn) to move) is a peptide that promotes inflammation. It causes arterioles to dilate (enlarge) via the release of prostacyclin, nitric oxide, and endothelium-derived hyperpolarizing factor and makes veins constrict, via prostaglandin F2, thereby leading to leakage into capillary beds, due to the increased pressure in the capillaries. Bradykinin is a physiologically and pharmacologically active peptide of the kinin group of proteins, consisting of nine amino acids.

<span class="mw-page-title-main">Captopril</span> Chemical compound

Captopril, sold under the brand name Capoten among others, is an angiotensin-converting enzyme (ACE) inhibitor used for the treatment of hypertension and some types of congestive heart failure. Captopril was the first oral ACE inhibitor found for the treatment of hypertension. It does not cause fatigue as associated with beta-blockers. Due to the adverse drug event of causing hyperkalemia, as seen with most ACE Inhibitors, the medication is usually paired with a diuretic.

<span class="mw-page-title-main">Angiotensin-converting enzyme</span> Mammalian protein found in Homo sapiens

Angiotensin-converting enzyme, or ACE, is a central component of the renin–angiotensin system (RAS), which controls blood pressure by regulating the volume of fluids in the body. It converts the hormone angiotensin I to the active vasoconstrictor angiotensin II. Therefore, ACE indirectly increases blood pressure by causing blood vessels to constrict. ACE inhibitors are widely used as pharmaceutical drugs for treatment of cardiovascular diseases.

The kinin–kallikrein system or simply kinin system is a poorly understood hormonal system with limited available research. It consists of blood proteins that play a role in inflammation, blood pressure control, coagulation and pain. Its important mediators bradykinin and kallidin are vasodilators and act on many cell types. Clinical symptoms include marked weakness, tachycardia, fever, leukocytosis and acceleration of ESR.

<span class="mw-page-title-main">DD-transpeptidase</span>

DD-transpeptidase is a bacterial enzyme that catalyzes the transfer of the R-L-αα-D-alanyl moiety of R-L-αα-D-alanyl-D-alanine carbonyl donors to the γ-OH of their active-site serine and from this to a final acceptor. It is involved in bacterial cell wall biosynthesis, namely, the transpeptidation that crosslinks the peptide side chains of peptidoglycan strands.

<span class="mw-page-title-main">Carboxypeptidase</span>

A carboxypeptidase is a protease enzyme that hydrolyzes (cleaves) a peptide bond at the carboxy-terminal (C-terminal) end of a protein or peptide. This is in contrast to an aminopeptidases, which cleave peptide bonds at the N-terminus of proteins. Humans, animals, bacteria and plants contain several types of carboxypeptidases that have diverse functions ranging from catabolism to protein maturation. At least two mechanisms have been discussed.

Kininogens are precursor proteins for kinins, biologically active polypeptides involved in blood coagulation, vasodilation, smooth muscle contraction, inflammatory regulation, and the regulation of the cardiovascular and renal systems.

<span class="mw-page-title-main">Phosphodiesterase 2</span> Class of enzymes

The PDE2 enzyme is one of 21 different phosphodiesterases (PDE) found in mammals. These different PDEs can be subdivided to 11 families. The different PDEs of the same family are functionally related despite the fact that their amino acid sequences show considerable divergence. The PDEs have different substrate specificities. Some are cAMP selective hydrolases, others are cGMP selective hydrolases and the rest can hydrolyse both cAMP and cGMP.

<span class="mw-page-title-main">Met-enkephalin</span> Chemical compound

Met-enkephalin, also known as metenkefalin (INN), sometimes referred to as opioid growth factor (OGF), is a naturally occurring, endogenous opioid peptide that has opioid effects of a relatively short duration. It is one of the two forms of enkephalin, the other being leu-enkephalin. The enkephalins are considered to be the primary endogenous ligands of the δ-opioid receptor, due to their high potency and selectivity for the site over the other endogenous opioids.

<span class="mw-page-title-main">Antamanide</span> Chemical compound

Antamanide is a cyclic decapeptide isolated from a fungus, the death cap: Amanita phalloides. It is being studied as a potential anti-toxin against the effects of phalloidin and for its potential for treating edema. It contains 1 valine residue, 4 proline residues, 1 alanine residue, and 4 phenylalanine residues with a structure of c(Val-Pro-Pro-Ala-Phe-Phe-Pro-Pro-Phe-Phe). It was isolated by determining the source of the anti-phalloidin activity from a lipophillic extraction from the organism. It has been shown that antamanide can react to form alkali metal ion complexes. These include complexes with sodium and calcium ions. When these complexes are formed, the cyclopeptide structure undergoes a conformational change.

<span class="mw-page-title-main">Carboxypeptidase A</span>

Carboxypeptidase A usually refers to the pancreatic exopeptidase that hydrolyzes peptide bonds of C-terminal residues with aromatic or aliphatic side-chains. Most scientists in the field now refer to this enzyme as CPA1, and to a related pancreatic carboxypeptidase as CPA2.

<span class="mw-page-title-main">Renin inhibitor</span> Compound inhibiting the activity of renin

Renin inhibitors are pharmaceutical drugs inhibiting the activity of renin that is responsible for hydrolyzing angiotensinogen to angiotensin I, which in turn reduces the formation of angiotensin II that facilitates blood pressure.

The angiotensin receptor blockers (ARBs), also called angiotensin (AT1) receptor antagonists or sartans, are a group of antihypertensive drugs that act by blocking the effects of the hormone angiotensin II in the body, thereby lowering blood pressure. Their structure is similar to Ang II and they bind to Ang II receptors as inhibitors, e.g., [T24 from Rhys Healthcare].

Dipeptidyl peptidase-4 inhibitors are enzyme inhibitors that inhibit the enzyme dipeptidyl peptidase-4 (DPP-4). They are used in the treatment of type 2 diabetes mellitus. Inhibition of the DPP-4 enzyme prolongs and enhances the activity of incretins that play an important role in insulin secretion and blood glucose control regulation. Type 2 diabetes mellitus is a chronic metabolic disease that results from inability of the β-cells in the pancreas to secrete sufficient amounts of insulin to meet the body's needs. Insulin resistance and increased hepatic glucose production can also play a role by increasing the body's demand for insulin. Current treatments, other than insulin supplementation, are sometimes not sufficient to achieve control and may cause undesirable side effects, such as weight gain and hypoglycemia. In recent years, new drugs have been developed, based on continuing research into the mechanism of insulin production and regulation of the metabolism of sugar in the body. The enzyme DPP-4 has been found to play a significant role.

Many major physiological processes depend on regulation of proteolytic enzyme activity and there can be dramatic consequences when equilibrium between an enzyme and its substrates is disturbed. In this prospective, the discovery of small-molecule ligands, like protease inhibitors, that can modulate catalytic activities has an enormous therapeutic effect. Hence, inhibition of the HIV protease is one of the most important approaches for the therapeutic intervention in HIV infection and their development is regarded as major success of structure-based drug design. They are highly effective against HIV and have, since the 1990s, been a key component of anti-retroviral therapies for HIV/AIDS.

<span class="mw-page-title-main">Peptidyl-dipeptidase Dcp</span> Class of enzymes

Peptidyl-dipeptidase Dcp (EC 3.4.15.5, dipeptidyl carboxypeptidase (Dcp), dipeptidyl carboxypeptidase) is a metalloenzyme found in the cytoplasm of bacterium E. Coli responsible for the C-terminal cleavage of a variety of dipeptides and unprotected larger peptide chains. The enzyme does not hydrolyze bonds in which P1' is Proline, or both P1 and P1' are Glycine. Dcp consists of 680 amino acid residues that form into a single active monomer which aids in the intracellular degradation of peptides. Dcp coordinates to divalent zinc which sits in the pocket of the active site and is composed of four subsites: S1’, S1, S2, and S3, each subsite attracts certain amino acids at a specific position on the substrate enhancing the selectivity of the enzyme. The four subsites detect and bind different amino acid types on the substrate peptide in the P1 and P2 positions. Some metallic divalent cations such as Ni+2, Cu+2, and Zn+2 inhibit the function of the enzyme around 90%, whereas other cations such as Mn+2, Ca+2, Mg+2, and Co+2 have slight catalyzing properties, and increase the function by around 20%. Basic amino acids such as Arginine bind preferably at the S1 site, the S2 site sits deeper in the enzyme therefore is restricted to bind hydrophobic amino acids with phenylalanine in the P2 position. Dcp is divided into two subdomains (I, and II), which are the two sides of the clam shell-like structure and has a deep inner cavity where a pair of histidine residues bind to the catalytic zinc ion in the active site. Peptidyl-Dipeptidase Dcp is classified like Angiotensin-I converting enzyme (ACE) which is also a carboxypeptidase involved in blood pressure regulation, but due to structural differences and peptidase activity between these two enzymes they had to be examined separately. ACE has endopeptidase activity, whereas Dcp strictly has exopeptidase activity based on its cytoplasmic location and therefore their mechanisms of action are differentiated. Another difference between these enzymes is that the activity of Peptidyl-Dipeptidase Dcp is not enhanced in the presence of chloride anions, whereas chloride enhances ACE activity.

Lysine carboxypeptidase is an enzyme. This enzyme catalyses the following chemical reaction:

<span class="mw-page-title-main">Scytalidopepsin B</span>

Scytalidocarboxyl peptidase B, also known as Scytalidoglutamic peptidase and Scytalidopepsin B is a proteolytic enzyme. It was previously thought to be an aspartic protease, but determination of its molecular structure showed it to belong a novel group of proteases, glutamic protease.

Four drugs from the class of direct Xa inhibitors are marketed worldwide. Rivaroxaban (Xarelto) was the first approved FXa inhibitor to become commercially available in Europe and Canada in 2008. The second one was apixaban (Eliquis), approved in Europe in 2011 and in the United States in 2012. The third one edoxaban was approved in Japan in 2011 and in Europe and the US in 2015. Betrixaban (Bevyxxa) was approved in the US in 2017.

References

  1. 1 2 3 4 5 Acharya, K.R.; Sturrock, E.D.; Riodan, J.K.; Ehlers, M.R. (2003), "ACE revisited: A New Target for Structure-Based Dug Design.", Nature Reviews Drug Discovery, 2 (11): 891–902, doi:10.1038/nrd1227, PMC   7097707 , PMID   14668810
  2. 1 2 3 4 Redelinghuys, P.; Nchinda, A.T.; Sturrock, E.D. (2005), "Development of Domain-Selective Enzyme Inhibitors.", Annals of the New York Academy of Sciences, 1056: 160–175, doi:10.1196/annals.1352.035, PMID   16387685, S2CID   25407204
  3. 1 2 3 4 5 6 Cushman, D.W.; Cheung, H.S.; Sabo, E.F.; Ondetti, M.A. (1977), "Design of Potent Competitive Inhibitors of Angiotensin-Converting Enzye. Carboxyalkanoyl and Mercaptoalkanoyl Amino Acid.", Biochemistry, 16 (25): 5484–5491, doi:10.1021/bi00644a014, PMID   200262
  4. Crantz, F.R.; Swartz, S.L.; Hollenberg, N.K.; Moore, T.J.; Dluhy, R.G.; Williams, G.H. (1980), "Differences in response to the peptidyldipeptide hydrolase inhibitors SQ 20,881 and SQ 14,225 in normal-renin essential hypertension.", Hypertension, 2 (5): 604–609, doi: 10.1161/01.hyp.2.5.604 , PMID   6158478
  5. 1 2 3 Cushman, D.W.; Ondetti, M.A. (1991), "History of the design of captopril and related inhibitors of angiotensin converting enzyme.", Hypertension, 17 (4): 589–592, doi: 10.1161/01.hyp.17.4.589 , PMID   2013486
  6. Byers, L.D.; Wolfenden, R. (1973), "Binding of the By-Product Analog Benzylsuccinic Acid by Carboxypeptidase A.", Biochemistry, 12 (11): 2070–2078, doi:10.1021/bi00735a008, PMID   4735879
  7. 1 2 Ondetti, M.A.; Rubin, B.; Cushman, D.W. (1977), "Design of Specific Inhibitors of Angiotensin-Converting Enzyme: New Class of Orally Active Antihypertensive Agents.", Science, 196 (4288): 441–444, Bibcode:1977Sci...196..441O, doi:10.1126/science.191908, PMID   191908
  8. 1 2 Condon, M.E.; et al. (1982), "Angiotensin-Converting Enzyme Inhibitors: Importance of the Amide Carbonyl of Mercaptoacyl Amino Acids for Hydrogen Bonding to the Enzyme", Journal of Medicinal Chemistry, 25 (3): 250–258, doi:10.1021/jm00345a011, PMID   6279843
  9. Patchett, A.A.; et al. (1980), "A new class of angiotensin-converting enzyme inhibitors", Nature, 288 (5788): 280–283, Bibcode:1980Natur.288..280P, doi:10.1038/288280a0, PMID   6253826, S2CID   4246377
  10. 1 2 Dive, V.; et al. (2004), "Review: Phosphinic peptides as zinc metalloproteinase inhibitors", Cellular and Molecular Life Sciences, 61 (16): 2010–2019, doi:10.1007/s00018-004-4050-y, PMID   15316651, S2CID   30797568
  11. Gerogiadis, D.; Guniasse, P.; Cotton, J.; Yiotakis, A.; Dive, V. (2004), "Structural Determinants of RXPA380, a Potent and Highly Selective Inhibitor of the Angiotensin-Converting Enzyme C-domain", Biochemistry, 43 (25): 8048–8054, doi:10.1021/bi049504q, PMID   15209500
  12. Nchinda, A.T.; Chibale, K.; Redelinghuys, P.; Stirrock, E.D. (2006), "Synthesis of novel keto-ACE analogues as domain-selective angiotensin I-converting enzyme inhibitors", Bioorganic & Medicinal Chemistry Letters, 16 (17): 4612–4615, doi:10.1016/j.bmcl.2006.06.003, PMID   16784850
  13. Redelinghuys, P.; Nchinda, A.T.; Chibale, K.; Sturrock, E.D. (2006), "Novel ketomethylene inhibitors of angiotensin I-converting enzyme (ACE): inhibition and molecular modelling", Biological Chemistry, 387 (4): 461–466, doi:10.1515/BC.2006.061, PMID   16606345, S2CID   30710745
  14. Almquist, R.G.; Chao, W.R.; Ellis, M.E.; Johnson, H.L. (1980), "Synthesis and Biological Activity of a Ketomethylene Analogue of a Tripeptide Inhibitor of Angiotensin-Converting Enzyme", Journal of Medicinal Chemistry, 23 (12): 1392–1398, doi:10.1021/jm00186a020, PMID   6256550
  15. Clive, D.L.J.; Yang, H.; Lewanczuk, E.Z. (2001), "Synthesis and in vitro activity of a non-epimerizable analog of the angiotensin-converting enzyme inhibitor A58365A", Comptes Rendus de l'Académie des Sciences - Series IIC - Chemistry, 4 (6): 505–512, doi:10.1016/s1387-1609(01)01263-4
  16. Kim, J.; Sieburth, S.M. (2004), "Silanediol peptidomimetics. Evaluation of four diastereomeric ACE inhibitors", Bioorganic & Medicinal Chemistry Letters, 14 (11): 2853–2856, doi:10.1016/j.bmcl.2004.03.042, PMID   15125946
  17. Kim, J.; Hewitt, G.; Carroll, P.; Sieburth, S.M. (2005), "Silanediol Inhibitors of Angiotensin-Converting Enzyme. Synthesis and Evaluation of Four Diastereomers of Phe[Si]Ala Dipeptide Analogues", Journal of Organic Chemistry, 70 (15): 5781–5785, doi:10.1021/jo048121v, PMID   16018669
  18. Dive, V.; et al. (1999), "RXP 407, a phosphinic peptide, is a potent inhibitor of angiotensin I converting anzyme able to differentiate between its two active sites", PNAS, 96 (8): 4330–4335, Bibcode:1999PNAS...96.4330D, doi: 10.1073/pnas.96.8.4330 , PMC   16332 , PMID   10200262
  19. Georgiadis, D.; Beau, F.; Czarny, B; Cottin, J; Yiotakis, A; Dive, V (2003), "Roles of the Two Active Sites of Somatic Angiotensin-Converting Enzyme in the Cleavage of Angiotensin I and Bradykinin: Insights From Selective Inhibitors", Circulation Research, 93 (2): 148–154, doi: 10.1161/01.RES.0000081593.33848.FC , PMID   12805239