Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

ORIGINAL ARTICLE

A Review of the Structural and Functional Features of


Olmesartan Medoxomil, An Angiotensin Receptor Blocker
David E. Mire, PhD,* Tonous N. Silfani, PhD,† and Michael K. Pugsley, PhD‡

Although precise mechanisms have not yet been elucidated to


Abstract: The angiotensin II (A-II) type 1 (AT1) receptor–mediated explain all observed clinical benefits, there can be little doubt
effects of A-II play a key role in the pathophysiology of hypertension. that the ability to significantly reduce systolic and diastolic
Effective inhibition of A-II is provided by the latest class of anti- blood pressure provides much of the benefit seen with ARB
hypertensive medications, the AT1 receptor blockers (ARBs). These therapy.5 Although this is not the only drug class capable of
orally available agents were developed around a common imidazole- producing clinically significant reductions in blood pressure,
based structural core. The most recent member of this drug class the ARBs are unique in their ability to provide such benefit
to be approved by the Food and Drug Administration, olmesartan with a limited side effect profile.6 Interestingly, although blood
medoxomil, contains unique features that may explain its clinical ef- pressure lowering occurs by the same mechanism, there are
ficacy. Key structural elements of olmesartan medoxomil include a differences in the antihypertensive efficacy of the various
hydroxyalkyl substituent at the imidazole 4-position and a hydrolyz- agents within the ARB class.
able ester group at the imidazole 5-position. Inter- and intramolecular The renin–angiotensin–aldosterone system (RAAS) plays
hydrogen bonding involving these groups may contribute to the po- a central role in blood pressure regulation and the pathogenesis
tentiation of antagonist activity. After oral administration, olmesartan of hypertension. The RAAS consists of an enzymatic cascade.
medoxomil is deesterified in the intestinal tract to produce the active Angiotensinogen, produced by the liver, is converted to angio-
metabolite olmesartan, which undergoes no additional metabolic change. tensin I (A-I) by renin, which is secreted by juxtaglomerular
The marked antihypertensive efficacy of olmesartan medoxomil may cells in the kidneys. Angiotensin-converting enzyme (ACE)
result from a unique pharmacological interaction of the drug with the converts A-I to A-II, a powerful peptide hormone responsible
AT1 receptor, resulting in a potent, long-lasting, dose-dependent for a myriad of cellular events (Fig. 1).7,8 ACE inhibitors block
blockade of A-II. This review article characterizes the structural the formation of A-II; however, they also prevent the break-
features of olmesartan that may be responsible for its clinical efficacy. down of bradykinin. The resulting accumulation of bradykinin
Inferential pharmacological studies compare and contrast the effects as well as substance P and related active peptides may con-
of olmesartan to those of other ARBs in comparable preclinical tribute to the development of cough and angioedema, which
animal models. are often associated with ACE inhibitor therapy.9,10 Moreover,
Key Words: angiotensin receptor blocker, antihypertensive, chem- ACE inhibition may not completely block the RAAS. In vitro
istry, methods, olmesartan medoxomil, structural features experiments have shown that angiotensinogen can be converted
to A-II by nonrenin enzymes such as tissue plasminogen acti-
(J Cardiovasc Pharmacol TM 2005;46:585–593) vator, cathepsin G, elastase, and chymostatin, and A-I can be
converted to A-II by non-ACE enzymes such as chymostatin-
sensitive A-II–generating enzyme, chymase, and cathepsin G.9
The primary effector hormone in this system, the octa-
peptide A-II, exerts numerous effects on many different tis-
sues, including vasoconstriction of vascular smooth muscle,
A ngiotensin II (A-II) type 1 (AT1) receptor antagonists
(ARBs) have become a key component in the antihyperten-
sive armamentarium. Recently reported clinical studies de-
aldosterone secretion from adrenal glomerulosa cells, and tro-
phic changes in the heart and blood vessels via binding with
the AT1 receptor.11
monstrate the important role that this drug class provides in the A brief examination of the origin of RAAS-modifying
treatment of hypertension,1 heart failure,2,3 and renal disease.4 agents provides a framework for understanding the develop-
ment of olmesartan medoxomil. The first series of com-
Received for publication March 17, 2005; accepted July 26, 2005. pounds that were developed to inhibit the RAAS were
From the *New Product Planning, Sankyo Pharma Inc, Parsippany, New
Jersey; †Medical and Scientific Affairs, Sankyo Pharma Inc, Parsippany, polypeptides that inhibited the action of angiotensin-converting
New Jersey; and ‡Pharmacology, Forest Research Institute, Jersey City, enzyme (ACE).12 The nonapeptide teprotide was isolated from
New Jersey. the venom of a Brazilian pit viper, Bothrops jararaca, and
This work was supported by Sankyo Pharma Inc. served as the first ACE inhibitor to be evaluated.13,14 Teprotide
Reprints: Michael K. Pugsley, Pharmacology, Forest Research Institute, 19th
Floor, Harborside Financial Center, Jersey City, NJ 07311 (e-mail:
(SQ 2881) was found to be a highly effective antihypertensive
Michael.Pugsley@frx.com). agent and was devoid of toxicities associated with the pit
Copyright Ó 2005 by Lippincott Williams & Wilkins viper toxin. However, because of the molecule’s peptidic

J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005 585


Mire et al J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005

The peptide antagonists saralasin and sarmesin were


initially synthesized by replacing amino acid residues found
in A-II.18,19 Amino acid modification either reduced or elimi-
nated peptide activity (ie, the ability to activate the AT1 receptor).20
The amino acid sequence of A-II is Asp1-Arg2-Val3-Tyr4-Ile5-
His6-Pro7-Phe8. Replacement of Asp1 with sarcosine, Ile5 with
valine, and Phe8 with alanine resulted in the generation of
saralasin: 1-sarcosine-5-valine-8-alanine angiotensin II or
[Sar1-Val5-Ala8]A-II. Similarly, replacement of Asp1 with
sarcosine and Tyr4 with the methoxy derivative of tyrosine
resulted in production of sarmesin: 1-sarcosine-4-methoxytyr-
osine angiotensin II or Sar-Arg-Val-Tyr(OMe)-Ile-His-Pro-
Phe-OH or [Sar1,Tyr(OMe)4]A-II.20,21
However, despite their efficacy, there were several major
drawbacks associated with this approach to development of
novel antihypertensive drugs. First, these molecules provided
FIGURE 1. The antihypertensive mechanism of angiotensin only partial antagonism of the AT1 receptor.22 Second, as
receptor blockers (ARBs). The diagram depicts the renin–
angiotensin–aldosterone system, in which the renal release of
peptides they had little or no oral bioavailability and, like the
renin catalyzes the conversion of angiotensinogen (derived first ACE inhibitors, had to be administered parenterally.
from the liver) to angiotensin I (A-I). A-I is subsequently Despite these drawbacks, the peptide antagonists provided a
converted to angiotensin II (A-II) by circulating and pulmonary great deal of important mechanistic information regarding the
angiotensin-converting enzyme (ACE). A-II produces a multi- mode of action of A-II binding to AT1 receptors. Modification
tude of physiological effects, including vasoconstriction, of A-II to produce saralasin and sarmesin uncovered crucial
release of aldosterone from the adrenal cortex, and catechol- stereoelectronic (3-dimensional structural and electronic mo-
amine release by activating angiotensin (AT1) receptors on lecular properties) features of the A-II molecule, notably the
vascular smooth muscle. Olmesartan and related ARBs block presence of a backbone bend at the Tyr4-Ile5-His6 segment.
A-II activation of AT1 receptors, providing effective antihyper- The information that resulted showed that to activate the AT1
tensive activity. BP indicates blood pressure.7,8
receptor, the A-II peptide must assume a specific conformation
in which the p-orbitals of the aromatic ring side chains of Tyr4,
structure, parenteral administration was required, thus limiting His6, and Phe8 overlap.23 This allows the establishment of a
its clinical utility. The antihypertensive effect of teprotide was charge-relay system, whereby charge transfer can occur from
shown to result from its ability to inhibit ACE.13 Subsequently, the A-II molecule to the AT1 receptor in the form of pro-
the first orally active ACE inhibitor, captopril, was developed.15 tonation (Fig. 2).11 It has been hypothesized that this charge
This prototypical ACE inhibitor was extensively studied and transfer plays a key role in the activity of A-II on AT1 and the
compared with other efficacious antihypertensive drugs at the ensuing conformational change associated with receptor ac-
time (including a- and b-adrenergic blockers and peripheral tivation and subsequent intracellular cascade mechanisms.24
sympathetic inhibitors such as guanethidine). The use of ACE This hypothesis has been validated through the synthesis of
inhibitors in hypertensive patient populations resulted in well- a cyclic analogue of angiotensin II.25 This conformationally
maintained long-term reductions in blood pressure. Additionally, restricted structure confirmed the necessity of a clustered ring
these compounds demonstrated cardioprotective and renopro- conformation for receptor activation.
tective properties, as evidenced by improved survival rates and
end-organ protection not attributable to blood pressure.16
The Development and Structure–Activity
Relationship (SAR) of Olmesartan
CHEMICAL DEVELOPMENT OF ARBS In the early 1980s, researchers at Takeda Chemical
There are 2 primary receptors for A-II, denoted AT1 Industries, Ltd, discovered the first nonpeptide-based A-II
and AT2.17 The binding of A-II to the AT1 receptor mediates antagonists.26,27 The compounds synthesized were based around
virtually all of the known pathologic effects of A-II that con- a substituted imidazole core (Fig. 3). The first series of mol-
tribute to hypertension. These effects include vasoconstric- ecules produced weak (;38%) inhibition of A-II–mediated
tion, sodium and water retention, and cellular proliferation and contraction of isolated rabbit aortic blood vessels up to
hypertrophy. Activation of the AT2 receptor inhibits gener- a maximal concentration of 10 mM.26 A second series of
alized cellular proliferation and hypertrophy, thereby antag- imidazole-5-acetic acid derivatives showed better in vitro
onizing the trophic responses in tissue induced by A-II.7 inhibitory actions. These molecules produced 100% inhibi-
In vascular endothelial cells, AT2 activation causes dilation of tion of A-II in isolated blood vessels at 1-mM concentra-
blood vessels by formation of nitric oxide and bradykinin.17 tions. However, these concentrations resulted in only 40%
Although the role of the AT2 receptor is less understood, it to 60% inhibition of an A-II–induced pressor effect in rat
usually exerts opposing effects to the AT1 receptor. Direct models.27 Although unsuitable as final marketable products
blockade of the site of action of A-II was first explored through because of low potency and poor oral bioavailability, these
modification of A-II to produce peptide-based antagonists. compounds served as useful leads for the development of

586 q 2005 Lippincott Williams & Wilkins


J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005 Structure and Function of Olmesartan Medoxomil

Many SAR studies examining the imidazole-based


antagonists (ARBs) have been reported and show key elements
required for effective antagonism of AT1, a 7-transmembrane-
spanning G-protein-coupled receptor (GPCR).29–31 The target
sequence for the nonpeptide, imidazole-based antagonists is
the A-II C-terminal pentapeptide Tyr4-Ile5-His6-Pro7-Phe8.31
Analysis of the binding affinity of ARBs to the AT1 receptor
has uncovered a number of common important structural
features. ARB binding involves key acid–base interactions
between acidic moieties of the ARB molecule and basic amino
acid residues on the receptor. Additionally, ARB binding
involves the interaction of hydrophobic substituents with
hydrophobic regions on the receptor, producing weak, but
important, binding forces. Finally, hydrogen bonding between
the ARB and the AT1 receptor is also important for potent
binding affinity.31
Enhancement of binding activity was observed when
a 2#-tetrazole-biphenylmethyl group was introduced at the
imidazole 1-position of the Takeda lead compound. The 2#-
tetrazole-biphenylmethyl group has recently been identified
as a privileged structure. (A privileged structure is a common
molecular framework on which ligands for multiple receptors
can be constructed. This concept has aided in the rational
development of new lead molecules.) This key ligand sub-
structure may increase binding affinity by targeting a conserved
minicore primarily made up of aromatic and nonpolar amino
acid residues within the AT1 receptor.32 The presence of an
acidic group (either a tetrazole or carboxylic acid) at the
2#-position of the biphenyl portion of the molecule provides
FIGURE 2. A schematic that depicts the putative charge for an additional binding position with a basic amino acid group
transfer that occurs from the A-II peptide to the AT1 receptor on, presumably, the highly conserved arginine-167 (Arg167) of
(adapted from Moore and Matsoukas11). transmembrane region IV of the AT1 receptor.33 Similarly, an
acidic group at the 5-position of the imidazole ring provides
a crucial acidic or hydrogen-bonding point of attachment to the
subsequent commercially available ARBs. Thus, currently
AT1 receptor.34 The positively charged lysine 199 (Lys199)
marketed agents are derived as either N-1 biphenyl analogues
residue on transmembrane region V of the AT1 receptor is pro-
of the original N-1 benzyl Takeda lead compounds or are
posed to be the complementary tight binding site for ARBs.35
derivatives in which thienyl modification has taken place at the
It has been proposed that the binding interaction of the
imidazole 5-position of the Takeda lead compounds.28
tetrazole might actually be different from the conventional salt
bridge interaction described and may result from an unusual
lysine–aromatic interaction.36 However, studies have shown
that the charge associated with the Lys199 residue is critical for
proper electrostatic interaction of A-II and high binding
affinity36; thus, potent and efficacious antagonists target this
amino acid binding site. In addition, the nitrogen atom located
at the 3-position of the imidazole ring provides a point for
hydrogen bonding of the receptor.37
Modifications to the original imidazole core led to the
development of olmesartan medoxomil by Sankyo chemists
(Fig. 4A).31,38 Olmesartan medoxomil exists as a prodrug
composed of the parent compound olmesartan that has been
esterified with a (5-methyl-2-oxo-1,3-dioxol-4-yl)methyl
(medoxomil) group (Fig. 4A). Olmesartan medoxomil is
deesterified in vivo via the arylesterase enzyme. This enzyme
is found in both the intestine and plasma. The conversion of
olmesartan medoxomil to the active agent olmesartan occurs
rapidly via the breakdown of the medoxomil ester moiety.39
FIGURE 3. The Takeda lead compound—the first nonpeptide A wide variety of 4-position imidazole antagonists were
AT1 blocker.26,27 constructed, including alkyl, alkenyl, and hydroxyalkyl

q 2005 Lippincott Williams & Wilkins 587


Mire et al J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005

FIGURE 4. A, Chemical structures of olmesartan and olme-


sartan medoxomil. X-ray–determined structure of olmesartan.
B, Olmesartan has intramolecular hydrogen bonding between FIGURE 5. Analogues examined during olmesartan medoxomil
the hydroxyl group and the carboxyl group.31,38 development.31

substituted derivatives. The resulting compounds were the presence of an intermediate-sized alkyl substituent in-
evaluated for their A-II antagonistic activity using in vitro creased binding affinity, likely the result of a lipophilic inter-
binding affinity to AT1 receptors and intravenous and oral action with the receptor, mimicking the isopropyl side chain
inhibitory efficacy on the A-II pressor response in rats.31 When of the Ile5 amino acid in A-II. Thus, as a result of chemical
the alkyl- and alkenyl-substituted molecules were examined and biologic analyses, the C(CH3)2OH substituent at the im-
for biologic activity, it became evident that the presence of idazole 4-position was selected and found to provide for both
bulky alkyl or alkenyl substituent groups at the imidazole 4- moderate bulk and hydrogen-bonding capability of olmesartan
position decreased the binding affinity of the molecule to the medoxomil (Fig. 5).
AT1 receptor. In the case of unsubstituted hydrocarbons, an In conjunction with the chemical and biologic studies
increase in size of the substituent led to a reduction in binding conducted, proton nuclear magnetic resonance spectra (1H
affinity such that H . CH3 ; CH2CH3 . C(=CH2)CH3 . NMR) and x-ray structural analysis of olmesartan provided
CH(CH3)2 in both 2#-tetrazole-biphenyl and 2#-carboxybi- additional insight into the key features of the molecule.31 The
phenyl series (Fig. 5). Addition of the hydroxy group to the x-ray structure revealed that an intramolecular hydrogen
bond formed between the hydroxyl group at the imidazole
alkyl groups (ie, formation of the hydroxyalkyl group)
4-position and the carboxylate anion at the imidazole 5-position
remarkably enhanced the binding affinities of the antagonists.
(Fig. 4B).31,38 Establishment of this hydrogen bond not only
In vivo antihypertensive activity of the hydroxyalkyl
appears to change the electrostatic nature of the imidazole
substituted compounds (as compared with the unsubstituted 5-carboxy group but also may help direct the isopropyl
hydrocarbon analogues) was increased 100-fold versus substituent (a gem-dimethyl group for olmesartan) at the
losartan.31 Additionally, it was found that hydroxyalkyl imidazole 4-position to a more favorable conformation within
substituents bulkier than C(CH3)2OH caused a reduction in the hydrophobic binding region of the AT1 receptor.31,41 This
antagonist activity. idea was further supported through conformational analysis
An assessment of the effect of the acidic substituent at techniques using the PM3 method of MOPAC (Molecular
the biphenyl 2#-position was studied by examining the iden- Orbital Package). MOPAC is an all-purpose semiempirical
tical series of 4- and 5-imidazole derivatives.31 The use of 5- quantum mechanics program that is used as the basis for
substituted-1H-tetrazoles as bioisosteric replacements for car- quantitative SAR studies to predict structural and biologic
boxylic acids has been recently reviewed.40 For all derivatives, properties of molecules.42 This molecular modeling technique
the tetrazole-substituted molecules consistently had greater generated a three-dimensional structure of olmesartan. A
antagonistic activity than the similarly substituted carboxylic stereochemical comparison with the C-terminal pentapeptide
acid derivatives. of A-II suggested that olmesartan overlapped with this region
The end results from these studies show the importance of A-II and emphasized the importance of the hydroxyl group
of a hydroxyl group at the 4-position of the imidazole ring for at the imidazole 4-position for optimal binding affinity and
high-affinity binding of olmesartan to the AT1 receptor. Also, antagonist properties.31

588 q 2005 Lippincott Williams & Wilkins


J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005 Structure and Function of Olmesartan Medoxomil

Mode of AT1 Receptor Antagonism may be the result of its pharmacokinetic properties and slow
To date, many selective, potent, and orally available dissociation from the AT1 receptor. As has been suggested
ARBs have been developed and are used clinically to treat for candesartan cilexitil, the long antihypertensive actions of
hypertension and reduce end-organ damage associated with olmesartan medoxomil may also result from drug accumula-
diseases such as atherosclerosis and diabetes.16 Although all tion at the receptor level or drug reassociation to the receptor
ARBs bind with high affinity to the AT1 receptor,43 there are after dissociation.45
differences in the modes of interaction of ARBs with the
receptor (Table 1).44 The binding of ARBs such as olmesartan,
valsartan, candesartan, telmisartan, and irbesartan to the AT1 An In Vitro Receptor Binding Profile of
receptor displays varying degrees of insurmountability. In con- Olmesartan on AT1 Receptors
trast, losartan, eprosartan, and tasosartan display surmount- The inhibitory effect of olmesartan on [125I]A-II binding
able binding characteristics.43,45 A 2-state, 2-step antagonist was compared with that of related ARBs from data obtained
(A)–AT1 receptor (R) kinetic model has been proposed to best using bovine adrenal cortex membranes that express AT1
explain the changes observed in the dose–response curves for receptors and bovine cerebellum membranes that express
surmountable and insurmountable antagonists.46 AT2 receptors.8 Adrenal cortical membranes (10 mg) were in-
In a study that compared olmesartan to losartan, cubated with [125I]A-II (20 fmol) and increasing concen-
EXP3174, and candesartan, it was observed that olmesartan trations of olmesartan. The binding of [125I]A-II to cell
behaved in an insurmountable manner, but the kinetics of the membranes was quantified using a radioreceptor assay.
inhibitory effect for olmesartan appeared to be greater than The concentration of olmesartan required to produce 50%
that for the other ARBs,8 which may be related to structural inhibition of [125I]A-II binding (IC50) to the AT1 receptor in
features unique to olmesartan.41 This apparent distinction may adrenal cortex was 7.7 6 1.0 nM. An examination of studies
result from the presence of the hydroxyisopropyl substituent at conducted using similar experimental conditions showed
the imidazole 4-position of olmesartan. This substituent can that olmesartan binds the AT1 receptor with an affinity
hydrogen bond with the AT1 receptor and form an intra- comparable to those of valsartan and telmisartan but ;2.0-fold
molecular hydrogen bond with the carboxylic acid moiety at greater than that of candesartan (Table 2). Olmesartan showed
the imidazole 5-position of olmesartan (Fig. 6).41 Although no antagonist activity on AT2 receptors in cerebellar
all ARBs share a common mechanism of action, differences membranes. Olmesartan’s affinity for the AT2 receptor in
in molecular structure may lead to enhanced pharmacolog- these membranes was .100,000 nM or .12,500 times less
ical properties and help explain inherent differences in than that for the AT1 receptor.
efficacy. Thus, although olmesartan and related ARBs are potent
inhibitors of binding of A-II to the AT1 receptor, these binding
studies showed comparable binding affinities between ARBs
THE PHARMACOLOGY OF that produce insurmountable AT1 blockade that is slightly
OLMESARTAN MEDOXOMIL greater than that for agents that produce surmountable an-
Olmesartan produces selective, insurmountable inhibi- tagonism. However, because these studies were not designed
tion of the AT1 receptor and displays no AT2 receptor activity to specifically examine the nature of receptor blockade or to
(AT1 blockade is ;12,500-fold greater than that for AT2).8,47 describe the differences in receptor kinetics, the results are
Olmesartan medoxomil displays a rapid onset of action and a difficult to interpret mechanistically other than that these drugs
long-lasting inhibition of A-II–induced pressor responses that are potent, selective inhibitors of the AT1 receptor.

TABLE 1. A Summary of the Pharmacological Differences Between Surmountable and


Insurmountable Antagonism
Surmountable Insurmountable
Duration of physiological effect Short (minutes) Long (hours)
Receptor dissociation kinetics* Fast (k1 ; k21) Slow, strong (k22 ... k2)
Effects on angiotensin II (A-II) Rightward shift Modest shift
dose–response curve in curve (ED50) in curve (ED50 may increase)
Magnitude of the shift Reduction in maximal
is proportional to response (efficacy [Emax] decreases)
antagonist concentration
No change Bmax decreases
in maximal response
(efficacy) (Emax = E’max)
No change in Bmax
Receptor binding Antagonist can be reversed Antagonism cannot be completely reversed
by increasing A-II concentration by increasing agonist concentration
*All angiotensin receptor blockers (ARBs) dissociate from the AT1 receptor; no clinically used ARB binds irreversibly.
Bmax describes the number of A-II binding sites. Emax describes the maximal tissue response or efficacy for the agonist, A-II. Data derived from Vauquelin
et al.44

q 2005 Lippincott Williams & Wilkins 589


Mire et al J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005

TABLE 3. A Comparison of the Potency of ARB Antagonism


Of A-II–Mediated Vasoconstriction in Isolated Aortic
Tissue Preparations
AT1 Functional Antagonism
ARB [IC50 (nM)]
Candesartan 0.11
Olmesartan 0.13
Eprosartan ;0.29
Telmisartan ;0.33
Valsartan ;0.55
EXP3174 2.45
Irbesartan 4.0
Losartan potassium 26.4
The olmesartan IC50 is derived from a pD2 of 9.9 (pD2 is the negative log of the IC50
value). The IC50 values for eprosartan, telmisartan, and valsartan were calculated from
their pKB values. Data derived from Mizuno et al,8 Merlos et al,48 Koike et al,47 and
FIGURE 6. Proposed interaction of olmesartan with the AT1 Brunner.49 Note that comparisons of AT1 functional antagonism between ARBs are based
receptor.38 on data published from several different studies (see text for details) using isolated aortic
tissue preparations under similar study conditions.

TISSUE-BINDING PROPERTIES: OLMESARTAN


AND RELATED ARBS Olmesartan antagonized the A-II–induced contraction
In addition to binding studies using cell membranes, of isolated guinea pig aortas in a concentration-dependent
functional studies were conducted in A-II–challenged isolated manner (Fig. 7).8 Olmesartan reduced the maximal response
tissue preparations to obtain estimates of the efficacy and (ie, contraction) induced by A-II. As an insurmountable an-
potency of the drug. Olmesartan showed no inhibitory activity tagonist, olmesartan produced a distinctly nonparallel dis-
on the contractile response of vascular tissue when depolariz- placement of the A-II concentration–response curve in these
ing agents such as potassium chloride or receptor-specific tissues. In isolated tissue preparations, the antagonist effect of
agonists such as norepinephrine were used in rabbit aortic olmesartan was increased as the time of drug exposure
preparations or histamine-induced contraction in guinea pig increased, and activity remained for at least 90 minutes after
tissue.8 However, olmesartan showed potent inhibition of washout of the drug.8
A-II–mediated vasoconstriction of isolated aortas. In these
studies, the tissue-blocking effects of the ARB are presented as ANTIHYPERTENSIVE EFFICACY: IN VIVO
IC50 (molar concentration that is required for 50% inhibition ANIMAL MODELS
of the maximal response). The pharmacological activity of
olmesartan was assessed and showed a greater potency than Inhibition of A-II–Mediated Pressor Response
that of other ARBs when data were compared from similar Activation of AT1 receptors by exogenous (intravenous)
studies (Table 3).8,47–49 Specifically, olmesartan was 4.2-fold administration of A-II elicits a pressor response. The pressor
more potent than valsartan, 30.7-fold more potent than
irbesartan, and 203-fold more potent than losartan.

TABLE 2. A Comparison of the Potency of Binding of Several


Arbs to Either AT1 or AT2 Receptors
IC50 (nM)
ARB AT1 (Adrenal Cortex) AT2 (Cerebellum)
Angiotensin II 1.5 0.57
Valsartan 6.5 .30,000
Olmesartan* 7.7 .100,000
Telmisartan 9.2 — FIGURE 7. The effect of olmesartan on guinea pig aortic tissue
Candesartan 12 .100,000 contraction induced by A-II. Denuded guinea pig thoracic aortas
EXP3174* 16 .100,000
were suspended in isolated organ baths containing Krebs-
Henseleit solution. Isometric tension was recorded, and A-II
Losartan potassium 92 .100,000
concentration–response curves were obtained in the absence and
*Indicates that these are the active forms of the molecule, not the prodrug. presence of olmesartan (0.1 and 0.3 nM). The concentration of
Note that EXP3174 is the active metabolite of losartan. Telmisartan binding studies olmesartan that produced half-maximal inhibition of the A-II
were conducted using rat aortic smooth muscle cell preparations. See Koike et al47 for
additional details. Comparisons are based on data published from several studies (see text
response (IC50) was 0.13 nM (Reprinted from European Journal of
for details) using similar study conditions. Pharmacology, Vol. 285, Mizuno et al, Pharmacology of CS-866,
(1995), with permission from Elsevier).

590 q 2005 Lippincott Williams & Wilkins


J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005 Structure and Function of Olmesartan Medoxomil

increases aldosterone levels and enhances sympathetic ner-


TABLE 4. The Inhibitory Effects of ARBs to A-II Pressor
vous system activity.57
Responses In Vivo
In Goldblatt (2K1C) renal hypertensive rats, single oral
Inhibition of A-II Pressor
Responses
doses of olmesartan medoxomil (0.01–0.3 mg/kg) produce a
dose-dependent reduction in blood pressure (Fig. 8).8,41,47
ED50 Relative Olmesartan
ARB (mg/kg) Potency
Inferential data examination from comparable studies for can-
desartan cilexitil, irbesartan, and valsartan are also shown us-
Olmesartan 0.0079 —
ing the same model. The data for the efficacy of each drug in
Candesartan (active metabolite) 0.03 3.8
this model were expressed as the percentage change from con-
Eprosartan 0.10 13
trol value in each study conducted, and data from each study
Telmisartan 0.10 13
were best fit using the Hill equation.41 When these studies were
EXP-3174
(losartan active metabolite) 0.27* 34
compared, the maximal dose of olmesartan medoxomil ad-
Irbesartan 0.45 57
ministered (0.3 mg/kg) produced a 60 mm Hg reduction in
Losartan potassium 0.70 89
blood pressure 3 hours after administration. This response was
Valsartan 10† N/A
similar to that for the 10-mg/kg dose of candesartan and the
30-mg/kg dose of valsartan.
*An anesthetized pithed rat model was used. Other studies used conscious or From the use of these in vitro and in vivo animal models,
anesthetized animals.
†Only an oral ED10 value could be determined in pithed, anesthetized rats. an understanding of the general pathophysiological principles
Data derived from Cazaubon et al,50 Criscione et al,51 Nishikawa et al,52 Mizuno associated with hypertension has been developed. In turn,
et al,8 and Yanagisawa et al.31 these models have lent themselves to characterizing the
Note that comparisons of inhibition of A-II pressor responses between ARBs are
based on data published from several different studies (see text for details). pharmacological potency and efficacy of ARBs that are used
clinically.

response model has routinely been used to assess the efficacy POTENTIAL CLINICAL SIGNIFICANCE
and potency of most ARBs currently used in the clinic. A ARBs share similar structural elements that are required
summary of the results from similar studies but conducted by for effective antagonism of the AT1 receptor. However,
different authors is presented in Table 4.8,31,50–52 A comparison olmesartan medoxomil has unique stereoelectronic features
of pressor response data shows that olmesartan medoxomil that distinguish it from other members of the class. An addi-
is ;4 times more potent than is candesartan and ;89 times tional binding site has been added through incorporation of
more potent than is losartan in this animal model. a hydroxyisopropyl substituent at the imidazole 4-position of
Although all of the ARBs examined in these different olmesartan medoxomil. The hydrogen bonding and hydro-
studies inhibited A-II–mediated pressor responses, drug effects phobic interaction capability provided by this hydroxyl group
were variable and time-dependent. Of all the ARBs examined, and isopropyl group, respectively, increases the binding affi-
olmesartan medoxomil produced the greatest dose-dependent nity of olmesartan medoxomil with the AT1 receptor and
inhibition of the pressor response (Table 4). Olmesartan may help explain its enhanced clinical efficacy. This clinical
medoxomil (0.01 mg/kg) produced ;85% inhibition of the
pressor response within 1 hour after dosing, whereas EXP3174
produced ;70% reduction at a dose of 1 mg/kg (100 times the
olmesartan medoxomil dose), and irbesartan produced ;60%
inhibition at a dose of 10 mg/kg (1000 times the olmesartan
medoxomil dose). This pressor response model has useful
links to human studies where a similar A-II challenge is used
to evaluate the relationship between effectiveness and dose of
a drug.53

GOLDBLATT RENAL HYPERTENSIVE RATS


Hypertension develops in animals in which unilateral
kidney stenosing is produced by placement of a vascular clip
on the renal artery. Goldblatt and colleagues54 published
the first results of a classic study in which experimental
constriction of a renal artery resulted in sustained systemic
hypertension—the 2-kidney, 1-clip (2K1C) model. In 2K1C
models used to evaluate ARBs, constriction of the renal artery FIGURE 8. The antihypertensive effect of olmesartan and
reduces perfusion pressure, resulting in an increase in renin related ARBs in Goldblatt (2K1C) renal hypertensive rats
synthesis and release from the clipped kidney.55,56 The 3 hours postdose.8,41,47 Note that comparisons are based on
elevated circulating A-II level is a crucial factor in the data published from several studies (see text for details) using
pathogenesis of hypertension in this model because A-II the same animal model and similar study conditions.

q 2005 Lippincott Williams & Wilkins 591


Mire et al J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005

efficacy has been demonstrated in a 12-week, randomized, 17. Goodfriend TL. Angiotensins: actions and receptors. In: Izzo JL, Black
parallel-group study with recommended starting doses of HR, eds. Hypertension Primer, The Essentials of High Blood Pressure.
Philadelphia: Lippincott Williams & Wilikins, 2003, pp 8–11.
olmesartan medoxomil (20 mg/d), losartan potassium (50 mg/d), 18. Matsoukas JM, Bigam G, Zhou N, et al. 1H-NMR studies of [Sar1]angiotensin
valsartan (80 mg/d), and irbesartan (150 mg/d). Olmesartan II conformation by nuclear Overhauser effect spectroscopy in the rotating
medoxomil 20 mg/d was significantly more effective than frame (ROESY): clustering of the aromatic rings in dimethylsulfoxide.
starting doses of losartan potassium, valsartan, and irbesartan Peptides. 1990;11:359–366.
19. Matsoukas JM, Yamdagni R, Moore GJ.1H-NMR studies of sarmesin and
in reducing cuff DBP in patients with essential hypertension.58 [des1]sarmesin conformation in dimethylsulfoxide by nuclear Overhauser
This was further demonstrated with significant differences in effect (NOE) enhancement spectroscopy: folding of the N- and C-terminal
24-hour ambulatory blood pressure goal rates versus losartan domains. Peptides. 1990;11:367–374.
potassium and valsartan and numerically greater differences 20. Bovy PR, O’Neal JM, Olins GM, et al. Structure-activity relationships for
from that of irbesartan.59 The clinical results are consistent the carboxy-terminus truncated analogues of angiotension II, a new class
of angiotensin II antagonists. J Med Chem. 1990;33:1477–1482.
with the marked, long-lasting antihypertensive efficacy of 21. Moore GJ, Ganter RC, Matsoukas JM, et al. Receptor interactions of the
olmesartan medoxomil observed in vivo, which may result position 4 side chains of angiotensin II analogues: importance of aromatic
from a unique pharmacological interaction of the molecule ring quadrupole. J Mol Recognit. 1994;7:251–256.
with the AT1 receptor. The nature of this interaction at the 22. Criscione L, Thomann H, Whitebread S, et al. Sarmesin is a partial agonist
of angiotensin-II receptors in rabbit, but not in rat, aortic rings. Biochem
receptor level is being further investigated. Biophys Res Commun. 1990;169:636–642.
Although the receptor binding interactions resulting 23. Carpenter KA, Wilkes BC, Schiller PW. The octapeptide angiotensin II
from the unique structural features of olmesartan medoxomil adopts a well-defined structure in a phospholipid environment. Eur J
are yet to be fully characterized, the observations made at the Biochem. 1998;251:448–453.
receptor, tissue, animal, and clinical levels are consistent. This 24. Rauk A, Hamilton G, Moore GJ. Mechanistic consequences of charge
transfer systems in serine proteases and angiotensin: semiempirical
information provides a framework for comparing angiotensin computations. Biochem Biophys Res Commun. 1987;145:1349–1355.
receptor antagonists and may help explain the clinical dif- 25. Matsoukas JM, Polevaya L, Ancans J, et al. The design and synthesis of
ferences seen within the class. a potent angiotensin II cyclic analogue confirms the ring cluster receptor
conformation of the hormone angiotensin II. Bioorg Med Chem. 2000;8:
1–10.
REFERENCES 26. Furukawa Y, Kishimoto S, Nishikawa K. Hypotensive imidazole
1. Weber MA. Angiotensin II receptor blockers and cardiovascular derivatives. US Patent 4,340,598. Issued to Takeda Chemical Industries,
outcomes: what does the future hold? J Renin Angiotensin Aldosterone Ltd, Osaka, Japan, 1982.
Syst. 2003;4:62–73. 27. Furukawa Y, Kishimoto S, Nishikawa K. Hypotensive imidazole-5-acetic
2. Wong M, Staszewsky L, Latini R, et al. Valsartan benefits left ventricular acid derivatives. US Patent 4,355,040. Issued to Takeda Chemical
structure and function in heart failure: Val-HeFT echocardiographic study. Industries, Ltd, Osaka, Japan, 1982.
J Am Coll Cardiol. 2002;40:970–975. 28. Timmermans PB. Development of Nonpeptidic Angiotensin II Receptor
3. Manohar P, Pina IL. Therapeutic role of angiotensin II receptor blockers in Antagonists. Philadelphia: Hanley & Belfus, 2001.
the treatment of heart failure. Mayo Clin Proc. 2003;78:334–338. 29. Duncia JV, Chiu AT, Carini DJ, et al. The discovery of potent nonpeptide
4. Norris K, Vaughn C. The role of renin–angiotensin–aldosterone system angiotensin II receptor antagonists: a new class of potent antihyperten-
inhibition in chronic kidney disease. Expert Rev Cardiovasc Ther. 2003;1: sives. J Med Chem. 1990;33:1312–1329.
51–63. 30. Timmermans PB, Carini DJ, Chiu AT, et al. Angiotensin II receptor
5. Segura J, Christiansen H, Campo C, et al. How to titrate ACE inhibitors antagonists. From discovery to antihypertensive drugs. Hypertension.
and angiotensin receptor blockers in renal patients: according to blood 1991;18:III136–III142.
pressure or proteinuria? Curr Hypertens Rep. 2003;5:426–429. 31. Yanagisawa H, Amemiya Y, Kanazaki T, et al. Nonpeptide angiotensin II
6. Mimran A, Ribstein J. Angiotensin receptor blockers: pharmacology and receptor antagonists: synthesis, biological activities, and structure-activity
clinical significance. J Am Soc Nephrol. 1999;10(Suppl 12):S273–S277. relationships of imidazole-5-carboxylic acids bearing alkyl, alkenyl, and
7. Carey RM, Wang ZQ, Siragy HM. Role of the angiotensin type 2 receptor hydroxyalkyl substituents at the 4-position and their related compounds.
in the regulation of blood pressure and renal function. Hypertension. J Med Chem. 1996;39:323–338.
2000;35:155–163. 32. Bondensgaard K, Ankersen M, Thogersen H, et al. Recognition of
8. Mizuno M, Sada T, Ikeda M, et al. Pharmacology of CS-866, a novel privileged structures by G-protein coupled receptors. J Med Chem. 2004;
nonpeptide angiotensin II receptor antagonist. Eur J Pharmacol. 1995; 47:888–899.
285:181–188. 33. Yamano Y, Ohyama K, Kikyo M, et al. Mutagenesis and the molecular
9. Ellis ML, Patterson JH. A new class of antihypertensive therapy: modeling of the rat angiotensin II receptor (AT1). J Biol Chem. 1995;270:
angiotensin II receptor antagonists. Pharmacotherapy. 1996;16:849–860. 14024–14030.
10. Israili ZH, Hall WD. Cough and angioneurotic edema associated with 34. Ashton WT, Hutchins SM, Greenlee WJ, et al. Nonpeptide angiotensin II
angiotensin-converting enzyme inhibitor therapy. A review of the antagonists derived from 1H-pyrazole-5-carboxylates and 4-aryl-1H-
literature and pathophysiology. Ann Intern Med. 1992;117:234–242. imidazole-5-carboxylates. J Med Chem. 1993;36:3595–3605.
11. Moore GJ, Matsoukas JM. Angiotensin as a model for hormone–receptor 35. Fierens FL, Vanderheyden PM, Gaborik Z, et al. Lys(199) mutation of the
interactions. Biosci Rep. 1985;5:407–416. human angiotensin type 1 receptor differentially affects the binding of
12. DiBianco R. Adverse reactions with angiotensin converting enzyme surmountable and insurmountable non-peptide antagonists. J Renin
(ACE) inhibitors. Med Toxicol. 1986;1:122–141. Angiotensin Aldosterone Syst. 2000;1:283–288.
13. Vane JR. The history of inhibitors of angiotensin converting enzyme. 36. Noda K, Saad Y, Kinoshita A, et al. Tetrazole and carboxylate groups of
J Physiol Pharmacol. 1999;50:489–498. angiotensin receptor antagonists bind to the same subsite by different
14. Patlak M. From viper’s venom to drug design: treating hypertension. mechanisms. J Biol Chem. 1995;270:2284–2289.
FASEB J. 2004;18:421. 37. Thomas AP, Allott CP, Gibson KH, et al. New nonpeptide angiotensin II
15. Cushman DW, Cheung HS, Sabo EF, et al. Development and design of receptor antagonists. 1. Synthesis, biological properties, and structure–
specific inhibitors of angiotensin-converting enzyme. Am J Cardiol. 1982; activity relationships of 2-alkyl benzimidazole derivatives. J Med Chem.
49:1390–1394. 1992;35:877–885.
16. Gavras H. Historical evolution of angiotensin II receptor blockers: 38. Koike H, Konse T, Sada T, et al. Olmesartan medoxomil, a novel potent
therapeutic advantages. J Am Soc Nephrol. 1999;10(Suppl 12):S255– angiotensin II blocker. Annual Report Sankyo Research Laboratories.
S257. 2003;55:1–91.

592 q 2005 Lippincott Williams & Wilkins


J Cardiovasc Pharmacol ä  Volume 46, Number 5, November 2005 Structure and Function of Olmesartan Medoxomil

39. Laeis P, Puchler K, Kirch W. The pharmacokinetic and metabolic profile 50. Cazaubon C, Gougat J, Bousquet F, et al. Pharmacological characteriza-
of olmesartan medoxomil limits the risk of clinically relevant drug tion of SR 47436, a new nonpeptide AT1 subtype angiotensin II receptor
interaction. J Hypertens Suppl. 2001;19(Suppl 1):S21–S32. antagonist. J Pharmacol Exp Ther. 1993;265:826–834.
40. Herr RJ.5-Substituted-1H-tetrazoles as carboxylic acid isosteres: medic- 51. Criscione L, de Gasparo M, Buhlmayer P, et al. Pharmacological profile of
inal chemistry and synthetic methods. Bioorg Med Chem. 2002;10:3379– valsartan: a potent, orally active, nonpeptide antagonist of the angiotensin
3393. II AT1-receptor subtype. Br J Pharmacol. 1993;110:761–771.
41. Pugsley MK, Mire DE, Silfani TN, et al. The dose-dependent anti- 52. Nishikawa K, Inada Y, Shibouta Y, et al. Pharmacological profile of a
hypertensive actions of olmesartan medoxomil, an angiotensin (AT) II novel nonpeptide angiotensin II subtype 1 receptor antagonist, TCV-116.
receptor blocker. Am J Hypertens. 2004;17:42. Blood Press Suppl. 1994;5:7–14.
42. Stewart JJP. MOSOL, MOPAC for solid-state physics. Quant Chem Prog 53. Brunner HR, Nussberger J. Relevance of clinical pharmacological models
Exch. 1985;5:62–63. for the evaluation of therapeutic dose range of an AT1-receptor antagonist.
43. Timmermans PB. Pharmacological properties of angiotensin II receptor J Hypertens Suppl. 2001;19(Suppl 1):S15–S20.
antagonists. Can J Cardiol. 1999;15(Suppl F):26F–28F. 54. Goldblatt H, Lynch J, Hanzel RF. Studies on experimental hypertension:
44. Vauquelin G, Van Liefde I, Vanderheyden P. Models and methods for I. The production of persistance elevation of systolic blood pressure by
studying insurmountable antagonism. Trends Pharmacol Sci. 2002;23: means of renal ischemia. J Exp Med. 1934;59:347–379.
514–518. 55. Robertson JI. Renin and the pathophysiology of renovascular hyperten-
45. Morsing P, Vauquelin G. How can the differences among AT1-receptor sion. In: Robertson JI, ed: The Renin–Angiotensin System. New York:
antagonists be explained? Cell Biochem Biophys. 2001;35:89–102. Gower Medical Publishing 1993, pp 55.1–55.34.
46. Vauquelin G, Morsing P, Fierens FL, et al. A two-state receptor model for 56. Navar LG, Zou L, Von Thun A, et al. Unraveling the mystery of Goldblatt
the interaction between angiotensin II type 1 receptors and non-peptide hypertension. News Physiol Sci. 1998;13:170–176.
antagonists. Biochem Pharmacol. 2001;61:277–284. 57. Ploth DW. Angiotensin-dependent renal mechanisms in two-kidney, one-
47. Koike H, Sada T, Mizuno M. In vitro and in vivo pharmacology of clip renal vascular hypertension. Am J Physiol. 1983;245:F131–F141.
olmesartan medoxomil, an angiotensin II type AT1 receptor antagonist. 58. Oparil S, Williams D, Chrysant SG, et al. Comparative efficacy of
J Hypertens Suppl. 2001;19(Suppl 1):S3–14. olmesartan, losartan, valsartan, and irbesartan in the control of essential
48. Merlos M, Rabasseda X, Silvestre JS. Discovery of new angiotensin II hypertension. J Clin Hypertens (Greenwich). 2001;3:283–291, 318.
receptor antagonists. A review of pharmacological studies. Methods Find 59. Smith DH, Dubiel R, Jones M. Use of 24-hour ambulatory blood pressure
Exp Clin Pharmacol. 1998;20:805–815. monitoring to assess antihypertensive efficacy: a comparison of olmesartan
49. Brunner HR. The new oral angiotensin II antagonist olmesartan medoxomil: medoxomil, losartan potassium, valsartan, and irbesartan. Am J Cardiovasc
a concise overview. J Hum Hypertens. 2002;16(Suppl 2):S13–S16. Drugs. 2005;5:41–50.

q 2005 Lippincott Williams & Wilkins 593

You might also like