You are on page 1of 14

This article was downloaded by: [Emory University]

On: 09 August 2015, At: 15:52


Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick
Place, London, SW1P 1WG

Expert Opinion on Drug Discovery


Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/iedc20

Recent advances in the design and synthesis of


prazosin derivatives
a a a a
Anna Minarini , Maria Laura Bolognesi , Vincenzo Tumiatti & Carlo Melchiorre
a
Department of Pharmaceutical Sciences, University of Bologna, Via Belmeloro 6,
40126 Bologna, Italy.
Published online: 31 Mar 2015.

To cite this article: Anna Minarini, Maria Laura Bolognesi, Vincenzo Tumiatti & Carlo Melchiorre (2006) Recent advances
in the design and synthesis of prazosin derivatives, Expert Opinion on Drug Discovery, 1:5, 395-407

To link to this article: http://dx.doi.org/10.1517/17460441.1.5.395

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained
in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no
representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose
of the Content. Any opinions and views expressed in this publication are the opinions and views of the
authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not
be relied upon and should be independently verified with primary sources of information. Taylor and Francis
shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and
other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation
to or arising out of the use of the Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any
form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://
www.tandfonline.com/page/terms-and-conditions
Review

Recent advances in the design and


synthesis of prazosin derivatives
Anna Minarini†, Maria Laura Bolognesi, Vincenzo Tumiatti
& Carlo Melchiorre
†Department of Pharmaceutical Sciences, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
1. Introduction
2. Design of prazosin derivatives
Due to the large number of therapeutic indications, particularly for antago-
3. Synthesis of prazosin derivatives nists, a field of constant interest in medicinal chemistry is the design, synthesis
4. Conclusion and pharmacological evaluation of ligands that bind at α1-adrenoreceptors,
5. Expert opinion which include α1a, α1b and α1d subtypes. This review, which is mainly based on
literature over the past 10 years, focuses on the developments of some of the
most potent, selective and widely used α1-adrenoreceptor antagonists that are
structurally related to prazosin (which is characterised by a quinazoline moi-
ety). Initially introduced in therapy for the management of hypertension, pra-
zosin derivatives have become increasingly common in the treatment of
Downloaded by [Emory University] at 15:52 09 August 2015

benign prostatic hyperplasia. During the past few years, new perspectives have
emerged for the use of these derivatives as a novel class of apoptosis-inducing
agents in prostate cancer.

Keywords: α-adrenergic antagonist, cyclazosin, prazosin, quinazoline

Expert Opin. Drug Discov. (2006) 1(5):395-:407

1. Introduction

The α1-adrenoreceptors (α1-ARs) are classified into three well-characterised sub-


types, based on their molecular and pharmacological properties namely, α1a, α1b,
and α1d [1,2]. The effort to design agents selective for each of the three α1-AR sub-
types has been an active area of research due to the wide number of possible thera-
peutic applications. The α1-AR family is of great therapeutic interest because of its
fundamental role in cardiovascular function, particularly in the regulation of blood
pressure [3]. Although initially introduced for the management of hypertension,
α1-AR antagonists have become increasingly common in the treatment of lower uri-
nary tract symptoms (LUTS) and benign prostatic hyperplasia (BPH), as they relax
smooth muscle and thereby relieve urinary retention [4-7].
Some of the most potent, selective and widely used α1-AR antagonists are struc-
turally characterised by a quinazoline ring. Prazosin (1) represents the progenitor of
this class, and was one of the first α1-AR antagonists able to discriminate between
α1- and α2- families that were introduced in therapy as an antihypertensive agent.
Its antihypertensive activity depends on peripheral vasodilation mediated by a
postjunctional α1-AR blockade. However, due to the lack of selectivity of prazosin
among the three α1-AR subtypes, much research effort has been directed towards
the design and synthesis of more selective compounds using the structure of pra-
zosin as a template. Some congeners of prazosin, such as terazosin (2), doxazosin (3)
and alfuzosin (4), which offer a similar antihypertensive effect with a longer duration
of action, have been widely studied for the treatment of BPH [6,8]. The efficacy of
these α1-AR antagonists in BPH is balanced against a small, but significant, inci-
dence of side effects, such as orthostatic hypotension. Due to its preferential
distribution into the prostate gland, compound 4 emerged as a uroselective agent for
the management of BPH associated with a significantly lower incidence of
cardiovascular effects [9] (Figure 1).

10.1517/17460441.1.5.395 © 2006 Informa UK Ltd ISSN 1746-0441 395


Recent advances in the design and synthesis of prazosin derivatives

NH 2
NH 2 N H2
O
N O O
N N
O N N O O
O N N O O N N
N
N N
O O
1 Prazosin 2 Terazosin O 3 Doxazosin O

NH 2 NH 2
O O
N O N H2
R2 O
O N
O N N N O N
H
R1 R2
O
4 Alfuzosin 5
6 R1
NH 2
NH 2 O N H2
N
O O
N N
O N N
O N N N O N N
Downloaded by [Emory University] at 15:52 09 August 2015

CO O CH 2 Ph
N N N O
O CO NHt-Bu
10 O O 11 L-765314 N N
12
O
N H2
O HN N O
N O
N HN
N
O N N
H R N N
N N CO O CH 2 CH 3
O
N
H N N
O H
13 33 FH-71
34 EW -154
O
O
O NH O R2 O
RO
NH O N N NH
Y
O N N O R1O N N Ar
N R3
N

36 37
35
O

NH 2 N H2
O O
O O
O N
N
N N3
N O N N
O N O
X N N
HN R N
Ar I

O O
38 39 52 IAAP

Figure 1. Structure of prazosin (1) and related α-AR antagonists.

The relative importance of the various α1-AR subtypes in selectivity for inhibition of prostatic pressure/tone versus
BPH is still controversial as it has yet to be determined blood pressure [10]. On the other hand, increasing evidence
which subtype-selectivity profile provides the best therapeu- indicates that the α1d-AR subtype predominates in the blad-
tic advantage in terms of targeting the prostate. However, der detrusor muscle, supporting its role in mediating the
high affinity and selectivity for prostatic α1a-ARs, irritative symptoms in BPH. In fact, the
particularly versus the α1b subtype, seem to confer optimal methoxybenzenesulfonamide-based α1-AR antagonist

396 Expert Opin. Drug Discov. (2006) 1(5)


Minarini, Bolognesi, Tumiatti & Melchiorre

Later, due to the large availability of α1-AR antagonists,


many groups have attempted to develop a pharmacophore for
α1-AR subtypes, reaching the conclusion that it is not possible
to construct a model that is valid for all the structurally differ-
ent classes of ligands. In particular, the relative positions of the
protonated nitrogen atom and the aromatic systems differ for
quinazoline derivatives, raising the so-called ‘prazosin prob-
lem’. From the obtained results, it is quite likely that the pra-
zosin derivatives (which generally exhibit very high affinity for
all three α1-AR subtypes, but little subtype selectivity) bind to
a site that is different from that of other antagonists, and
common to all α1-ARs [14,15].
More recently, a model of the prazosin α1-AR interaction,
built by the use of the coordinates of rhodopsin, confirmed
the N-1 function on the quinazoline ring to be one of the key
groups involved in receptor binding, together with the two
methoxy moieties, the 4-amino group and the carbonyl func-
Downloaded by [Emory University] at 15:52 09 August 2015

tion. The docking studies into all the α1-AR subtypes sug-
gested that the 4-amino group on the quinazoline ring
possibly interacts with the carboxyl group of an aspartate resi-
due in transmembrane (TM)3 and the N-1 atom with the
Figure 2. Complex of prazosin (1) at the ligand binding site.
EL: Extracellular loop; TM: Transmembrane domain.
hydroxyl group of a serine in TM5, through hydrogen bond
Reprinted from M Ishiguro, Y Futabayashi, T Ohnuki, M Ahmed, formations. Furthermore, the two methoxy groups of quina-
I Muramatsu, T Nagamoto: Identification of binding site of prazosin, tamsulosin
and KMD-3213 with α-adrenergic receptor subtypes by molecular modeling.
zoline ring may also interact with hydroxyl groups of a threo-
Life Sciences, 71, 2531-2541, Copyright (2002), with permission from Elsevier. nine in TM3 and of a serine in TM5, whereas the carbonyl
group is presumed to form a hydrogen bond with a serine in
tamsulosin, which has high affinity for α1a-AR and α1d-AR TM6 (Figure 2). The suggested identical interactions for pra-
subtypes over α1b-ARs, shows efficacy in BPH associated zosin within all the α1-AR subtypes, accounted for its lack of
with fewer side effects [7]. This is similar to the nonsubtype selectivity [16].
selective compounds 2 and 3. Therefore, in the last decade, One of the first attempts to manipulate the structure of
research efforts in this area have been directed towards the prazosin was the replacement of its furan ring with a 1,4-ben-
discovery of subtype-selective quinazoline derivatives. The zodioxane moiety, which is a well known pharmacaphore for
aim of this review is to provide an update on the current antihypertensive activity [17]. This modification led to the dis-
knowledge of the design and synthesis of α1-antagonists covery of compound 3, endowed with a longer duration of
based on the structure of prazosin. action that allowed a once-daily administration in humans.
To demonstrate the importance of the N-1 atom and the
2. Design of prazosin derivatives effect of its protonability, the quinazoline nucleus of prazosin
was replaced by isosteric heteroaromatic systems such as quin-
To investigate the role of the different main fragments of pra- oline (5) and isoquinoline (6) rings. The comparable biologi-
zosin in receptor binding, a wide range of structure–activity cal activity in binding assays of compound 5 derivatives with
relationship studies were performed on the structure of pra- prazosin demonstrated that, presumably, their nitrogen atoms
zosin. To this end, multiple modifications involved the play a common role in the α-AR recognition process, as their
piperazine and furan rings, mainly by conserving the basic values allowed protonation at physiological pH. The
2,4-diamino-6,7-dimethoxyquinazoline moiety. poor affinity showed by compound 6 derivatives confirmed
The role of the 2,4-diamino-6,7-dimethoxyquinazoline that isosteric relocation of the nitrogen atom within the ring
moiety in the receptor recognition process has been was unacceptable, and outlined the importance of N-1-proto-
investigated in depth. Initially, it was suggested that this nation in the receptor interaction process [18,19].
moiety could act as a conformationally restricted form of On the basis of these results and once the electronic
the noradrenaline cation and that a dominant role in the requirements of the quinazoline moiety were satisfied, most of
receptor recognition process was played by the protonated the efforts to modulate binding affinities were directed to the
N-1 of the quinazoline system, which mimicked the amine modification of the quinazoline 2-substituent. Thus, the role
function of the neurotransmitter [11]. In particular, a of the piperazine ring was initially investigated through its
charged-reinforced H-bond interaction involving the replacement by an α,ω-alkanediamine chain, revealing that
quinazoline-N-1 function and an anionic site of the this moiety may not be essential for α1 blocking activity, and
receptor was highlighted [12,13]. that the length of the alkane chain and N-methylation of both

Expert Opin. Drug Discov. (2006) 1(5) 397


Recent advances in the design and synthesis of prazosin derivatives

NH2
O
N

O N N
N
O
O

Prazosin 1 α1A = 8.60


α1B = 8.99
α1D = 8.91

NH2
O
Downloaded by [Emory University] at 15:52 09 August 2015

O N N
N
O
O

7: α1A = 9.04
α1B = 9.84
α1D = 9.19

NH2 NH2
O O
N N

O N N O N N
H
N N
O O
H
O S O
S

(+)-Cyclazosin 8: α1A = 7.73 Cystazosin 9: α1A = 7.53


α1B = 8.85 α1B = 7.49
α1D = 7.27 α1D = 8.54

Figure 3. Design strategy and affinity profile (pA2 values) at α-AR subtypes of compounds 7 – 9 [23].
the amide and the 2-amino function could efficiently tune (in which the hexamethylene chain of compound 7 is partially
affinity and selectivity towards α1- over α2-ARs. In this con- or totally incorporated into a constrained nucleus) were
text, compound 7 with a hexamethylenediamino chain, dis- designed. The aim of this structural modification was to keep
played the higher antagonist potency and selectivity (Figure 3) the quinazoline and furan rings in a position similar to that of
[20]. Consequently, different structural modifications were prazosin, forcing the alkane moiety to assume a definite spa-
performed on the piperazine ring, all of them leading to com- tial arrangement. The difference in activity between the two
pounds supporting the presence of a lipophilic binding area cis/trans diasteroisomers not only supported the presence of
on the surface of the α1-AR [21]. To better define the size and the lipophilic binding area on α1-ARs surface, but, in addi-
the sterochemical requirements of this area, several derivatives tion, demonstrated that the lipophilic pocket is endowed with

398 Expert Opin. Drug Discov. (2006) 1(5)


Minarini, Bolognesi, Tumiatti & Melchiorre

OCH 3
X NH 2
N ( ) N
H 4 H O
N
15 X = S
O N N
2 R
N
14
O

NH 2
O
N
CH 2 NH(CH 2)6 NH(CH 2)2 X
O N N
N
Downloaded by [Emory University] at 15:52 09 August 2015

O 2
16 – 21 X = S, CH 2

NH 2
O
N
CH 2 NH(CH 2)6 NH(CH 2)2 X
O N N
N

O 2
22 – 27 X = S, CH 2

Figure 4. Design strategy of derivatives 16 – 27.

a well-defined size and spatial orientation. Among the synthe- been confirmed in functional studies that have been performed
sised compounds (+)-cyclazosin (8) [22], which incorporates a on rat and rabbit tissues, making it a useful tool for the pharma-
cis-perhydroquinoxaline nucleus, displayed the most promis- cological characterisation of the α1b-AR [28].
ing affinity profile, leading to a selective α1b-AR antagonist, as In the search for new analogues compound 8 may demon-
reviewed in detail [23] (Figure 3). strate increased affinity and selectivity for α1b-ARs, a
A steric discrimination is probably responsible for the number of racemic compounds, bearing different substitu-
α1b-AR selectivity of compound 8, in line with the trend of ents as an alternative to the 2-furoyl moiety, were synthe-
affinity found for compound 10 [24] and L-765314 (11) [25], sised and pharmacologically evaluated [29]. Binding results
which also bear bulky substituents and a preferential interac- indicated that the majority of the compounds showed a
tion with the α1b-AR subtype. Furthermore, cystazosin (9), trend of preferential affinity for α1d- and α1b-ARs, similar to
obtained by replacing the hexanediamino chain of compound that of racemic compound 8, although the most potent of
7 with a cystamine moiety, displayed a favourable affinity the series, the 4,6-dimethoxy-substituted triazine derivative
profile, leading to a selective α1d-AR antagonist [26]. compound 12 (Figure 1) presented comparable affinity at
In 2004, the (4aR,8aS) configuration of compound 8 was α1a-, α1b- and α1d-ARs, with pKi values of 10.15, 10.22 and
assigned by means of an X-ray crystallographic study of the 10.40, respectively. Taken together, the binding results indi-
(R,R)-(+)-tartrate salts of the chiral precursor (+)- and (-)-2-furyl cated that the α1a-AR would express a more specific struc-
(cis-perhydro-1-quinoxalinyl)-methanone [27]. The higher tural demand in the N-4-substituent for optimum
α1b-AR selectivity of compound 8 relative to the (-)-enantiomer interaction, when compared with α1d- and α1b-ARs [29].
has to be ascribed to its lower affinity towards the other α1-AR In a subsequent work on compound 8 derivatives, the role
subtypes, which might be due to a negative influence of the of different 5-furan ring substituents towards selectivity and
tetramethylene portion of the perhydroquinoxaline system in the affinity for α1-AR subtypes was explored. To this end, substit-
interaction process with α1a- and α1d-AR subtypes. The α1b-AR uents with different electronic and/or lipophilic properties
selectivity of compound 8 over α1a- and α1d-AR subtypes has like bromine, methyl, methoxy or acetyl, were chosen (see

Expert Opin. Drug Discov. (2006) 1(5) 399


Recent advances in the design and synthesis of prazosin derivatives

NH2
O
N

O
O N N N O
N O O N
N S
O O R
R 28a R = 3-Ph
28b R = 4-Ph

NH2
HO ( )
O n NH2
N S S
O O
N
O N N O
N O N N
N ( )
O n
1 S
Downloaded by [Emory University] at 15:52 09 August 2015

29 n = 4 O S
30 n = 1
31 n = 0
O

NH2
O
N O

O O N N
N

32 O O

Figure 5. Hybrid 1-related derivatives 28 – 32.

compound 13 for a general structure – Figure 1). All of the nists, whereas all the polyamine disulfides displayed a nonho-
new designed compounds maintained the α1b-AR selectivity, mogeneous mechanism of inhibition at the three subtypes as
although it was lower than that of compound 8 [30]. they were, like compound 15 noncompetitive antagonists at
As increasing the number of contacts between the ligand the α1a- and α1b-AR subtypes. However, unlike compound
and its receptor would hopefully increase subtype selectivity, 15, the polyamine disulfides were competitive antagonists at
the furan ring of prazosin and compound 9 and its carbon the α1d-AR. These results may suggest the presence, in the
analogue 7 was replaced by a phenyl ring (14), thus offering binding pocket of α1a- and α1b-ARs, of a suitable binding
the possibility of incorporating additional structural elements thiol function that would experience an interchange reaction
at different positions (Figure 4). Interestingly, the structural with the disulfide moiety of the antagonist and which would
modifications performed did not improve affinity for α1-AR be missing, or not accessible, in the α1d-AR subtype [32].
subtypes; however, importantly, they did give rise to selectiv- An attractive strategy for new prazosin derivatives was
ity [23,31]. Moreover, a most intriguing finding was the obser- applied by Gasco et al. [33] to obtain antihypertensive agents
vation that the insertion of a diaminohexane chain on the with two distinct vasodilation mechanisms: α1-AR
benzene ring did not induce negative interactions with the antagonism and nitric oxide (NO) mediated vasodilation [33].
receptor, thus opening the path for the development of new For this purpose, some hybrid compounds were synthesised
quinazolines bearing a polyamine backbone. On the basis of by combining two pharmacophoric units, such as the quina-
these results, hybrid tetra-amine disulfides were synthesised by zoline ring of prazosin and the NO-prodrug furoxan moiety
combining the structural features of prazosin or compound 7 for the furanylcarbonyl function in prazosin. The
and benextramine 15, an irreversible α1/α2-AR antagonist. (furoxanylsulfonyl)piperidine derivatives 28a and 28b (Figure
With the aim of verifying the role of the disulfide moiety of 5) showed NO vasodilation and α1-AR antagonism activities
the latter on the interaction with α1-AR subtypes, the corre- in an appropriate balance.
sponding carbon analogues were included in the study. Of the Following an analogous approach, further modifications
synthesised compounds (16 – 27), quinazolines that lacked the were performed on the lead structure (1) with the aim of wid-
disulfide bridge behaved, like prazosin, as competitive antago- ening its biological profile. Thus, to generate antioxidant and

400 Expert Opin. Drug Discov. (2006) 1(5)


Minarini, Bolognesi, Tumiatti & Melchiorre

Table 1. Affinity profile of compounds 29 – 32 at α1-AR subtypesa.

NH2 NH2
O O
N N

O N N O N N O
N (CH2)n N
S S
O 32

29 – 31
O

No n pKB or pA2 pKi

α1A α1B α1D α1a α1b α1d


1 8.93 ± 0.10 9.06 ± 0.11 8.93 ± 0.10 9.23 9.39 9.65
29 4 7.19 ± 0.10 7.39 ± 0.12 7.92 ± 0.22 8.85 8.90 9.05
Downloaded by [Emory University] at 15:52 09 August 2015

30 1 9.00 ± 0.11 8.99 ± 0.21 9.04 ± 0.06 9.54 9.51 9.41


31 0 7.73 ± 0.05 8.76 ± 0.20 9.06 ± 0.06 9.09 9.27 9.31
32 7.62 ± 0.06 8.53 ± 0.12 9.07 ± 0.01 8.90 9.17 9.03
Lipoic acid <5 <5 <5

a Affinity constants, expressed as pK or pA (isolated tissues) or pK (Chinese hamster ovary cells) values.
B 2 i
Data from [34].

antiproliferative activities in the same molecule, the furoyl performed at the piperazine and aromatic rings, keeping the
moiety of prazosin was replaced either with the lipoyl frag- 2,4-diamino-6,7-dimethoxyquinazoline portion unaltered.
ment of the well-known antioxidant lipoic acid (LA) (or its However, the pharmacological data of some quinazo-
lower homologues), or with 1,4-naphthoquinone, thereby lin-4-(3H)-one derivatives, patented in recent years by phar-
affording (29 – 31) and compound 32, respectively [34] maceutical companies [37], attested that structural variations
(Figure 5). In particular, the pharmacological profiles of com- of the quinazoline ring can be successfully attempted. Com-
pounds (29 – 32) at α1-AR subtypes, evaluated by functional pound 35 displays very high antagonistic potency at the puta-
and binding studies, were very promising in terms of both tive rabbit aorta α1L-AR subtype and is claimed to be useful
affinity and selectivity (Table 1). Compounds 29 and 32 5were for treating BPH, sexual dysfunctions and hypertension. This
able to protect prostate cells against reactive oxygen species putative α1L-AR subtype, has been found in vascular and
formation evoked by oxidative stress, indicating that antioxi- lower urinary tract smooth muscle, and has been referred to as
dant properties were retained only by compounds that incor- α1L-AR because of its low affinity for prazosin. However,
porated the 1,4-naphthoquinone and LA structure, but not some studies have reported the hypothesis that α1L-AR could
that of its lower homologues 30 and 31. In contrast to com- represent a functional phenotype of the α1a-AR subtype.
pound 29, compounds 30 – 32 displayed an antiproliferative Other quinazolinone derivatives, exemplified by compound
activity higher than that of prototype prazosin [35]. This study 36, are stated to be selective towards α1a/α1b-AR subtypes over
showed that it is possible to obtain multipotent drugs able to the α1d-AR subtype and are claimed for the treatment of BHP
display both a potent α1-AR antagonism and antiproliferative with reduced side effects [37]. More recently, a series of 2-aryl
activity combined with the capacity to inhibit oxidative stress. substituted quinazolinone derivatives of general formula 37,
The amino-quinazoline moiety of prazosin and its conge- emerged as α1a- and α1b-AR selective antagonists, particularly
ners has also been modified by introducing a substituent on when the R2 functional group is not hydrogen [101] (Figure 1).
the 4-amino group, leading to a series of compounds from Further modifications of the quinazoline substructure
which FH-71 (33) and EW-154 (34) emerge for their selectiv- were reported in a study describing the design, synthesis and
ity for α1a- and α1b-ARs, respectively, in both functional and biological evaluation of a series of compounds where the
binding experiments [36] (Figure 1). quinazoline moiety of prazosin was replaced with a tetrahy-
As a consequence of the early evidence on the essential role droacridine system. The constrained structure preserved the
played by the piperazine ring, most of the quinazolines N-1 basicity, but forced key features to assume a reciprocal
described so far have been derived from modifications arrangement, probably similar to that of prazosin in one of

Expert Opin. Drug Discov. (2006) 1(5) 401


Recent advances in the design and synthesis of prazosin derivatives

H
O NH2 1) NaOCN O N OH O N O

N NH
O COOH O O
2) HCl
OH O
40
41
PCl5
POCl3
O
HN N O
O N Cl O N Cl
NH3
1
N N
O O
NH2 Cl
43
42

Figure 6. Classical synthesis of 1.


Downloaded by [Emory University] at 15:52 09 August 2015

O NH2 N

1) (COCl)2 H H
O O O N N
N

OH O S
2) Bu4NNCS N C S O
O
44 45 46
O

N
O
N
H
N

H
N N
CF3COOH
1. CF3COOH O N
O
O
N

O
O
47

Figure 7. Synthesis of 1 by solid-phase approach.

its low-energy conformations. Despite the intriguing simi- interesting pharmacophore at the α1b-AR subtype, leading
larity shown by prazosin and compound 38 with respect to to a significant affinity, which does not seem to be influ-
the relative positions of the pharmacophoric functions, the enced by the nature of a lateral chain [38].
overall series of derivatives was less potent in all the α1-AR Finally, in 2006, some imidazo[5,1-b]quinazolines linked
subtypes relative to prazosin, with most displaying a ten- to aryl piperazines, exemplified by the general structure com-
dency to selectivity for the α1b-AR subtype in both func- pound 39 (Figure 1) were synthesised as prazosin-constrained
tional and binding experiments. This finding suggests that analogues and proved that the structure–activity relationship
the newly introduced tricyclic system represents an for α1-AR antagonists (hypotensive activity) included the

402 Expert Opin. Drug Discov. (2006) 1(5)


Minarini, Bolognesi, Tumiatti & Melchiorre

Cl
O
N
R'
R' N
O N Cl N
O
NH O R 2R 3 N N
N
R' R2
O N N
O N Cl
R3
48 49 50

CF3COOH

R'
NH

O
N
R2
O N N
Downloaded by [Emory University] at 15:52 09 August 2015

R3
51

Figure 8. Solid-phase approach for the synthesis of a 2,4-diaminoquinazoline library.

absence of any substituents at the phenyl group of the quinazoline nucleus, exhaustively reviewed by Vogtle [40]. The
arylpiperazine moiety and had no branching at the oxoethyl authors limit their report to the synthesis of the
linker. Unfortunately, data on subtype selectivity of the syn- 2,4-diaminoquinazoline scaffold, and in particular, to the
thesised compounds were not reported [39]. solid-phase synthesis of prazosin, but several other methods
associated with the synthesis of other quinazoline-related
3. Synthesis of prazosin derivatives nuclei are present in the literature. Wilson [41] reported the
solid-phase synthesis of 2,4-diaminoquinazoline, starting
Due to the importance of quinazoline derivatives for the from an acylisothiocyanate resin (45) and generated from
pharmaceutical community and in parallel with the search carboxy polystyrene (44). Resin (45) was condensated with
for new lead compounds, many research efforts have been 2-amino-4,5-dimethoxybenzonitrile and subsequently treated
devoted to developing readily accessible and efficient with 1-(2-furoyl)-piperazine to give the guanidine derivative
synthetic methodologies. 47, which was treated with trifluoroacetic acid to afford
The first synthesis of prazosin, was described by Hess prazosin with a yield of 70% (Figure 7).
et al. [102]. Prazosin (1) was prepared by the cyclisation of the A more general and versatile method was described by Dhanoa
ureido derivative of the 6-aminoveratric acid (40) with et al. [42] and later slightly modified by Dener et al. [43]. The start-
sodium or potassium cyanate in acidic aqueous media. The ing material was represented by polymer bound amines (48)
resulting 2,4-dihydroxy-6,7-dimethoxyquinazoline (41) was which reacted with 6,7-dimethoxy-2,4-dichloroquinazoline fol-
chlorinated at the 2- and 4-positions with a mixture of phos- lowed by an aromatic nucleophilic substitution with the appro-
phorus pentachloride and phosphoryl chloride. By reacting priate amine and subsequent cleavage to give the desired
with an excess of ammonia in tetrahydrofuran, the resulting quinazoline (51) (Figure 8)
2,4-dichloroquinazoline (42), was converted in A modified synthesis of the antihypertensive agent iodoazi-
2-chloro-4-aminoquinazoline (43). Finally, the chloro doarylprazosin (52) [44] (Figure 1) has been published recently.
derivative was condensed with 1-(2-furoyl)piperazine to Compound 52 is a prazosin derivative shown to function as a
generate prazosin (Figure 6). multi-drug-resistance reversal agent and used as a standard
A more recent approach to the synthesis of quinazoline photo-affinity label for competition assays on P-glycoprotein.
derivatives is represented by solid-phase and combinatorial The newly developed convergent route to compound 52
methodologies. These innovative techniques allowed an involved a direct addition of an acylated piperazine
intense production of several libraries based on the intermediate to the 2,4-dichloroquinazoline. Moreover,

Expert Opin. Drug Discov. (2006) 1(5) 403


Recent advances in the design and synthesis of prazosin derivatives

palladium imidazolium heteroatom coupling was also prostate cancer prevention and therapy [49]. It is plausible
investigated as an alternative and more efficient route to that quinazoline-based derivatives, through competing
obtain 6,7-dimethoxy-2-piperazin-1-yl-quinazo- with ATP binding, interfere with an as yet, unidentified
lin-4-ylamine, a useful intermediate in the preparation of pra- tyrosine kinase that uses Akt as an effector. It is worth not-
zosin derivatives. ing that these compound 3-derived agents are structurally
distinct from the quinazoline-based inhibitors of epidermal
4. Conclusion growth factor receptor (EGFR) tyrosine kinases such as
erlotinib and gefitinib. These two anilinoquinazoline
Although some of the early α1-AR antagonists are still used in derivatives recently approved for the second- and third-line
the treatment of hypertension, the newly disclosed com- treatment of non-small-cell lung cancer, act as EGFR tyro-
pounds are mainly used in urological pathologies. Through sine kinase inhibitors and compete for the ATP-binding
the developments of some of the most potent, selective and site in the intracellular domain, inhibiting EGFR auto-
widely used α1-AR antagonists structurally related and unre- phosphorylation. This strategy for developing targeted
lated to prazosin, the past 10 years have seen considerable anticancer therapy has become more important than the
gains in understanding the role of α1-ARs in the aetiology of optimisation of therapy with conventional anticancer
LUTS/BPH. As a result, more uroselective and less vasoactive drugs [50,51].
α1-AR antagonists such as compound 4 have been developed. Further support for the multiple biological actions of
Downloaded by [Emory University] at 15:52 09 August 2015

A wealth of evidence has established the α1-AR antagonists to quinazoline derivatives comes from the discovery of a series of
be clinically effective in relieving the symptoms associated anilinoquinazolines, previously described as competitive
with BPH by relaxing prostatic smooth muscle tone. More inhibitors at the ATP site of EGFR tyrosine kinase, which
recently, α1-AR antagonists were found to be useful in showed allosteric inhibition of fructose 1,6-bisphosphatase
increasing the frequency of spontaneous passage of the distal (F16Bpase). Selective inhibition of this enzyme was obtained
ureteral calculi [45]. However, this action alone does not fully through the addition of appropriate polar functional groups
account for the long-term clinical response to these drugs in at the quinazoline 2-position, thus separating the F16Bpase
BPH patients. Furthermore, the advances in understanding inhibitory activity from the EGFR tyrosine kinase activity
the molecular mechanism governing prostate cell proliferation previously observed. An F16Bpase inhibitor should reduce
have led to the identification of possible novel effects of hepatic glucose output and lower blood glucose by inhibiting
quinazoline derivatives against prostate cancer growth. Exper- the elevated rate of gluconeogenesis present in diabetic
imental clinical studies have provided new evidence to sup- patients and would, thus, be a useful therapeutic tool for the
port a differential growth-suppressing function of the α1-AR treatment of type 2 diabetes [52].
antagonists against prostate cancer, that is independent of an Finally, as recently as 2006, a patent reported the design
α1-AR mechanism [46]. The quinazoline-based antagonists 2 and synthesis of some 2-amino-3,4-dihydroquinazoline
and 3 suppress prostate growth by inducing apoptosis and derivatives that are useful in the treatment of Alzheimer’s
preventing cell invasion and migration of prostate tumour disease as enzyme β-secretase (BACE) inhibitors [103].
epithelial cells and vascular endothelial cells. Differently, tam-
sulosin, an α1-AR antagonist clinically effective for BPH 5. Expert opinion
treatment, fails to exert a similar apoptotic action, indicating
that this effect is mediated by the quinazoline nucleus [47]. The effort to design and synthesise agents selective for each
This and other evidence challenges conventional knowl- of the three α1-AR subtypes is still an active area of research
edge of the mechanism of action of α1-AR antagonists, and in medicinal chemistry. Due to a high percentage of amino
points to a new therapeutic value for these drugs by provid- acids that are identical in the active binding pocket of the
ing a differential molecular basis for their antitumour effi- different α1-AR subtypes and despite the impressive results
cacy. In 2005, it was found that compound 2 gives rise to an obtained by molecular biology studies, the availability of
apoptotic/anti-angiogenic effect in transitional cell selective ligands that are able to recognise only one among
carcinoma of the bladder, similar to the effect seen in the α1-AR subtypes, is limited. The picture is further com-
prostate cancer [48]. plicated as it is still unclear whether most of the so-called
A 2004 study demonstrated that three derivatives could competitive antagonists are neutral antagonists or inverse
be pharmacologically exploited to develop a novel class of agonists [53] and whether or not constitutively active native
anticancer agents that mediate apoptosis in PC-3 receptors have any physiological or pathological significance.
androgen-independent prostate cancer cells through the Recent experimental evidence suggests that a large number
inhibition of intracellular protein kinase B/Akt activation. of structurally different α1-AR antagonists, including quina-
The apoptosis-inducing activity of compound 3 was further zolines, are inverse agonists at both α1a- and α1b-AR
optimised by replacing the (2,3-dihydro-benzo[1,4]diox- subtypes (and this finding should explain the unwanted
ane)-carbonyl moiety with aryl-sulfonyl functions, thus effects in vivo on the cardiovascular system such as orthos-
highlighting the translational potential of these agents in tatic hypotension) [54]. The distinction of receptor antago-

404 Expert Opin. Drug Discov. (2006) 1(5)


Minarini, Bolognesi, Tumiatti & Melchiorre

nists as inverse agonists versus neutral antagonists will allow that is found in a broad variety of biologically active com-
a better interpretation of the pharmacological effects of clin- pounds that hit different pharmaceutical targets.
ically used drugs with respect to their mechanism of action. Therefore, it meets the requirements of a privileged struc-
Unfortunately, as spontaneous activity of receptors in vivo is ture that is, ‘a single molecular framework able to provide
difficult to assess, the therapeutic implications for the use of ligands for diverse receptors’ [55]. As it is difficult to reach
inverse agonists versus neutral antagonists remain a matter lead discovery or optimisation without combinatorial
of debate. methodologies, the solid-phase synthesis of a privileged
As detailed in the Section 4, it is well known that, structure or substructure based on quinazoline could pro-
besides the essential role played by the vide a way of generating numerous lead compounds for a
2,4-diamino-6,7-dimethoxy-quinazoline moiety for α1-AR variety of receptor and enzymatic targets, including the
antagonism, the quinazoline ring is a heterocyclic motif α1-AR [56].

Bibliography 10. LOWE FC: Role of the newer 17. ACHARI B, MANDAL S, DUTTA P,
Papers of special note have been highlighted as α-adrenergic-receptor antagonists in the CHOWDHURY C: Perspectives on
Downloaded by [Emory University] at 15:52 09 August 2015

either of interest (•) or of considerable interest treatment of benign prostatic 1,4-benzodioxins, 1,4-benzoxazines and
(••) to readers. hyperplasia-related lower urinary tract their 2,3-dihydro derivatives. Synlett. (2004)
symptoms. Clin. Ther. (2004) (14):2449-2467.
1. LANGER SZ: History and nomenclature of
26(11):1701-1713. CAMPBELL SF, HARDSTONE JD,
α1-adrenoceptors. Eur. Urol. (1999) 18.
36(Suppl. 1):2-6. 11. CAMPBELL SF, DAVEY MJ, PALMER MJ:
HARDSTONE JD, LEWIS BN, 2,4-Diamino-6,7-dimethoxyquinoline
2. HIEBLE JP: Adrenoceptor
PALMER MJ: derivatives as α-adrenoceptor antagonists
subclassification: an approach to improved
2,4-Diamino-6,7-dimethoxyquinazolines. and antihypertensive agents.
cardiovascular therapeutics.
1.2-[4-(1,4-benzodioxan-2-ylcarbonyl)piper J. Med. Chem. (1988) 31(5):1031-1035.
Pharm. Acta Helv. (2000) 74(2-3):163-171.
azin-1-yl] derivatives as α1-adrenoceptor 19. BORDNER J, CAMPBELL SF,
3. ZUSMAN RM: The role of α1-blockers in antagonists and antihypertensive agents. PALMER MJ, TUTE MS:
combination therapy for hypertension. Int. J. Med. Chem. (1987) 30(1):49-57. 1,3-Diamino-6,7-dimethoxyisoquinoline
J. Clin. Pract. (2000) 54(1):36-40.
12. BORDNER J, CAMPBELL SF, derivatives as potential α1-adrenoceptor
4. KAPLAN SA, KAPLAN NM: α-blockade: PALMER MJ, TUTE MS: antagonists. J. Med. Chem. (1988)
monotherapy for hypertension and benign 1,3-Diamino-6,7-dimethoxyisoquinoline 31(5):1036-1039.
prostatic hyperplasia. Urology (1996) derivatives as potential α1-adrenoceptor 20. GIARDINÀ D, BRASILI L, GREGORI M
48(4):541-550. antagonists. J. Med. Chem. (1988) et al.: Structure–activity relationships in
5. HIEBLE JP, RUFFOLO RR Jr: The use of 31(5):1036-1039. prazosin-related compounds. Effect of
α-adrenoceptor antagonists in the 13. CAMPBELL SF, HARDSTONE JD, replacing a piperazine ring with an
pharmacological management of benign PALMER MJ: alkanediamine moiety on
prostatic hypertrophy: an overview. 2,4-Diamino-6,7-dimethoxyquinoline α1-adrenoreceptor blocking activity.
Pharmacol. Res. (1996) 33(3):145-160. derivatives as α1-adrenoceptor antagonists J. Med. Chem. (1989) 32(1):50-55.
6. CHAPPLE CR: Pharmacological therapy of and antihypertensive agents. J. Med. Chem. 21. GIARDINÀ D, GULINI U, MASSI M
benign prostatic hyperplasia/lower urinary (1988) 31(5):1031-1035. et al.: Structure-activity relationships in
tract symptoms: an overview for the 14. BREMNER JB, COBAN B, GRIFFITH R, prazosin-related compounds. 2. Role of the
practising clinician. BJU Int. (2004) GROENEWOUD KM, YATES BF: piperazine ring on α-blocking activity.
94(5):738-744. Ligand design for α1 adrenoceptor subtype J. Med. Chem. (1993) 36(6):690-698.
7. ROEHRBORN CG, SCHWINN DA: selective antagonists. Bioorg. Med. Chem. 22. GIARDINÀ D, CRUCIANELLI M,
α1-adrenergic receptors and their inhibitors (2000) 8(1):201-214. ROMANELLI R, LEONARDI A,
in lower urinary tract symptoms and benign 15. BREMNER JB, GRIFFITH R, COBAN B: POGGESI E, MELCHIORRE C:
prostatic hyperplasia. J. Urol. (2004) Ligand design for α(1) adrenoceptors. Synthesis and biological profile of the
171(3):1029-1035. Curr. Med. Chem. (2001) 8(6):607-620. enantiomers of
8. AKDUMAN B, CRAWFORD ED: 16. ISHIGURO M, FUTABAYASHI Y, [4-(4-amino-6,7-dimethoxyquinazolin-2-yl)
Terazosin, doxazosin, and prazosin: current OHNUKI T, AHMED M, -cis-octahydroquinoxalin-1-yl]furan-2-ylme
clinical experience. Urology (2001) MURAMATSU I, NAGATOMO T: thanone (cyclazosin), a potent competitive
58(6 Suppl. 1):49-54. Identification of binding sites of prazosin, α1B- adrenoceptor antagonist.
9. MCVARY KT: Alfuzosin for symptomatic tamsulosin and KMD-3213 with J. Med. Chem. (1996) 39(23):4602-4607.
benign prostatic hyperplasia: long-term α1-adrenergic receptor subtypes by •• This paper describes the design and
experience. J. Urol. (2006) 175(1):35-42. molecular modeling. Life Sci. (2002) synthesis of one of the most selective
71(21):2531-2541. α1b-AR antagonists discovered so far.

Expert Opin. Drug Discov. (2006) 1(5) 405


Recent advances in the design and synthesis of prazosin derivatives

23. MELCHIORRE C, ANGELI P, subtypes. J. Med. Chem. (1998) 41. WILSON LJ: Traceless solid-phase synthesis
BOLOGNESI ML et al.: α1-adrenoreceptor 41(24):4844-4853. of 2,4-diaminoquinazolines. Org. Lett.
antagonists bearing a quinazoline or a 32. BOLOGNESI ML, MARUCCI G, (2001) 3(4):585-588.
benzodioxane moiety. Pharm. Acta Helv. ANGELI P et al.: Analogues of prazosin 42. WU ZD, KIM J, SOLL RM,
(2000) 74(2-3):181-190. that bear a benextramine-related polyamine DHANOA DS: Solid-phase synthesis of
24. LEONARDI A, MOTTA G, BOI C et al.: backbone exhibit different antagonism 2,4-diaminoquinazolines.
Synthesis, pharmacological evaluation, and toward α1-adrenoreceptor subtypes. Biotechnol. Bioeng. (2000) 71(2):87-90.
structure–activity relationship and J. Med. Chem. (2001) 44(3):362-371. 43. DENER JM, LEASE TG, NOVACK AR,
quantitative structure–activity relationship 33. FRUTTERO R, BOSCHI D, PLUNKETT MJ, HOCKER MD,
studies on novel derivatives of DI STILO A, GASCO A: The furoxan FANTAUZZI PP: Solid-phase synthesis of
2,4-diamino-6,7-dimethoxyquinazoline system as a useful tool for balancing 2,4-diaminoquinazoline libraries.
α1-adrenoceptor antagonists. ‘hybrids’ with mixed α1-antagonist and J. Comb. Chem. (2001) 3(6):590-597.
J. Med. Chem. (1999) 42(3):427-437. NO-like vasodilator activities. J. Med. 44. ANDRUS MB, METTATH SN, SONG C:
25. PATANE MA, SCOTT AL, BROTEN TP Chem. (1995) 38(25):4944-4949. A modified synthesis of iodoazidoaryl
et al.: 34. ANTONELLO A, HRELIA P, prazosin. J. Org. Chem. (2002)
4-Amino-2-[4-[1-(benzyloxycarbonyl)-2(S)- LEONARDI A et al.: Design, synthesis, and 67(23):8284-8286.
[[(1,1-dimethylethyl)amino]carbonyl]-piper biological evaluation of prazosin-related 45. YILMAZ E, BATISLAM E, BASAR MM,
azinyl]-6, 7-dimethoxyquinazoline
Downloaded by [Emory University] at 15:52 09 August 2015

derivatives as multipotent compounds. TUGLU D, FERHAT M, BASAR H: The


(L-765,314): a potent and selective α1b J. Med. Chem. (2005) 48(1):28-31. comparison and efficacy of 3 different
adrenergic receptor antagonist.
35. BENNING CM, KYPRIANOU N: α1-adrenergic blockers for distal ureteral
J. Med. Chem. (1998) 41(8):1205-1208.
Quinazoline-derived α1-adrenoceptor stones. J. Urol. (2005) 173(6):2010-2012.
26. MINARINI A, BUDRIESI R, antagonists induce prostate cancer cell 46. TAHMATZOPOULOS A,
CHIARINI A, LEONARDI A, apoptosis via an ROWLAND RG, KYPRIANOU N: The
MELCHIORRE C: Search for α1-adrenoceptor-independent action. role of α-blockers in the management of
α1-adrenoceptor subtypes selective Cancer. Res. (2002) 62(2):597-602. prostate cancer. Expert Opin. Pharmacother.
antagonists: design, synthesis and biological
36. CHUEH SC, CHERN JW, (2004) 5(6):1279-1285.
activity of cystazosin, an α1D-adrenoceptor
CHOONG CM, GUH JH, TENG CM: •• A thorough review on apoptotic activity of
antagonist. Bioorg. Med. Chem. Lett. (1998)
Characterization of some novel quinazoline-based α1-AR antagonists.
8(11):1353-1358.
α1-adrenoceptor antagonists in human 47. TAHMATZOPOULOS A,
27. GIARDINÀ D, SAGRATINI G, hyperplastic prostate. Eur. J. Pharmacol. KYPRIANOU N: Apoptotic impact of
GRELLI M, GULINI U, (2002) 445(1-2):125-131. α1-blockers on prostate cancer growth: a
MELCHIORRE C, ANTOLINI L:
37. ROMEO G, MATERIA L, SALERNO L, myth or an inviting reality? Prostate (2004)
Absolute configuration of the
RUSSO F, MINNEMAN KP: Novel 59(1):91-100.
α1B-adrenoceptor antagonist (+)-cyclazosin.
antagonists for α1-adrenoceptor subtypes. 48. TAHMATZOPOULOS A,
Farmaco (2004) 59(12):965-969.
Expert Opin. Ther. Patents (2004) LAGRANGE CA, ZENG L,
28. MARUCCI G, ANGELI P, BUCCIONI M 14(5):619-637. MITCHELL BL, CONNER WT,
et al.: (+)-Cyclazosin, a selective
38. ROSINI M, ANTONELLO A, KYPRIANOU N: Effect of terazosin on
α1B-adrenoceptor antagonist: functional
CAVALLI A et al.: Prazosin-related tissue vascularity and apoptosis in
evaluation in rat and rabbit tissues.
compounds. Effect of transforming the transitional cell carcinoma of bladder.
Eur. J. Pharmacol. (2005)
piperazinylquinazoline moiety into an Urology (2005) 65(5):1019-1023.
522(1-3):100-107.
aminomethyltetrahydroacridine system on 49. SHAW YJ, YANG YT, GARRISON JB,
29. GIARDINÀ D, POLIMANTI O, the affinity for α1-adrenoreceptors. KYPRIANOU N, CHEN CS:
SAGRATINI G et al.: Searching for J. Med. Chem. (2003) 46(23):4895-4903. Pharmacological exploitation of the
cyclazosin analogues as α1B-adrenoceptor
39. ISMAIL MA, ABOUL-ENEIN MN, alpha1-adrenoreceptor antagonist doxazosin
antagonists. Farmaco (2003)
ABOUZID KA, SERYA RA: Ligand design to develop a novel class of antitumor agents
58(7):477-487.
and synthesis of new that block intracellular protein kinase B/Akt
30. SAGRATINI G, BUCCIONI M, imidazo[5,1-b]quinazoline derivatives as activation. J. Med. Chem. (2004)
GULINI U et al.: (+)-Cyclazosin derivatives α1-adrenoceptor agonists and antagonists. 47(18):4453-4462.
as α1-adrenoceptor antagonists. Bioorg. Med. Chem. (2006) 14(4):898-910. 50. SHEINERMAN FB, GIRAUD E,
Med. Chem. Res. (2004) 13(3/4):190-199.
40. VOGTLE MM, MARZINZIK AL: LAOUI A: High affinity targets of protein
31. BOLOGNESI ML, BUDRIESI R, Synthetic approaches towards quinazolines, kinase inhibitors have similar residues at the
CHIARINI A, POGGESI E, quinazolinones and quinazolinediones on positions energetically important for
LEONARDI A, MELCHIORRE C: solid phase. Qsar Comb. Sci. (2004) binding. J. Mol. Biol. (2005)
Design, synthesis, and biological activity of 23(6):440-459. 352(5):1134-1156.
prazosin-related antagonists. Role of the •• A complete review on quinazolines, 51. NOBLE ME, ENDICOTT JA,
piperazine and furan units of prazosin on quinazolinones and quinazolinediones JOHNSON LN: Protein kinase inhibitors:
the selectivity for α1-adrenoreceptor solid-phase synthesis developed over the
last decade.

406 Expert Opin. Drug Discov. (2006) 1(5)


Minarini, Bolognesi, Tumiatti & Melchiorre

insights into drug design from structure. 55. EVANS BE, RITTLE KE, BOCK MG Patents
Science (2004) 303(5665):1800-1805. et al.: Methods for drug discovery:
52. WRIGHT SW, CARLO AA, CARTY MD development of potent, selective, orally 101. FHOFFMAN-LA ROCHE AG:

et al.: Anilinoquinazoline inhibitors of effective cholecystokinin antagonists. WO2005395 (2005).


fructose 1,6-bisphosphatase bind at a novel J. Med. Chem. (1988) 31(12):2235-2246. 102. PFIZER: US3511836 (1970).
allosteric site: synthesis, in vitro 56. KAMAL A, REDDY KL, DEVAIAH V, 103. JANSSEN PHARMACEUTICA NV:
characterization, and X-ray crystallography. SHANKARAIAH N, RAO MV: Recent WO2006024932 (2006).
J. Med. Chem. (2002) 45(18):3865-3877. advances in the solid-phase combinatorial
synthetic strategies for the quinoxaline,
53. MELCHIORRE C, BOLOGNESI ML, Affiliation
BUDRIESI R et al.: Search for selective quinazoline and benzimidazole based Anna Minarini†, Maria Laura Bolognesi,
antagonists a tα1-adrenoreceptors: neutral privileged structures. Mini Rev. Med. Chem. Vincenzo Tumiatti & Carlo Melchiorre
or negative antagonism? Farmaco (1998) (2006) 6(1):71-89. †Author for correspondence

53(4):278-286. • A very recent overview highlighting the Department of Pharmaceutical Sciences,


combinatorial synthesis for the generation University of Bologna, Via Belmeloro 6,
54. ROSSIER O, ABUIN L, FANELLI F,
of heterocycles-based privileged structures. 40126 Bologna, Italy
LEONARDI A, COTECCHIA S: Inverse
agonism and neutral antagonism at α(1a)- Tel: +39 0512099709; Fax: +39 0512099734;
and α(1b)-adrenergic receptor subtypes. E-mail: anna.minarini@unibo.it
Downloaded by [Emory University] at 15:52 09 August 2015

Mol. Pharmacol. (1999) 56(5):858-866.

Expert Opin. Drug Discov. (2006) 1(5) 407

You might also like