PHARMACOCHEMISTRY LIBRARY- VOLUME 24 PERSPECTIVES IN RECEPTOR RESEARCH
PHARMACOCHEMISTRY LIBRARY- VOLUME 24 PERSPECTIV...
7 downloads
673 Views
21MB Size
Report
This content was uploaded by our users and we assume good faith they have the permission to share this book. If you own the copyright to this book and it is wrongfully on our website, we offer a simple DMCA procedure to remove your content from our site. Start by pressing the button below!
Report copyright / DMCA form
PHARMACOCHEMISTRY LIBRARY- VOLUME 24 PERSPECTIVES IN RECEPTOR RESEARCH
PHARMACOCHEMISTRY LIBRARY- VOLUME 24 PERSPECTIVES IN RECEPTOR RESEARCH
PHARMACOCHEMISTRY LIBRARY, edited by H. Timmerman Other titles in this series Volume 10 QSAR in Drug Design and Toxicology, Proceedings of the Sixth European Symposium on Quantative Structure-Activity Relationships, Portoro~-Portorose (Yugoslavia), September 22-26, 1986 edited by D. Had~i and B. Jerman-Bla~_i~; Volume 11 Recent Advances in Receptor Chemistry. Proceedings of the Sixth CamerinoNoordwijkerhout Symposium, Camerino (Italy), September 6-10. 1987 edited by C. Melchiorre and Giannella Volume 12 Trends in Medicinal Chemistry '88. Proceedings of the Xth International Symposium on Medicinal Chemistry, Budapest, 15-19 August, 1988 edited by H. van der Groot, G. Domany, L. Pallos and H. Timmerman Volume 13 Trends in Drug Research. Proceedings of the Seventh Noordwijkerhout-Camerino Symposium, Noordwijkerhout (The Netherlands), 5-8 September, 1989 edited by V. Claassen Volume 14 Design of Anti-Aids Drugs edited by E. De Clerq Volume 15 Medicinal Chemistry of Steroids by F.J. Zeelen Volume 16 QSAR: Rational Approaches to the Design of Bioactive Compounds. Proceedings of the Eighth European Symposium on Quantitative Structure-Activity Relationships, Sorrento (Italy), 9-13 September, 1990 edited by C. Silipo and A. Vittoria Volume 17 Antilipidemic Drugs- Medicinal, Chemical and Biochemical Aspects edited by D.T. Witiak, H.A.I. Newman and D.R. Feller Volume 18 Trends in Receptor Research. Proceedings of the Eighth Camerino-Noordwijkerhout Symposium, Camerino (Italy), September 8-12, 1991 edited by P. Angeli, U. Giulini and W. Quaglia Volume 19 Small Peptides. Chemistry, Biology and Clinical Studies edited by A.S. Dutta Volume 20 Trends in Drug Research. Proceedings of the 9th Noordwijkerhout-Camerino Symposium, Noordwijkerhout (The Netherlands), 23-27 May, 1993 edited by V. Claassen Volume 21 Medicinal Chemistry of the Renin-Angiotesin System edited by P.B.M.W.M. Timmermans and R.R. Wexler Volume 22 The Chemistry and Pharmacology of Taxol| and its Derivatives edited by V. Farina Volume 23 Qsarand Drug Design: New Developments and Applications edited by T. Fujita
PHARMACOCHEMISTRY
LIBRARY
E d i t o r : H. T i m m e r m a n
Volume
24
PERSPECTIVES IN RECEPTOR RESEARCH Proceedings of the 10th Camerino-Noordwijkerhout Symposium, Camerino, Italy, 10-14 September 1995 Edited by"
DARIO GIARDIN~,
ALESSANDRO PIERGENTILI MARIA PIGINI Department of Chemical Sciences University of Camerino Via S. Agostino 1 62032 Camerino (MC), Italy
ELSEVIER Amsterdam - Lausanne - New York-
O x f o r d - S h a n n o n - T o k y o 1996
ELSEVIER SCIENCE B.V. P.O. Box 1527 1000 BM Amsterdam, The Netherlands
ISBN 0-444-82204-6 91996 Elsevier Science B.V. All rights reserved. No part of this publication may be reproduced, stored in a retrieval system or transmitted in any form or by any means, electronic, mechanical, photocopying, recording or otherwise, without the prior written permission of the publisher, Elsevier Science B.V., Copyright & Permissions Department, P.O.Box 521, 1000 AM Amsterdam, The Netherlands. Special regulations for readers in the USA- This publication has been registered with the Copyright Clearance Center Inc. (CCC), 222 Rosewood Drive, Danvers, MA 01923. Information can be obtained from the CCC about conditions under which photocopies of parts of this publication may be made in the USA. All other copyright questions, including photocopying outside of the USA, should be referred to the publisher. No responsibility is assumed by the publisher for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions or ideas contained in the material herein. This book is printed on acid-free paper. Printed in The Netherlands
PHARMACOCHEMISTRY LIBRARY ADVISORY BOARD r. Fujita
Department of Agricultural Chemistry, Kyoto University, Kyoto, Japan
E. Mutschler
Department of Pharmacology, University of Frankfurt, Frankfurt, F.R.G.
N.J. de Souza Research Centre, Hoechst India Ltd., Bombay, India D.T. Witiak
College of Pharmacy, The Ohio State University, Columbus, OH, U.S.A.
F.J. Zeelen
Organon Research Centre, Oss, The Netherlands
This Page Intentionally Left Blank
~
Vll
PREFACE This proceedings book contains the invited lectures delivered at the 10th CamerinoNoordwijkerhout Symposium, held in Camerino from the 10th to the 14th September 1995. In 1987, the Italian and Dutch scientific groups decided to join forces in organizing meetings, that were to become known as the Camerino-Noordwijkerhout Symposia, on specific aspects of Medicinal Chemistry. Since the beginning, in 1978, a particular feature of the Camerino Symposia has been that of reporting and debating the most recent knowledge and discoveries of chemists, pharmacologists and biologists involved in studying receptors and their mechanism of action. In this 1995 edition, too, an extremely intense exchange of information and opinions took place during the five days of the Symposium, when 27 invited lectures (16 plenary and 1 lmain ones), 8 oral communications, and 90 posters were presented and discussed in depth. Since that first 1978 meeting, Camerino has offered hospitality to 950 scientists, more and more of them intemational, as the proportion of foreign participants has risen from 12% to 60%. In the 1987 and 1991 editions two Nobel prize winners, Sir John Vane and Sir James Black, respectively, gave the opening lectures of the Symposia, and on three occasions, in 1987, 1991, and 1995, Honofis Causa degrees were conferred on Drs B. Belleau, E.J. Miens, and D.J. Tfiggle, respectively. During these 17 years, the existence of distinct receptofial subpopulations has been confirmed, something hypothesized at the 1978 Camerino Symposium by N.J.M. Birdsall, who during his paper "Muscarinic receptors: biochemical and binding studies" by himself, A.S.V. Burgen and E.C. Hulme, when analyzing data from their experiments, affu-med:
"These results are precisely the behaviour predicted for a heterogeneous population of binding sites" In addition, that first Symposium saw the setting up of a work relation between Nigel Birdsall and Rudolf Hammer that was subsequently to lead to the discovery of pirenzepine as a selective ligand for M1 muscarinic receptor subtype. As organizers of the 10th Symposium, we feel extremely gratified by how the exchange of opinion between the various lines of research in our meetings has opened the road to a deeper knowledge of the receptor as a biological target.
viii In conclusion, we hope that other meetings will continue to be held in Camerino, with the same high quality of scientific content and the same high values of friendly human relationships that have characterized all the past ones. The encouraging results obtained so far will thereby be carried even further forward.
Dario Giardin~t Alessandro Piergentili Maria Pigini
ix CONTENTS Giardin~t D, Piergentili A, Pigini M
Preface Triggle DJ Medicinal chemistry: through a glass darkly Klotz K-N, Jesaitis AJ, Lohse MJ
Mechanisms regulating G protein-coupled receptors
11
Shapiro G
Introduction to muscarinic receptors
27
Wess J, Blin N, Yun J, Sch6neberg T, Liu J
Structure-function analysis of muscarinic acetylcholine receptors
31
Mutschler E, Ensinger HA, Gross J, Leis A, Mendla K, Moser U, Pfaff O, Reichel D, Rtihlmann K, Tacke R, Waelbroeck M, Wehrle J, Lambrecht G
Muscarinic receptor subtypes - Search for selective agonists and antagonists
51
Casagrande C, Bertolini G
Perspectives in the design and application of dopamine receptor agonists
67
Branchek TA
Serotonin receptor complexity: relationships and roles
85
Becker DP, Goldstin B, Gullikson GW, Loeffler R, Moormann A, Moummi C, Nosal R, Spangler D, Villamil CI, Yang D-C, Zabrowski DL, Flynn DL
Design and synthesis of agonists and antagonists of the serotonin 5-HT4 receptor subtype Theroux TL, Esbenshade TA, Minneman KP oti-Adrenergic receptor subtypes and signal transduction
99
121
Leonardi A, Testa R, Motta G, De Benedetti PG, Hieble P, Giardinh D O~l-Adrenoceptors: subtype- and organ-selectivity of different agents
135
Jacobson KA, van Rhee AM, Siddiqi SM, Ji X-d, Jiang Q, Kim J, Kim HO
Molecular recognition in adenosine receptors
153
Cristalli G, Camaioni E, Di Francesco E, Vittori S, Volpini R
Chemical and pharmacological profile of selective adenosine receptor agonists
165
IJzerman AP, van der Wenden EM, Roelen HCPF, Math6t RAA, von Frijtag Drabbe Ktinzel JK
Partial agonists for adenosine receptors
181
Leurs R, Timmerman H
New histamine 1-13receptor ligands as pharmacological tools
193
Hibert M, Hoflack J, Trumpp-Kallmeyer S, Paquet J-L, Leppik R, Mouillac B, Chini B, Barberis C, Jard S
Three-dimensional structure of G protein-coupled receptors: from speculations to facts
205
Triggle DJ
Ion channels as targets for drug design
215
Evans JM
Recent advances in potassium channel activators
227
Lodge D, Bond A
Spinal glutamate receptors
241
Froestl W, Mickel SJ, Mondadori C, Olpe H-R, Pozza MF, Waldmeier PC, Bittiger H
GABAB receptor antagonists: new tools and potential new drugs
253
xi DiMaio J, Winocour P, Leblond L, Saifeddine M, Laniyonu A, Hollenberg MD Thrombin inhibitors and thrombin receptor agonists/antagonists
271
Rees DC Structure-activity relationships of non-peptide kappa-opioid analgesics:
a perspective of the last 10 years
291
Portoghese PS Selective nonpeptide ligands as probes to explore ~ opioid receptor
architecture
303
de Costa B R, He X-s, Dominguez C, Williams W, Rice KC, Bowen WD
The role of novel ligands in the biological characterization of sigma receptors
313
Melchiorre C, Angeli P, Bolognesi ML, Budriesi R, Cacciaguerra S, Chiarini A, Crucianelli M, Giardin~ D, Gulini U, Marucci G, Minarini A, Spampinato S, Tumiatti V
Tetraamines as lead compounds for the design of neurotransmitter receptor ligands: focus on o~-adrenergic and muscarinic receptors recognition
321
Lambrecht G, Ardanuy U, B~iumert HG, Bo X, Hoyle CHV, Nickel P, Pfaff O, Ralevic V, Windscheif U, Ziganshin AU, Ziyal R, Mutschler E, Burnstock G
Design and pharmacological characterization of selective Pepurinoceptor antagonists
337
Feldman J, Dontenwill M, Greney H, Bennai F, Bousquet P
Imidazoline receptors: an update
351
Brasili L, Pigini M, Bousquet P, Carotti A, Dontenwill M, Giannella M, Moriconi R, Piergentili A, Quaglia w, Tayebati SK
Discovery of highly selective imidazoline receptor ligands
361
xii Gaviraghi G, Cassar~ P, Corsi M, Curotto G, Donati D, Feriani A, Finch H, Finizia G, Pentassuglia G, Polinelli S, Ratti E, Reggiani A, Tarzia G, Tedesco G, Tranquillini ME, Trist DG, Ursini A
Synthesis and structure-activity relationship of new 1,5-benzodiazepine CCK-B antagonists
375
Author index
389
Subject index
393
Perspective in Receptor Research D. Giardinh, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved. MEDICINAL
CHEMISTRY:
THROUGH
A GLASS
DARKLY
David J. Triggle, State University of New York, Buffalo, New York, USA INTRODUCTION
0 brave new world, That has such people in't! William Shakespeare,
The Tempest.
The patenting of a mouse predisposed to cancer and genetically engineered by Philip Leder and Timothy Stewart at Harvard University in April 1988 generated important scientific, commercial and ethical questions. Some five years later a selfimposed moratorium has ended and the United States Patent and Trademark Office is again issuing patents to a variety of genetically engineered animals, including those that exhibit prostate enlargement, develop insulin-resistant diabetes and produce human proteins including antibodies ( i ) . There is little question but that these transgenic species will accelerate progress towards treatment of a variety of human disorders. Similarly, the production of transgenic plants, whilst generating less immediate ethical attention, will prove of value to the generation of medicinal products of natural origin such as scopolamine through the introduction of the hyoscyamine hydroxylase gene into Atropa belladonna (2). During the same time period there has been a similar increase in human gene therapy (3). Some thirty-seven gene transfer trials have been approved worldwide and eighteen of these are designed to produce immediate therapeutic benefits. These include several trials inserting the gene for adenosine deaminase to alleviate the absence of a natural immune system and interleukin genes for several cancer types (4,5). With continued progress in overcoming the major problems of gene therapy including low gene transfer efficiency, poor cell targeting and premature gene turnoff it is clear that the horizons for human gene therapy will continue to expand. The potential benefits of alleviating dopamine deficiency in the brains of Parkinson's patients by tissue implantation have been widely discussed from both clinical and ethical perspectives. Conventional dopamine replacement therapy suffers from well known and serious limitations. The most recent data indicate that fetal-tissue transplants into patients with idiopathic or MPTP-induced Parkinson's disease are clinically encouraging and may be highly beneficial for at least some patients (6,7). Do these and similar developments in gene and tissue therapy signal the end of medicinal chemistry and the arrival at a solely biotechnology-oriented therapy mall where patients shop for both designer jeans and genes? Or, and more plausibly perhaps, will the molecular and medical sciences continue to adjust to a clinical interface that is positioned by science, economics and ethics?. It is clear that the issues of ethics raised by our rapid approach to the human genome will be of increasing importance to defining new clinical interfaces (8).
THE
HXSTORY
heavenly messengers bringing evil reports of armies on the move and time r u n n i n g short and famines and earthquakes and train wrecks and the tearing down of the wall bob dylan
The search for power and the explanation of natural events led naturally to the attribution of specific properties to plants and to their extracts. These properties were used in murder, magic and medicine, processes inextricably linked in man's history (9). Predictive, perhaps, of contemporary issues drugs were used also for their recreational properties; r e c e n t studies of Egyptian mummies have revealed the presence of cocaine, nicotine and hashish (I0). Frequently, specific features of the plant were associated with and predictive of these properties. Thus the mandrake [ Mandragora o f f i c i n a r u m ] was associated with the human form and special techniques were believed to be necessary in the collection of the plant to avoid madness or death. Those hominids partaking of Aminata m u s c a r i a would have been exposed to both the toxic and h a l l u c i n o g e n i c effects of the muscarine, muscimol and ibotenic acid c o n t a i n e d in this fungus. Aminata may have been the source of the soma of Vedic India and the inspiration for the soma of Aldous H u x l e y ' s Brave New World which had "all of the advantages of C h r i s t i a n i t y and alcohol; none of their defects". One of the plagues of Egypt may well have been caused by the proliferation of d i n o f l a g e l l a t e s causing a red tide, fish killing and water p o i s o n i n g (II). The history of hashish dates back to at least 2000 BC and is associated with a considerable folk-lore. The word "assassin" derives from the Arabic for "hashish-eater" and the i n t r o d u c t i o n of hashish to Europe by Marco Polo initiated investigations that have culminated in the identification of a specific neural receptor for the active constituent c a n n a b i n o i d s of m a r i j u a n a and in the isolation of an endogenous ligand, anandamide, for this receptor (12). The concept of specific receptors and sites of drug action has many origins. However, the work of Thomas Fraser and A l e x a n d e r Crum Brown of Edinburgh University described in their p u b l i c a t i o n of 1869, "On The Connection Between Chemical C o n s t i t u t i o n and Physiological Action", provides both a d e f i n i t i o n of structure-activity relationships and a p i o n e e r i n g example of chemist-pharmacologist collaboration (13). The definition of receptors as specific sites for drug action owes much to the work of John Newton Langley [ 18521925 ] and Paul Ehrlich [ 1854-1915 ]. Their separate w o r k on the autonomic nervous system and toxins and c h e m o t h e r a p e u t i c agents led to the concept of a receptor that possesses both r e c o g n i t i o n and transduction components and of c h e m o t h e r a p e u t i c m o l e c u l e s possessing discrete molecular features s u b s e r v i n g specific functions:
"It is convenient to have a term for the specially excitable constituent and I have called it the r e c e p t i v e substance. It r e c e i v e s the stimulus [ recognition ] and by transmitting it [ transduction ] causes contraction [ response ]" John Newton Langley, 1906
"---they must p o s s e s s a certain definite grouping which is c h e m i c a l l y a l l i e d to one of the c h e m o r e c e p t o r s of the p a r a s i t e . - - - T h e r e must, therefore, be in addition to the f i x i n g g r o u p a n o t h e r which brings about the d e s t r u c t i o n -is to be c h a r a c t e r i z e d as the toxophoric" Paul Ehrlich, 1913
These predictions have been admirably fulfilled by the isolation and characterization of receptors themselves and by the assignment within receptors and individual molecules of domains responsible for such specific functions; these include ligand binding, voltage sensitivity and effector coupling in ion channels and detection, initiation and delivery in the DNA cleaving enediynes (14,15). Although he died during World War 1 Ehrlich was an optimist and writing some two years before his death: "Now that the l i a b i l i t y to, and danger of, disease are to a large extent circumscribed--the effects of c h e m o t h e r a p e u t i c s are directed as far as possible to fill up the gaps left in this ring".
Ehrlich had not, however, contemplated drug resistance, including the recent appearance of drug-resistant tuberculosis (16,17), nor presumably had he contemplated the dual impact of mass migrations and habitat destruction on the emergence of new and lethal infections including Lassa rift valley, Ross River, Crimean-Congo and HI viruses (18). THE P R E S E N T "The a c h i e v e m e n t s of chemical synthesis are firmly b o u n d to our a t t e m p t to break the shackles of disease and poverty" Roald Hoffmann, 1993.
The emergence of both old and new diseases focuses continuing efforts on drug discovery and the central role of medicinal chemistry. These foci are switching increasingly from the empirical and intuitive discovery processes to structureand mechanism-based discovery routes. These changes are coupled to the emergence of new targets and roles for medicinal chemistry that are increasingly at an interface with molecular biology. New cellular messengers with unconventional properties continue to define new targets. Nitric oxide, or a labile derived species, is now established to mediate physiological and pathological events in cardiovascular, secretory and neuronal cells (19-21). The recent postulate that CO may also be a messenger species (22,23) serving, with nitric oxide, a common guanylyl cyclase effector defines new signalling pathways that link space and time to coordinate the activities and functions of cell populations (24). A major role for such species can be envisaged in the regulation of the brain and its multiple neuronal populations. The modulation of the activities of such novel species presents both major opportunities and major problems of site specificity and delivery. The critical role of calcium as a cellular regulator has been recognized for over a century (25). Cyclic adenosine
diphosphate ribose [ cADP-ribose ] is the latest messenger serving to modulate the release of intracellular Ca ~T and perhaps representing the endogenous ligand for the ryanodinesensitive Ca 2+ release channel (26,27). Not only is this m e s s e n g e r an important link between nutrient levels and insulin release in pancreatic beta-cells, b ~ it may serve quite generally to link nutrients with Ca fluxes. The availability of both cADP ribwse and inositol phosphate pathways to control intracellular Ca ~T relwase provides a parallel to the multiple entry processes for Ca ~T during cell excitation and provides an additional base for the spatial and temporal control of this messenger (28). Additionally, this discovery raises again the issue of endogenous ligands for other types of Ca 2+ channels (29). New targets demand new ligands and more subtle definition of the ligand-target interactions. Target definition by X-ray and NMR as structure-based drug design promises to become a predictive component of the discovery process (30). The design of two-fold symmetric inhibitors of HIV protease followed directly from the discovery that the retroviral enzymes are heterodimers (31-33). Other examples are likely to follow for other soluble proteins. Membrane-bound proteins constitute, however, particular problems for both X-ray and N M R m e t h o d s , but the high resolution electron microscopic techniques applied to two-dimensional membrane sheets including bacteriorhodopsin and porin may well be useful for other systems (34,35). The d e t e r m i n a t i o n of the structure of the human growth hormone complexed with the soluble extracellular domain of its receptor suggests an alternative strategy for membrane proteins (36). Additionally, extension to ligand interactions with other m a c r o m o l e c u l e s including DNA as in the glucocorticoid-DNA complex is occurring (37). Gene isolation has been a major force in the characterization, expression and screening of pharmacological receptors. Two lessons learned are that pharmacological specificity may depend upon a single amino acid residue and that for receptors composed of subunits only a limited number of permutations may exist. The antagonist specificity of the rodent 5-HT.IB receptor is determined by a single residue [ asparaglne-355 ] from that of a human brain receptor [ threonine-355 ] with overall sequence homology, although both receptors bind 5-HT equally well (38). The benzodiazepinesensitive GABA A receptor, composed of multiple alpha, beta, gamma and delta subunits derived from some 15 genes and with several hundred thousand possible permutations, actually exists in very restricted distribution and combinations (39,40). The generation of lead structures and their optimization constitutes a central theme of medicinal chemistry. This theme places continuing demands on synthetic chemistry including the specification of new catalytic processes and consideration of chiral interactions (41,42). Increasingly, these demands will interface with biological processes including catalytic antibodies for disfavored chemical reactions (43) and chiral control through enzymatic and biomimetic processes (41). The mimicking of the biological world by chemistry has become extremely productive. The award of the 1987 Nobel Prize in Chemistry to Cram, Lehn and Pedersen was an explicit acknowledgment of the role of molecular recognition processes and of the analogies between host-guest chemistry and ligandreceptor interactions (45,46). Thus, a simple A4B6
cyclooligomer of trimesic acid and R,R-diaminocyclohexane exhibits very high chirality and side-chain specificity in peptide recognition (47). In turn, chemistry is expanding the synthetic capabilities of biological systems. The "natural" biological code specifies only 20 amino acids. Expansion of this code is possible through the creation of new codonanticodon pairs that specify novel residues (48). The nonstandard pair (iso-C)AG:(iso-dG)CU specifies L-iodotyrosine and indicates the possibilities of a semi-synthetic genetic code. A general method for incorporation of unnatural amino acids into proteins derives from replacement of a specific codon by the "blank" nonsense codon TAG through oligonucleotide-directed mutagenesis and use of a suppressor tRNA directed against this codon that has been aminoacylated by the amino acid residue of interest (49). Although ligand design may ultimately be completely rational and predictable from target structures, iterative processes of design and selection will continue to be of major significance despite its traditional heavy consumption of time and money. It is of increasing importance to devise methods that facilitate lead generation and selection A variety of combinatorial techniques are now available that generate libraries of compounds from amino acids or nucleotides. These range from extensions of peptide resin synthesis to the use of fusion phage which express on their surface a single peptide encoded by a randomly mutated region of the phage genome (5054). Affinity-mediated selection followed by physical or sequence-based localization permits the characterization of a lead structure. A combination of these processes entitled "encoded combinatorial chemistry" has been proposed in a theoretical treatment by Brenner and Lerner to consist of parallel syntheses where each monomeric chemical unit is encoded by a nucleotide sequence (55). Active molecules are affinityselected and identity and amplification achieved through the PCR applied to the retrogenetic tag. In principle, this method is not restricted to amino acid or nucleotide precursors and any set of building blocks could be employable. Such molecular construction kits are now becoming available and some are termed, appropriately enough, "Molecular Meccano" (56-58). A final component to the process of ligand selection may be accomplished by closing the circle of variation/mutation, selection and amplification/replication to provide evolutiondirected ligand selection (59-61). THE FUTURE
give me back my broken night my secret room my secret life its lonely here leonard cohen
Advances in synthetic methodologies notwithstanding, the future of medicinal chemistry will make increasing demands on chiral control, on peptidomimetics and on target- and sitespecific delivery. Exploration of specific cellular receptors as surrogate entry loci for peptides and proteins offers significant opportunity for target-specific delivery particularly if combined with mechanisms to selectively enhance the expression
of delivery receptors. Antibodies to the transferrin receptors can cross the blood-brain barrier by internalization through these receptors localized in brain capillary endothelial cells. Antibodies conjugated to nerve growth factor deliver the latter to neurons and enhance the survival of cholinergic and noncholinergic neurons transplanted into the eye (62). Similar techniques may be used to accomplish receptormediated gene delivery (63,64). Gene transfer technology is exploitable to create both target- and site-specific delivery systems. Retroviral vectors may be particularly and selectively useful because of their ability to transfer genes only into dividing cells. Thus, murine fibroblasts expressing a retroviral vector containing the herpes simplex thymidine kinase gene transduced proliferating rat glioma cells. Addition of the anti-herpes drug ganciclovir regressed the gliomas since these cells only expressed the herpes enzyme (65). The goal of peptide mimicry is the discovery of structural leads, or the development of strategies for molecular design, which result in bioavailable peptide analogs (66). The development of a strategy is awaited and most active compounds derive from lead identification and optimization. However, "retrofitting" of such leads as the benzodiazepine nucleus found in ligands for opiate, CCK, gastrin and glutamate receptors (7671) and the 1,4-dihydropyridine structure as a component of drugs active at several voltage-gated ion channels (72) is likely to be productive. The importance of chirality to the drug-receptor interaction has been recognized since at least the work of Cushney (73). An increasing need to consider chirality as a theme in all aspects of drug discovery from synthesis and manufacture to pharmacodynamics, pharmacokinetics and toxicology derives from the new FDA guidelines (74-77). Indeed, since 1985 almost 50% of the optically active molecules developed as drugs have been developed as active isomers and there are probably some 50-60 currently marketed racemates that are prime candidates for chiral switching (76,78). Fortunately, perhaps, for the pharmaceutical industry m o l ~ u l e s such as palytoxin with 64 dissymmetric carbon atoms and i0 isomers are rare (79). However, even simple chiral molecules may present complex pharmacology because of opposing chiral effects of pharmacokinetic and pharmacodynamic factors. This may be particularly so for drugs active at ion channels where statedependent interactions may modify both quantitatively and qualitatively the expression of drug action (80,81) Thus the stereoselectivity of local anesthetic interactions at Na + ' channels varies according to the frequency of stimulus and the activation mode (82,83). At the synthetic level we may expect increased emphasis on enantioselective catalysis using metal complex catalysts as a key component of the manufacturing process (84). For biocatalysts there will unquestionably continue to be increasing interest in the "custom synthesis" of enzymes engineered for specific functions and conditions. The first example of the "ultimate" enzyme has been reported with the synthesis of the all-D form of HIV-I protease (85-87). This enzyme exhibits a chiral specificity opposite to that of the naturally occurring L form and it may be generally predicted that enantiomeric proteins will exhibit reciprocal chiral specificity in all aspects of their interactions. These reciprocal chiral
s p e c i f i c i t i e s of D - a n d L - p r o t e i n s are of c o n s i d e r a b l e interest; they s u g g e s t t h a t in worlds a l t e r n a t i v e to ours their FDA will be the m l r r o r image of ours. On the other hand, .......... ACKNOWLEDGMENTS.
This w o r k was p r e s e n t e d in part as a l e c t u r e d e l i v e r e d to The XII I n t e r n a t i o n a l C o n g r e s s on M e d i c i n a l C h e m i s t r y in Basle, S e p t e m b e r 1992. It was o r i g i n a l l y p u b l i s h e d in " A n n u a l R e p o r t s In M e d i c i n a l Chemistry", V o l u m e 28, 1993 and is r e p r i n t e d h e r e in its e n t i r e t y and w i t h o u t change w i t h the kind p e r m i s s i o n of A c a d e m i c Press Inc, Orlando, Florida.
REFERENCES
1 2
3 4 5 6 7 8 9 i0 ii 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29
30
N e w Y o r k Times, Wed. Feb. 3, 1993, page AI. Yun SJ, H a s h i m o t o T, Yamada Y. Proc Nat A c a d Sci USA, 1992: 98: 11799. T h o m p s o n L. S c i e n c e 1992: 258: 744. M i l l e r AD. N a t u r e 1992: 455: 357. R o e m e r K, F r i e d m a n n T. Eur J B i o c h e m 1992:327: 1589. Fahn S. N e w Eng J Med 1992: 327: 1591. K a s s i r e r JP, A n g e l i M. New Eng J Med 1992: 327- 1591. H o o d L, K e u l e s DJ. [Editors] In: The Code of Codes, H a r v a r d U n i v e r s i t y Press, B o s t o n MA, 1992. M a n n J. M a g i c and Medicine, O x f o r d U n i v e r s i t y Press, O x f o r d UK, 1992. B a l a n o v a S, P a r s c h e F, Pirsig W. N a t u r w i s s 1992: 79: 358. Exodus, 7: 20,21. D e v a n e WA, H a n u s L, Brewer A, P e r t w e e RG, S t e v e n s o n LA, G r i f f i n G, G i b s o n D, M a n d e l b a u m A, E t i n g e r A, M e c h o u l a n R. S c i e n c e 1992: 258- 1946. F r a s e r T, C r u m B r o w n A. On The c o n n e c t i o n B e t w e e n C h e m i c a l C o n s t i t u t i o n and P h y s i o l o g i c a l Action. N e i l l e and Co. E d i n b u r g h , 1869. C a t t e r a l l WA. S c i e n c e 1991" 253: 1499. N i c o l a o u KC, Dai W-M, Tsay SC, Estevez VA, W r a s i d l o W. S c i e n c e 1992: 256: 1172. Anon, N a t u r e 1992: 359: 657. B l o o m BR. N a t u r e 1992: 348: 538. B e n f o r d G. Proc Nat A c a d Sci USA 1992: 89: 11098. N a t h a n C. FASEB J 1992: 6: 3051. S t a m l e r JS, Singel DJ, L o s c a l z o J. S c i e n c e 1992: 258: 1898. E d i t o r i a l , S c i e n c e 1992- 258- 1862. B a r i n o g a M. S c i e n c e 1993- 259: 309. V e r m a A, H i r s c h DJ, Glatt CE, R o n n e t t GV, Snyder SH. S c i e n c e 1993: 381: 259. E d e l m a n GM, G a l l y JA. Proc Nat A c a d Sci USA 1992: 89: 11651. T r i g g l e DJ. I n : A b r a h a m S and A m i t a i G, eds. C a l c i u m C h a n n e l M o d u l a t o r s in H e a r t and Smooth Muscle. VCH Weinheim, Germany, 1990; 1-13. G a l i o n e A. S c i e n c e 1993; 259: 325. T a k e s a w a S, Nata K, Y o n e k u r a H and K a m o t o HO. S c i e n c e 1993; 259: 370. B e r r i d g e MJ. N a t u r e 1993; 361- 315. T r i g g l e DJ. In: M o r a d M, Nayler WG, Kazda S and S c h r a m m M, eds. The C a l c i u m channel: Structure, F u n c t i o n and Implications. Springer-Verlag, B e r l i n and H e i d e l b e r g . 1988; 549-563. E r i c k s o n JW and Fesik SW. Ann Rep Med Chem 1992; 27"" 271.
31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56
57 58 59 60 61 62 63 64
E r i c k s o n J, N i e d h a r t DJ, Vandrie J, Kempf DJ, Wang XC, N o r b e c k DW, P l a t t n e r JJ, Rittenhouse JW, Turon M, W i d e b u r g N, K o h l b r e n n e r WE, Simmer R, Helfrich R, Paul DA and K n i g g e M. Science 1990; 249: 527. Des Jarlais RL, Seibel GL, Kuntz ID, Furthy PS, Alvarez JC, Ortiz de M o n t e l l a n o PR, DeCamp DL, Babe LM, Craik CS. Proc Nat A c a d Sci USA 1990; 87: 6644. Bone R, Vacca JP, Anderson PS, Holloway MK. J Med Chem 1991; 113: 9382. H e n d e r s o n R, B a l d w i n JM, Ceska JA. J Mol Biol 1991; 213: 899. W e i s s M, Abele U, Weckesser J, Weito M, Schultz E, Schultz GE. Science 1991; 254: 1627. De Vos AM, U l t s c h M, Kossiakoff AA. Science 1992; 225: 306. Lusi BF, Xu WX, Otwinowski Z, Freedman LP, Yamomoto KR, S i g l e r PB. Nature 1991; 352: 497. O k s e n b e r g D, M a r s t e r s SA, O'Dowd BF, Jin H, Havlik S, P e r o u t k a SJ, Ashkenazi A. Nature 1992; 360: 161. W i s d e n W, Laurie DJ, Monger H, Seeburg PH. J Neurosci 1992; 12: 1040. D e L o r e y TM, Olsen RW. J Biol Chem 1992; 267- 16747. S e e b a c h D. A n g e w Chem Ed Int 1990; 29; 1320. H e n d e r i c k s o n JB. Angew Chem Ed Int 1990; 29: 1286. Janda KD, Sevlin CG, Lerner RA. Science 1993; 259- 490. R e b e k J. A n g e w Chem 1990; 29: 245. Cram DJ. Science 1986; 240: 760. Lehn JM. A n g e w Chem Ed Int 1990; 29: 1304. Yoon SS, Still WC. J Amer Chem Soc 1993; 115: 823. Bain JD, Switzel C, Chamberlain AR, Benner SA. Nature 1992; 356: 537. N o r e n CJ, A n t h o n y - C a h i l l SJ, Griffith MC, Schultz PG. S c i e n c e 1992; 244: 182. Jung G, B e c k - S i c k i n g e r AG. Angew Chem Ed Int 1992; 31: 367. H o u g h t e n RA. Proc Nat Acad Sci USA 195; 82: 5131. Kam KS, Almon SE, Hersh EM, Hruby VJ, Kazmierski WM, K n a p p RJ. Nature 1991; 354: 82. Scot JK, Smith GP. Science 1990; 249: 386. D e v l i n JJ, P a n g a n i b a n LC, Devlin PE. Science 1990; 249: 404. B r e n n e r S, Lerner RA. Proc Nat Acad Sci USA 1992; 89: 5381. A n e l l i PL, A s h t o n PR, Ballardini R, Balzani V, Delgado M, G a n d o l f i MT, G o o d n o w TT, Kaifer AE, Philp D, P i e t r a s z k i e w i i c z M, Prodi L, Reddington MV, Slawin AMZ, S p e n c e r N, Stoddart F, ~ Vicent C, Williams DJ. J Amer Chem Soc 1992; 114: 193. Yang X, Jiang J, Knobler CB, Hawthorne MF. J Amer Chem Soc 1992; 114: 9719. M u l l e r J, Base K, Magnera TF, Michi J. J Amer Chem Soc 1992; 114: 9721. B e a u d r y AA, Joyce GF. Science 1992; 257: 635. Joyce GF. Sci Amer 1990; 90: Dec. L e h m a n N, Joyce GF. Nature 1993; 361: 182. F r i d e n PM, Walus LR, Watson P, Doctrow SR, K o z a r i c h JW, B a c k m a n C, B e r g m a n H, Hoffer B, Bloom F, Granholm AC. S c i e n c e 1993; 259: 373. Cotten M, Wagner E, Zatloukal K, Phillips S, Curieland DT, B i r n s t i e l ML. Proc Nat Acad Sci USA 1992; 89: 6094. W a g n e r E, Zatloukal K, Cotten M, Kirlappos H, M e c h t l e r K, Curiel DT, Birnstiel ML. Proc Nat Acad Sci USA 1992; 89: 60999.
65 66 67 68 69 70 71 72 73 74 75 76 77 78 79
80 81 82 83 84 85 86 87
Culver KW, Ram Z, Wallbridge S, Ishii H, Oldfield EH, Blaese RM. Science 1992; 256- 1550. Morgan BA, Gainer JA. Ann Rep Med Chem 1989; 24: 243. Evans BE, Rittle KE, Bock MG, DiPardo RM, Freidinger RM, Whitter WL, Lundell GF, Veber DF, Anderson PS, Chagi RSL, Lotti VJ, Cerino DJ, Chen TB, Kling PJ, Kunkel KA, Springer JP, Hirshfield J. J Med Chem 1988; 31: 2235. Bock MG, Dipardo RM, Evans BE, Rittle KE, Whitter WL, Veber DF, Anderson PS, Friedinger KM. J Med Chem 1989;32: 13. Romer D, Buscher HH, Hill RC, Maurer R, Pechter TJ, Zeuger H, Benson W, Finner E, Milkowski W, Thies PW. Life Sciences 1982; 31: 1217. Zorumski CF, Yamada KA, Price MT, Olney JW. Neuron 1993; I0: 61. Pincus MR, Charty RP, Chen J, Lubowski J, Avitable M, Shah D, Scheraga HA, Murphy RB. Proc Nat Aad Sci USA 1987; 844821. Rampe D, Triggle DJ. Prog Drug Res (in press) Cushny AR, Biological Relations of Optically Isomeric Substances. Balliere, Tyndall and Cox, London 1926. Gross M. Ann Rep Med Chem 1989; 25: 323. Announcement. Chirality 1992; 4" 338. Cayen MN. Chirality 1991; 3: 94. Hodgson J. Biotechnology 1992; i0- 1093. Stinson SC. Chem Eng News 1992; 46: Sep. 28. Armstrong RW, Beau JM, Cheon SH, Christ WJ, Fujioka H, Ham WH, Kawkins LD, Jin H, Kang SH, Kishi Y, Martinelli MJ, McWhorter WW, Mzuno M, Nakata M, Stutz AE, Talaas FX, Taniguchi M, Tino JA, Ueda K, Uensiishi JI, White JB, Yonaga M. J Amer Chem Soc 1989; Iii: 7525. Kwon YW, Triggle DJ. Chirality 1991; 3: 393. Crossley R. Tetrahedron 1992; 48: 8155. Yeh JA, In: Fink BR, ed. Molecular Mechanisms of Anesthesia, Raven Press, 1980; 2: 35-44. Wang GK, Wang SY. J Gen Physiol 1992; i00: 1003. Nugent WA, RajanBabu TV, Burk MJ. Science 1993; 259: 479. del Milton RC, Milton Sc, Kent SBH. Science 1992; 256: 1445. Petsko GA. Science 1992; 256- 1403. Jung G. Angew Chem Ed Int 1992; 3 1 : 1 4 5 7
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardina, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
11
Mechanisms regulating G protein-coupled receptors K.-N. Klotz 1, A.J. Jesaitis 2 and M.J. Lohse I l Institut fCir Pharmakologie und Toxikologie der Universit/it W~irzburg, Versbacher Str. 9, D-97078 W/irzburg, Germany 2Department of Microbiology, Montana State University, Bozeman, MT 59717, U.S.A. INTRODUCFION Receptor-mediated signal transduction plays a pivotal role in cell communication and is essential for a coordinate functioning of the organs of our body. Several classes of receptors can be distinguished on the basis of functional and structural differences. The still growing family of G protein-coupled receptors has been investigted in great detail and a wealth of structural and functional information is available for many members of this family. It has long been recognized that receptor-mediated signal transduction is a highly regulated process but only recently have we begun to understand some of the regulatory events that modulate signal intensity resulting from a given stimulus. In particular different mechanisms of desensitization for short-term as well as for long-term modulation of receptor function have been investigated in great detail (for review see [11). Several receptors including the ~-adrenergic receptor, rhodopsin, muscarinic receptors and formylpeptide receptors served as prototypes to study various forms of desensitization [2-5]. In this overview we will focus on the growing number of components of the [3-adrenergic receptor system that are implicated in modulation of signal transduction. In addition, we will present a mechanism for desensitization of N-formylmethionyl peptide receptor (FPR) which might be specific for regulation of chemoattractant receptors. This mechanism might serve as a model for the aspect of spatial organization of signal transduction that appears to be required for an effective interplay of different partners in this multi-component system.
COMPONENTS OF THE ~-ADRENERGICRECEPTORSYSTEM The ~-adrenergic receptor system comprises the main membrane-bound components of transmembrane signaling, which are the receptors themselves, the G-proteins G s, and adenylyl cyclases. In addition, the receptor system contains several cytosolic proteins which regulate the signaling process (Figure 1). The most important ones of these are the [~-adrenergic receptor kinases and the
12 ~-arrestins, which inhibit receptor/G-coupling, and phosducin, which has been proposed to function as an inhibitor of G-protein activation. Protein kinase A is the effector kinase of the system which modulates cellular functions by phosphorylating key proteins of multiple cellular processes. In addition, it has feedback functions at the level of receptor/G-coupling and of phosducin/G-coupling. COMPONENTS OF THE B-ADRENERGIC RECEPTOR SYSTEM agonist
0~S ::
Figure 1. Components of the s receptor system. The abbreviations denote: ~AR, ~-adrenergic receptor; ~ARK, ~-adrenergic receptor kinase; AC, adenylyl cyclase; PKA, protein kinase A; PhD, phosdudn. For all of these components, several isoforms have been described. The potential specificities of all of these isoforms are as yet ill-defined and do not appear to play a major in the principles of the regulatory mechanisms discussed here. The function of these cytosolic components of the ~-adrenergic receptor system is the tuning of the signaling process. Signalling via ~ - a d r e n e r g i c receptors is subject to a variety of regulatory mechanisms that can affect both the number and the function of these receptors. Most importantly, repeated or prolonged stimulation of the receptors results in a reduction of responsiveness, which is called agonist-induced receptor desensitization (reviewed in [1, 2]). Receptor desensitization involves several molecular mechanisms, which may be operative to varying extents in different cells and different receptor systems. These desensitization mechanisms can be broadly classified into those that alter receptor function and those that affect receptor expression.
13 Receptor expression is regulated over many hours, and changes are usually not visible before =15 min. Agonist-induced changes in receptor expression involve the degradation of the receptor protein as well as reduced receptor synthesis due to reductions in its mRNA levels. In contrast, receptor function can be altered within seconds. It occurs primarily by uncoupling receptors from their G-protein, G s.
DESENSITIZATION ~-ARRESTINS
BY ~-ADRENERGIC
RECEPTOR KINASES
AND
The most specific and rapid mechanism desensitizing [3-adrenergic receptors is triggered via their phosphorylation by [3-adrenergic receptor kinases ([3ARK). This process is initiated by agonist-activation of the receptors and proceeds in two steps: first, the receptors are phosphorylated by [3ARK in an agonist-dependent manner [6, 7]; subsequently, the [3-arrestins bind to the phosphorylated receptors, resulting in inhibition of receptor/Gs-coupling [8-10]. Depending on the cell type this mechanism occurs within seconds after agonist exposure and is complete in less than a minute [11]. The cDNAs of six ~ARK-like kinases, which are more generally called G protein-coupled receptor kinases, have been cloned (reviewed in [12]). However, only two have been shown to be involved in the regulation of ~-adrenergic receptor function, even though all six appear to be capable of phosphorylating the receptors in vitro. These two are termed [3-adrenergic receptor kinase-1 and 2. Both isoforms of [3ARK are cytosolic proteins which need to be translocated to the cell membrane in order to phosphorylate the receptors. This translocation appears to occur via three "membrane anchors": First, the kinases require the agonist-occupied, active conformation of the receptors to which they bind [13]. Second, the kinases bind to the G protein [3y subunits which may, in addition to the translocation, result in activation of the kinases [14, 15]. This process appears to be supported by specific combinations of [3T subunits much better than by others [16]. And third, recent data indicate that the kinase binds to phosphatidylinositol bisphosphate (PIP2) via a C-terminal pleckstrin homology domain [17]. At least the first two modes of membrane anchoring and activation appear to be involved in the strictly agonist-dependent action of [3ARK on [3-adrenergic receptors. Phosphorylation of receptors by [3ARK increases their affinity for the [3-arrestins. The [3-arrestin isoforms 1 and 2 are members of the larger family of arrestin proteins (reviewed in [18]). In contrast to the other arrestins which are localized essentially in the retina, [3-arrestin-1 and 2 are widely distributed and are p r o b a b l y most i m p o r t a n t for desensitization of hormone and
14 neurotransmitter receptors. Binding of these proteins to phosphorylated receptors blocks the interaction between receptors and G proteins [8, 9, 19]. DESENSITIZATION BY PROTEIN KINASES A AND C In addition to phosphorylation by ~ARK, ~-adrenergic receptors can also be phosphorylated by protein kinases A and C. While the phosphorylation by ~ARK occurs on multiple sites of the extended C-terminus of these receptors, these kinases phosphorylate two consensus sites which are located close to the membrane in the third cytoplasmic loop of the receptors and in the C-terminus. This phosphorylation results in direct impairment of receptor-Gs-coupling [20, 21]. However, it appears that this type of receptor uncoupling occurs more slowly and is of smaller extent than the [3ARK/~-arrestin-mediated p a t h w a y [11, 22]. Only GRK-mediated phosphorylation of receptors causes enhanced affinity for arrestins. In contrast, phosphorylation of ~2-adrenergic receptors by PKA or PKC has no effect on the affinity or activity of arrestins [15]. REGULATION OF G-PROTEIN FUNCTION In addition to the modulation of signaling at the level of the receptors signal transduction appears also to be regulated at the next level, the G proteins. Again, regulation can occur either via alterations in the function of a G protein or by alterations in its number. Alterations in the number of G proteins have been described mostly for the G proteins regulating adenylyl cyclase, G s and G i. In these cases there appears to be pronounced 'cross-regulation'. Activation of the Gs-pathway leads to enhanced expression of Gi, and activation of the Gi-pathway causes reduced expression of G i [23, 24]. Such regulation occurs not only in isolated cells, but has also been observed in intact animals and in humans. A well-documented pathophysiological example of such regulation is the increased expression of Gia_2 in heart failure [25]. More recently it has become clear that also the function of G proteins can be altered. There are a small number of proteins that have been found to associate with G proteins and to affect their function. These proteins include the growth cone associated protein GAP-43, which has been found to enhance GTP binding by G o in a manner similar to receptors [26], and a complex between the small GTP-binding protein ras p21 and its GTPase activating protein (ras-GAP) which impair coupling of muscarinic receptors to potassium channels [27]. A regulatory protein which can inhibit signaling via multiple G proteins including G s is the cytosolic 33kDa protein phosducin [28]. Phosducin had earlier been believed to be expressed only in the retina and in the developmentally
15 related pineal gland [29, 30], but its mRNA has more recently been found in many other tissues [28]. Phosducin interacts preferentially with the ~, subunits of G proteins, but probably also with their 0~-subunits [28, 31, 32]. This interaction with the G proteins results in inhibition of the activation of G s, but also of G o, Gi and transducin. This inhibition of G protein activation causes a disruption of signaling from [~-adrenergic receptors to adenylyl cyclase [28]. Protein kinase A can phosphorylate phosducin [33]. This phosphorylation markedly inhibits the effects of phosducin on G proteins [28, 34]. Since phosducin inhibits the function of G s, and protein kinase A prevents this inhibition, G s, adenylyl cyclase, protein kinase A and phosducin form a positive feed-back loop. The biological implication of such positive feed-back are as yet not understood. Phosducin and [3ARK both bind to the ~, subunits of G proteins. Because of its very high affinity for the [3~,subunits phosducin can compete very effectively with ~ARK and thereby prevent its translocation to the plasma membrane [35]. As a consequence, phosducin can inhibit ~ARK-mediated phosphorylation and desensitization of ~-adrenergic receptors. Again, this inhibition is relieved following phosphorylation of phosducin by protein kinase A. This indicates that the cytosolic components of the ~-adrenergic receptor system interact with each other in multiple ways. Together, they form a complex network which regulates the signaling function of the receptor system. N-FORMYLMETHIONYL PEPTIDE RECEPTORS AND MEMBRANE DOMAINS N-formylmethionyl peptide receptors (FPR) are chemoattractant receptors on phagocytic cells like human neutrophils. Stimulation of FPR with N-formylmethionyl peptides induces a variety of responses of these cells including chemotaxis, superoxide production, release of hydrolytic enzymes and reorganization of cytoskeletal structures [36, 37]. Most of these responses appear to be mediated through activation of phospholipase C, however, participation of other effector enzymes such as adenylyl cyclase, phospholipase D and phospholipase A 2 have also been demonstrated [38]. In analogy to other receptor systems desensitization of FPR was observed. N-formylmethionyl peptides induce a transient burst of superoxide production in human neutrophils with a maximum after 1-2 min. In the past, several observations suggested that desensitization of FPR may involve the cytoskeleton [39]: (1) The microfilament-disrupting agent cytochalasin B substantially increased the rate and duration of the superoxide production induced by formyl peptides [40]. (2) FPR in desensitized neutrophils accumulated in a Triton X-100-insoluble fraction of the cells [40, 41]. (3) Plasma membrane fractionation revealed that after desensitization FPR appear to be redistributed to a membrane fraction enriched
16 in cytoskeletal proteins like actin and fodrin that was depleted from G proteins [42]. Two membrane fractions can be distinguished in neutrophil plasma membrane preparations based on different densities. The fraction with the higher density (PM-H) contains cytoskeletal proteins whereas the fraction with the lower density (PM-L) is enriched in G proteins. These membrane fractions are thought to correspond to plasma membrane domains with the respective specific distribution of proteins associated with the inner face of the membrane. In the two different membrane domains FPR were expected to show different coupling to G proteins. Solubilized FPR prepared from the PM-L fraction of neutrophils sedimented in a detergent-containing sucrose density gradient as 7 S particles while FPR prepared from PM-H sedimented with an apparent sedimentation coefficient of 4 S. The 7 S form was shifted to the 4 S form in a GTP~/S-sensitive manner indicating that the 7 S form represents receptorG protein complexes.
RESPONSIVE
PM-L
DESENSITIZED
G proteins
G proteins FPR
PM-H
Cytoskeletal proteins
] I I I FPR
Cytoskeletal proteins FPR
Figure 2. Domains and protein distribution in responsive and desensitized neutrophil plasma membranes. For details see text. These data led to the proposal of a model for desensitization based upon lateral segregation of signaling proteins in the plane of the plasma membrane. FPR in responsive cells are able to transduce a signal in the PM-L compartment that is enriched in G proteins. Activation of receptors would then allow for interaction with their signal transduction partners. In desensitized neutrophils this interaction is thought to be blocked because FPR are laterally segregated into the PM-H domain where only low levels of G protein are observed (Figure 2).
17 Recently, an agonist-induced redistribution of FPR in h u m a n neutrophils consistent with the lateral segregation model was d e m o n s t r a t e d by direct m e a s u r e m e n t of receptor lateral mobility with fluorescence recovery after photobleaching [43]. FPR-COUPLING TO THE MEMBRANE SKELETON
The lateral segregation mechanism of FPR desensitization in neutrophils (Figure 2) may be an attractive model that integrates the receptor-mediated actin polymerization/depolymerization and feed-back regulation of receptor function. What do we know about FPR binding to cytoskeletal proteins and membrane disribution of FPR as a function of the activation state of neutrophils to support such an idea? The initial observation that FPR accumulate in the Triton X-100-insoluble fraction of neutrophils upon desensitization led to a more detailed study of the cellular structures that might be implicated in the regulation of receptor function. The signal transduction steps that involve the receptor protein as well as the crucial desensitization events appear to take place in the plasma membrane because desensitization does occur at 15~ a temperatur that inhibits internalization of receptors [41]. Therefore, a potential interaction of receptors with cytoskeletal proteins on the level of the plasma m e m b r a n e was investigated. An association of FPR with the submenbraneous cytoskeleton, respectively the m e m b r a n e skeleton, was shown by centrifugation of detergent-solubilized membranes from responsive neutrophils in sucrose density gradients. When octylglucoside was used as a detergent all receptors were soluble. In contrast, use of Triton X-100 as a detergent that does not solubilize m e m b r a n e skeletal structures, about 50% of the receptors sedimented to the pellet along with m e m b r a n e skeletal actin. Solubilization with Triton X-100 in the presence of agents that cause actin filaments to depolymerize, caused FPR to be released from the membrane skeletal pellet along with actin (Figure 3, [44]). This indicates that some linkage exists between FPR and membrane skeletal actin. In order to define the functional significance of this linkage of FPR to the membrane skeleton in responsive neutrophils, this association was investigated in cells that were desensitized to various degrees. In cells partially desensitized by incubation with an agonist at 4~ about 70% of the receptors were found in the membrane skeletal pellet. In fully desensitized neutrophils virtually all (>90%) receptors sedimented to the pellet (Figure 3).
18
100
80
60
9 !,,,I
40
20
0
C
PD
FD
KC1
pCMPS
DNAse I
Figure 3. Membrane skeletal association of FPR. Neutrophils were solubilized with Triton X-100 and the extracts were spun over sucrose density gradients. The distribution of photoaffinity labeled FPR was analyzed on SDS-polyacrylamide gels with a Phosphor Imager. Percent receptors that are coupled to the membrane skeleton are shown for responsive control cells (C), partially (PD) and fully (FD) desensitized neutrophils. In addition, values are shown for cells after solubilization in the presence of agents causing depolymerizaiton of actin filaments, like KC1, p-chloromercuriphenylsulfonic acid (pCMPS) or DNAse I. For experimental details see [44]. Previous characterization of FPR revealed three distinct ligand-receptor complexes corresponding to defined activation states of the receptor [45]. The most striking feature of the desensitized state, named LRX in order to describe the interaction of the ligand-receptor complex with an unidentified component X, is the energy-dependent formation upon agonist stimulation with a timecourse similar to the desensitization of the response [45]. Recently we have shown that the agonist-induced coupling FPR of the membrane skeleton is also energy-dependent [46], confirming that the membrane skeleton may indeed represent the unidentified component X that is needed to accomplish
19 desensitization. Table I shows that depletion of adenine and guanine nucleotides results in a dramatically reduced FPR coupling to the membrane skeleton. Table 1 FPR coupling to the membrane skeleton in energy-depleted cells percent FPR in pellet Control 15 min fluoride treatment 60 min fluoride treatment
74.9 + 4.3 30.2 + 4.6 28.9 + 2.7
Neutrophils were energy-depleted by treatment with 40 mM NaF for 15 min (depletion of ATP and GTP) and 60 rain (depletion of ATP and GTP including soluble diphosphates, [45]). After this pretreatment the cells were desensitized by incubation with an agonist. For details see [46]. Taken together, these data suggest that an increased FPR coupling to the membrane skeleton in response to agonist stimulation is a basic mechanism for h o m o l o g o u s desensitization of this receptor. This novel desensitization processes may be specific for chemoattractant receptors as a subclass of G proteincoupled receptors [47]. However, the possibility that phosphorylation-dependent mechanisms similar to those observed during desensitization of the [3-adrenergic receptor play an additional regulatory role in the FPR p a t h w a y cannot be excluded (see below). WHAT IS THE LINK OF FPR TO THE MEMBRANE SKELETON?
A central role of the membrane skeleton in the desensitization of FPR in h u m a n neutrophils has been well documented in recent years (for review see [5, 48]). H o w e v e r , the nature of the molecular link of the receptor to the m e m b r a n e skeleton remained elusive. The release of FPR from m e m b r a n e skeletal pellets with agents that depolymerize actin [44] suggests that F-actin may participate in the immobilization process that prevents the receptor from interaction with G proteins. Although it is difficult to demonstrate a specific interaction of a receptor with a protein as abundant as actin there are a series of observations that suggest a specific and direct interaction of FPR with actin. First, an overlay assay was used to demonstrate binding of photoaffinity labeled FPR to actin from neutrophil cytosol [49]. Second, actin increased the sedimentation rate of Triton X-100-solubilized FPR in a sucrose density gradient [49]. Third, anti-actin antibodies were able to immunosediment FPR suggesting
20 the existence of FPR-actin complexes [44, 49]. Fourth, exogenously added actin p r o m o t e d agonist binding to FPR in actin-stripped membranes or FPR solubilized from actin-stripped membranes [49]. These observations suggest that the FPR is indeed an actin-binding protein. The function of actin would be to serve as a matrix that allows for immobilization of FPR in a membrane domain that is depleted in G proteins and, thus, would interrupt the signaling cascade in the process of desensitization. Similarily, binding to actin of the EGF receptor as a member of the tyrosine kinase receptors has been well documented [50]. However, a function for actin binding of the EGF receptor has not yet been identified. At present it is not known what triggers the binding of FPR to the membrane skeleton. In theory several mechanisms are conceivable. A covalent modification like phosphorylation could change the affinity of FPR for G proteins a n d / o r actin. In such a model actin might have a preference for phosphorylated receptors in analogy to the increased affinity of arrestins for phosphorylated rhodopsin or [3-adrenergic receptors [1]. Another possibility might be a conformational change caused by agonist binding to the receptor. So far, there is no indication for phosphorylation being involved in FPR desensitization via the "membrane skeleton pathway". Okadaic acid and staurosporine, inhibitors of serine/threonine phosphatases and protein kinases A and C, respectively, have no significant effect on FPR coupling to the membrane skeleton [46]. In addition, desensitization of neutrophils does not change the affinity of FPR for G protein [51]. An appealing possibility would be that local actin polymerization is involved in the immobilization process leading to desensitization. The energy-dependence of the membrane skeletal association [46] may serve as a hint to such a possibility. FPR PHOSPHORYLATION
In h u m a n neutrophils receptor phosphorylation does not appear to be necessary for the homologous desensitization pathway that implicates receptor immobilization at the membrane skeleton [46]. Nevertheless, phosphorylation of FPR has been demonstrated in differentiated HL60 cells [52] and in rat basophilic leukemia cells (RBL-2H3) with stably expressed FPR [53]. Both studies showed that an agonist-induced and staurosporine-insensitive phosphorylation occured, suggesting a role for a kinase in homologous desensitization. More recently, phosphorylation of the C-terminus of FPR with a kinase different from protein kinase A, protein kinase C or tyrosine kinases has been demonstrated [54]. This phosphorylation, most likely by GRK2, affects serine and threonine residues in a portion of the C terminus that is involved in receptor-G protein coupling [55].
21 How is this observation related to the desensitization via receptor coupling to the membrane skeleton? At present there is no evidence for a role of phosphorylation in FPR desensitization via the lateral segregation pathway. Although inhibition of protein kinase A and protein kinase C with staurosporine does not affect FPR coupling to the membrane skeleton [46], phosphorylation by a G protein-coupled receptor kinase (GRK) may still trigger FPR binding to actin. However, the finding that inhibition of serine/threonine phosphatases with ocadaic acid has also no significant effect on FPR immobilization [46] argues against such a possibility. It appears more likely that phosphorylation and membrane skeleton coupling are parallel mechanisms with the former representing a general mechanism for all G protein-coupled receptors and the latter serving as a mechanism that is operative for the functional subclass of chemoattractant receptors. RECEPTOR-CLASS DESENSITIZATION The possible existence of a mechanism of desensitization specific for chemoattractant receptors was proposed based on the observation that FPR and C5a receptors exhibited heterologous desensitization whereas no crossdesensitization was observed with c~1 adrenergic receptors although all three receptor subtypes are coupled to phospholipase C as an effector enzyme [47]. The cross-regulation of G protein-coupled receptors in neutrophils appears to be a highly complex process because the susceptibility to cross-desensitization is dramatically different for different Ca2+-mobilizing receptors such as peptide and lipid chemoattractant receptors or receptors for ligands other than chemoattractants [56]. Phosphorylation by PKC might be one mechanism for differential crossregulation because not all receptors coupled to the PLC pathway possess a consensus sequence for phosphorylation by PKC [56]. The complexity of the network that appears to be involved in the cross-talk between various receptors of the PLC pathway suggests the existence of multiple mechanisms to accomplish this complicated regulatory task. It is conceivable that interaction of GRKs with specific G protein ~, subunits allows for differential desensitization of different receptors [16]. However, in addition to the well characterized GRK mechanism the lateral segregation model provides an appealing mechanism to explain some of the observed phenomena [5].
22 SIGNAL TRANSDUCTION AND MEMBRANE ORGANIZATION The discovery of an increasing number of proteins that are involved in fine tuning of the intensity of receptor-mediated signaling raises the general question of the spatial organization of the relevant components. Simple collision coupling by lateral motion of proteins in the plasma membrane might be appropriate to explain receptor-G protein or G protein-effector coupling. For an efficient interplay of all proteins involved in signal modulation an organized distribution of the regulatory components appears to be required. Although a clear picture of such a membrane compartmentation remains to be developed, there are some data that point to an important contribution of cytoskeletal structures in the organizational control of the signal trasnduction machinery. As has been shown for FPR and other receptors diffusion of a receptor protein may be restricted by interaction with the membrane skeleton [5, 50]. On the other hand, interaction of G proteins with cytoskeletal elements has also been demonstrated providing for a means to also control G protein distribution in the plasma membrane (for review see [57]). An intriguing idea explaining distinct membrane distribution of G proteins has been put forward by Rodbell and coworkers who observed polydisperse forms of G proteins caused by polymerization of G a subunits in analogy to tubulin [58, 59]. Formation of these large molecular complexes would result in restricted mobility of the G proteins and could serve as an explanation for membrane domains in neutrophils that are enriched in G proteins (see above). CONCLUSION Receptor-mediated signal transduction is no longer considered to be just a simple interaction of a limited number of proteins. It has become a highly regulated process in a complicated network of an increasing number of components. The increasing complexity of the regulation of receptors suggests that spatial organization of proteins is necessary in order to achieve a highly reliable and efficient regulatory machinery [57]. The proposed model for desensitization of FPR that requires lateral segregation of m e m b r a n e components may serve as a simple example for spatial control of signal transduction. It appears that also in systems like the [~-adrenergic receptor a subtle control of the interaction of all the known components must be operative resulting in a well organized and specific signaling process in the cell.
23 Acknowledgement Work in the authors' laboratories is supported by grants from the American Cancer Society (IRG-172B, KNK); the National Institute of Health (RO1-AI-22735 and DMB 900058P, AJJ); the Deutsche Forschungsgemeinschaft, the European Commission, the Fonds der Chemischen Industrie (MJL). REFERENCES
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20
Lohse MJ. Biochim Biophys Acta 1993; 1179: 171-188. Hausdorff WP, Caron MG, Lefkowitz RJ. FASEB J 1990; 4: 2881-2889. Hargrave PA, McDowell JH. FASEB J 1992; 6: 2323-2331. Hosey MM, Benovic JL, DebBurman SK, Richardson RM. Life Sci 1995; 56: 951-955. Klotz K-N, Jesaitis AJ. Bioessays 1994; 16: 193-198. Benovic JL, Strasser RH, Caron MG, Lefkowitz RJ. Proc Natl Acad Sci USA 1986; 83: 2797-2801. Benovic JL, DeBlasi A, Stone WC, Caron MG, Lefkowitz RJ. Science 1989; 246: 235-240. Lohse MJ, Benovic JL, Codina J, Caron MG, Lefkowitz RJ. Science 1990; 248: 1547-1550. Lohse MJ, Andexinger S, Pitcher J, Trukawinski S, Codina J, Faure J-P, Caron MG, Lefkowitz RJ. J Biol Chem 1992; 267: 8558-8564. Attramadal H, Arriza JL, Aoki C, Dawson TM, Codina J, Kwatra MM, Snyder SH, Caron MG, Lefkowitz RJ. J Biol Chem 1992; 267: 17882-17890. Roth N, Campbell PT, Caron MG, Lefkowitz RJ, Lohse MJ. Proc Natl Acad Sci USA 1991; 88: 6201-6204. Premont RT, Inglese J, Lefkowitz RJ. FASEB J 1995; 9: 175-182. Chen C-Y, Dion SB, Kim CM, Benovic JL. J Biol Chem 1993; 268: 7825-7831. Haga K, Haga T. J Biol Chem 1992; 267: 2222-2227. Pitcher JA, Inglese J, Higgins JB, Arriza JL, Casey PJ, Kim C, Benovic JL, Kwatra MM, Caron MG, Lefkowitz RJ. Science 1992; 257: 1264-1267. M~iller S, Hekman M, Lohse MJ. Proc Natl Acad Sci USA 1993; 90: 1043910443. Touhara K, Koch WJ, Hawes BE, Lefkowitz RJ. J Biol Chem 1995; 270: 1700017005. Wilson CJ, Applebury ML. Curr Biol 1993; 3: 683-686. Wilden U, Hall SW, Kuhn H. Proc Natl Acad Sci USA 1986; 83: 1174-1178. Benovic JL, Pike LJ, Cerione RA, Staniszewski C, Yoshimasa T, Codina J, Caron MG, Lefkowitz RJ. J Biol Chem 1985; 260: 7094-7101.
24 21 Bouvier MB, Leeb-Lundberg LMF, Benovic JL, Caron MG, Lefkowitz RJ. J Biol Chem 1987; 262: 3106-3113. 22 Lohse MJ, Benovic JL, Caron MG, Lefkowitz RJ. J Biol Chem 1990; 265: 32023209. 23 Hadcock JR, Ros M, Watkins DC, Malbon CC. J Biol Chem 1990; 265: 1478414790. 24 Eschenhagen T, Mende U, Nose M, Schmitz W, Scholz H, Schulte am Esch J, Warnholtz A, Sch/ifer H. Circ Res 1992; 70: 688-696. 25 Eschenhagen T, Mende U, Diederich M, Nose M, Schmitz W, Scholz H, Schulte am Esch J, Warnholtz A, Sch/ifer H. Mol Pharmacol 1992; 42: 773-783. 26 Strittmatter SM, Valenzuela D, Sudo Y, Linder ME, Fishman MC. J Biol Chem 1991; 26: 22465-22471. 27 Yatani A, Okabe K, Polakis P, Halenbeck R, McCormick F, Brown AM. Cell 1990; 61: 769-776. 28 Bauer PH, Mfiller S, Puzicha M, Pippig S, Helmreich EJM, Lohse MJ. Nature 1992; 358: 73-76. 29 Lee RH, Flower A, McGinnis JF, Lolley RN, Craft CM. J Biol Chem 1990; 265: 15867-15873. 30 Reig JA, Yu L, Klein DC. J Biol Chem 1990; 265: 5816-5824. 31 Lee RH, Ting TD, Liebermann BS, Tobias DE, Lolley RN, Ho Y-K. J Biol Chem 1992; 267: 25104-25112. 32 Xu J, Wu D, Slepak VZ, Simon MI. Proc Natl Acad Sci USA 1995; 92: 20862090. 33 Lee RH, Brown BM, LoUey RN. J Biol Chem 1990; 265: 15860-15866. 34 Yoshida T, Willardson BM, Wilkins JE, Jensen GJ, Thornton BD, Bitensky MW. J Biol Chem 1994; 269: 24050-24057. 35 Hekrnan M, Bauer PH, S6hlemann P, Lohse MJ. FEBS Lett 1994; 343: 120-124. 36 Boxer GJ, Curnutte JT, Boxer LA. Hosp Practice 1985; 40: 69-90. 37 Snyderman R, Uhing RJ. In: Gallin JI, Goldstein IM, Snyderman R, eds. Inflammation. New York: Raven Press, 1988; 309-323. 38 Cockcroft S. Biochim Biophys Acta 1992; 1113: 135-160. 39 Jesaitis AJ, Bokoch GM, Allen RA. In: Condeelis J, Lazarides E, Satir P, eds. Signal Transduction in Cytoplasmic Organization and Cell Motility. New York: Alan R Liss, Inc, 1988; 325-337. 40 Jesaitis AJ, ToUey JO, Allen RA. J Biol Chem 1986; 261: 13662-13669. 41 Jesaitis AJ, Naemura JR, Sklar LA, Cochrane CG, Painter RG. J Cell Biol 1984; 98: 1378-1387. 42 Jesaitis AJ, Bokoch GM, Tolley JO, Allen RA. J Cell Biol 1988; 107: 921-928. 43 Johansson B, Wymann MP, Holmgren-Peterson K, Magnusson K-E. J Cell Biol 1993; 121- 1281-1289.
25 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59
Klotz K-N, Krotec KL, Gripentrog J, Jesaitis AJ. J Immunol 1994; 152: 801-810. Sklar LA, Mueller H, Omann G, Oades Z. J Biol Chem 1989; 264: 8483-8486. Klotz K-N, Jesaitis AJ. Cell Signal 1994; 6: 943-947. Didsbury JR, Uhing RJ, Tomhave E, Gerard C, Gerard N, Snyderman R. Proc Natl Acad Sci USA 1991; 88: 11564-11568. Jesaitis AJ. Comments Mol Cell Biophys 1992; 8: 97-114. Jesaitis AJ, Erickson RW, Klotz K-N, Bommakanti RK, Siemsen DW. J Immunol 1993; 151: 5653-5665. den Hartigh JC, van Bergen en Henegouwen PMP, Verkleij AJ, Boonstra J. J Cell Biol 1992; 119: 349-355. Klotz K-N, Jesaitis AJ. Biochem Pharmacol 1994; 48: 1297-1300. Tardif M, Mery L, Brouchon L, Boulay F. J Immunol 1993; 150: 3534-3545. Ali H, Richardson RM, Tomhave ED, Didsbury JR, Snyderman R. J Biol Chem 1993; 268: 24247-24254. Prossnitz ER, Kim CM, Benovic JL, Ye RD. J Biol Chem 1995; 270: 1130-1137. Bommakanti RK, Klotz K-N, Dratz EA, Jesaitis AJ. J Leukocyte Biol 1993; 54: 572-577. Tomhave ED, Richardson RM, Didsbury JR, Menard L, Snyderman R, Ali H. J Immunol 1994; 153: 3267-3275. Neubig RR. FASEB J 1994; 8: 939-946. Nakamura S-I, Rodbell M. Proc Natl Acad Sci USA 1990; 87: 6413-6417. Coulter S, Rodbell M. Proc Nail Acad Sci USA 1992; 89: 5842-5846.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardin~, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
27
Introduction to Muscarinic Receptors
Gideon Shapiro Sandoz Pharma Ltd., Basel Switzerland CH-4002
The muscarinic system is one of the earliest established components of the nervous system. Its pharmacology was already well defined over one hundred years ago using poisons or medicinal agents of the time, the natural products muscarine [1] pilocarpine, atropine and physostigmine. Interestingly the elucidation of acetylcholine [2] as the endogenous neurotransmitter was achieved many decades later highlighting the importance of natural products in medicine and pharmacology which remains with us.
OH
____/
I...../-~"0)'''' OH3 N+ OH3 / ~CH3 CH3
Muscarine Amanita Muscaria
Atropine Belladona
I
N'~I N
Me
o NH o.
/ Me \
Pilocarpine
Pilocarpus Jaborandi
Physostigmine
I
o II ~o "7"" Me
Acetylcholine
Physostigma venenosum
Over the past several decades with the ripening of the discipline of organic synthesis, man made compounds have been prepared which act on the functional unit of the muscarinic system which we now recognize as a receptor protein. Some of these have or remain to be used as therapeutic agents. More recently, the efforts of synthetic organic chemists have yielded muscarinic receptor antagonist ligands such as pirenzepine, AFDX-112, 4-DAMP, and
28 methoctramine which have catalyzed a revival of muscarinic receptor pharmacology. With these tools it has been possible to classify muscarinic receptors in terms of subtypes M1, M2 and M3 [3] which presumably allow subtle regulation of physiological processes. Again basic pharmacological science and the development of therapeutic agents have gone hand in hand, pirenzepine, an agent which inhibits gastric acid release, reaching the marketplace for ulcer treatment [4]. Many other selective agents have since been synthesized which are under investigation for various indications.
N~
~ OH
~N~)
~
/
Pirenzepine(M1)
N(Et)2
AF-DX116 (M2)
Hexahydrosiladifenidol(M3) I
oH
~ N ~
o O " ~ ~ N ~ */
v
N
-O H
1
4-DAMP(M3)
Methoctramine
(M2)
The latest impetus in the muscarinic field has come from molecular biology which has brought the muscarinic receptor, once a hypothetical construct, to the level of an expressed gene with a corresponding protein primary sequence. The cloning of muscarinic receptors has not only confmned but expanded the subtype division from three pharmacologically characterizable to five (ml-m5) molecular entities [3]. Compound screening has been greatly enhanced by the availability of cell lines expressing homogenous populations of specific receptor subtypes on which binding and functional studies can be performed. A great impact may be forseen on the search for therapeutically useful subtype selective compounds. Recent examples of this are the ml selective agonists Xanomeline and PD-151832 which are potential Alzheimer therapeutics. Finally, the structure of the muscarinic receptor(s) may be determined someday in the future ideally as ligand complexes. Until this is achieved the primary sequence used in tandem with mutagenesis experiments [5] and the only structure determined to date of
29 a membrane bound G-protein coupled receptor, namely bacteriorhodopsin [6], will have to continue to guide current efforts [7] to understanding how muscarinic ligands interact with the
~
N "" S
Xanomeline
\
N
\o
PD151832
/.O
receptor. Previously, structure activity relationships and pharmacophore models have been formulated solely on the basis of ligand structure and the receptor has been considered to be a rigid entity. The limitations of this assumption have been pointed out using a simplified hypothetical model for the binding of muscarinic agonists to a segment of transmembrane helix-3 of the ml receptor [8]. Conformation mobility of the receptor is necessary to account for optimal binding for all of the potent agonists studied. Where is the future of muscarinic receptor ligands/drugs headed? High capacity screening technologies using optimized cloned systems may tuna up interesting selective compounds already sitting on the shelf. Selective antagonists may certainly be improved upon while the difficulty in making selective agonists may be of an intrinsic nature. For the design of new therapeutically useful ligands the new insights into muscarinic receptor structure and function provide a sound basis for progress.
References 1. Schmiedeberg O and Koppe R. Das Muscarin, das Giftige Alkaloid des Fliegenpilzes, Leipzig: Vogel, 1869. 2. Dale HH and Dudley FH. J Physiol 1929; 68: 97-123. 3. Receptor and Ion Nomenclature Supplement Trends Pharmacol Sci 1995; 16: 4. 4. Blum AL and Hammer R, eds. Die Behandlung des Ulcus Pepticum mit Pirenzipin, Munich: Demter, 1979. 5. Liu J, Schoeneberg T, Van Rhee M and Wess J. J Biol Chem 1995; 270:1953219539. 6. Henderson R, Baldwin JM, Ceska TA, Zemlin F, Beckmann E and Downing KH. J Mol Biol 1990, 213: 899-929. 7. Hibert MF, Tnmapp-Kallmeyer S, Bruinvels A and Hoflack J. Trends Pharmacol Sci 1993; 14: 7-12. 8. Shapiro G, Floersheim P, Boelsterli J, Amstutz R, Bolliger G, Gammenthaler H, Gmelin G, Supavilai P and Walkinshaw M. J Med Chem 1992; 35:15-27.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardinh, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
31
Structure-function analysis of muscarinic acetylcholine receptors J. Wess, N. Blin, J. Yun, T. SchSneberg and J. Liu Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, Bldg. 8A, Room B 1A-09, Bethesda, MD, U.S.A. INTRODUCTION The muscarinic acetylcholine receptors (mAChRs) are typical members of the superfamily of G protein-coupled receptors (GPCRs; refs. 1,2). Following the cloning of five distinct mAChR genes (ml-m5), the last decade has witnessed a flood of new information about how these receptors function at a molecular level. Molecular genetic and biochemical approaches have been applied to identify receptor regions/single amino acids involved in ligand binding, G protein coupling, regulation of receptor activity, and other fundamental aspects of mAChR function. In this chapter, we will describe recent work dealing with the structure and assembly of mAChRs as well as with the molecular mechanisms governing the selectivity of receptor/G protein interactions. A R R A N G E M E N T OF T H E T R A N S M E M B R A N E HELICES
Like all other G protein-coupled receptors (GPCRs), the five mAChRs are predicted to be composed of seven a-helically arranged transmembrane domains (TM I-VII) connected by three extraceUular (o2-o4) and three intracellular (il-i3) loops [1,2]. Several lines of evidence suggest that the seven TM helices are arranged in a ring-like fashion, thus forming a tightly packed helical bundle [3-6]. Site-directed mutagenesis [7,8] and affinity labelling studies [9,10] have shown that residues present on the inner surfaces of different TM helices (facing the cavity enclosed by TM I-VII) are involved in the binding of muscarinic agonists (including the neurotransmitter, acetylcholine) and antagonists. Given the lack of high-resolution structural information on any GPCR, several laboratories have employed mutagenesis approaches to identify molecular interactions between individual TM helices [11-16]. We previously identified a series of hybrid m2/m5 receptors containing m2 receptor sequence in TM VII and m5 receptor sequence in TM I (such as C4 in Fig. 1B) which were unable to bind significant amounts of muscarinic radioligands [12]. However, replacing TM I in these apparently misfolded mutant receptors with the corresponding m2 receptor segment could restore proper ligand binding activity [12], indicating that TM I and TM VII are located directly adjacent to each other and that molecular interactions between these two TM helices are required for proper receptor folding. Immunocytochemical and ELISA studies showed that the pharmacologically inactive m2/m5 hybrid receptors were properly expressed on the cell surface [16], suggesting that the virtual lack of binding activity seen with these mutant receptors is due to a specific folding defect. It seemed reasonable to assume that the amino acids responsible for this folding defect are located at the TM Ifi'M VII
32
A
m2
m5 143v36G47
m, f424,~ ~-A~ ~L428 m32:..:.~.~~'.V~ W427r ,,,- ~C~9 / VI
isles41
B
Y4?..26S~3~
I
m5
~'''l
Ligand Binding Cavity
m2
m5"i'37->m2A30
[3H]NMS Binding Bmax (fmol/mg) K D (pM)
m2 (wt) 698 _+84 53 __.10
m2T423->m5H4 78
C4 < 25
541 +_79
556 +_54
52 _+4
76 +_20
Figure 1. Pharmacological activity of hybrid m2/m5 mAChRs. (A) Helical wheel model of the possible orientation of TM I and VII in mAChRs, assuming an anticlockwise connectivity of TM I-VII. The view is from the extracellular surface of the membrane. Shown here is a pharmacologically inactive hybrid receptor [12] containing m5 receptor sequence in TM I and m2 receptor sequence in TM VII (only the first 18 amino acids of TM I and VII are shown). Amino acids which differ between the m2 and m5 mAChRs (human) are marked with asterisks. The two Thr residues (m5Thr37 and m2Thr423) which face each other at the TM IfVII interface in the pharmacologically inactive hybrid receptors are highlighted. The model shown here slightly differs from the Baldwin projection [4,5] in that TM VII has been rotated by approximately 30 degrees (counterclockwise) to allow m2Tyr426 (underlined) to project into the ligand binding cavity formed by TM III-VII [8,48]. (B) Structure and [3H]NMS binding properties of hybrid m2/m5 mAChRs transiently expressed in COS cells [16].
33 interface and that the identification of these residues might therefore provide useful information as to how TM I and TM VII are oriented relative to each other. Helical wheel models [4,5] of the possible a r r a n g e m e n t of TM I-VII (assuming an anticlockwise connectivity of TM I-VII, as viewed from the extracellular membrane surface) showed that there are only two residues at the TM IfrM VII interface in which the m2 and m5 mAChRs differ [16]. As shown in Fig. 1A, in all misfolded m2/m5 hybrid receptors, m5Thr37 (TM I) is predicted to be located adjacent to m2Thr423 (TM VII). We therefore speculated that a novel hydrogen bond can form between these two Thr residues (which are both located 1-2 helical turns away from the membrane surface), thus interfering with proper helix/helix packing and receptor folding. To test this hypothesis, two single point mutations, m5Thr37->m2Ala30 or m2Thr423->m5His478, were introduced into all pharmacologically inactive hybrid receptors (shown for C4 in Fig. 1B). If the model shown in Fig. 1A is correct, these mutations should "recreate" the TM IfI?M VII interfaces present in the wild type m2 and m5 mAChRs, respectively [16]. Consistent with our working hypothesis, we found that either of the two point mutations resulted in mutant receptors which, when transiently expressed in COS-7 cells, were able to bind significant amounts ofmuscarinic radioligands (shown for C4 in Fig. 1B) and to couple to G proteins [16]. These findings therefore indicated that the model shown in Fig. 1A that guided this mutagenesis study is probably correct. This model was based on the recently proposed "Baldwin projection" [4,5], assuming an anticlockwise connectivity of the TM helices (as viewed from the extracellular membrane surface). In a model assuming a Clockwise arrangement of the TM helices, m5Thr37 (TM I) would still face TM VII, whereas m2Thr423 (TM VII) would lie adjacent to TM VI [16]. Such an a r r a n g e m e n t makes it very difficult to rationalize how mutational modification of either of these two Thr residues can restore a proper receptor fold. Our data therefore strongly support the concept that the TM helices in GPCRs are arranged in an anticlockwise fashion (as viewed from the cell surface). RECEPTOR ASSEMBLY
Previous studies have shown that GPCRs are composed of two major building blocks, one containing TM I-V and the other, TM VI and VII [17-19]. It was found that these two domains, when coexpressed as two separate polypeptides, can fold independently of each other and then properly assemble to form functional receptor complexes [17-19]. Based on these results, we have recently tested the hypothesis that GPCRs are assembled from many, r a t h e r t h a n only two, autonomous folding units. To address this question, the r a t m3 mAChR was "split" in all three intracellular (il-i3) and all three extracellular loops (o2-o4), thus generating six polypeptide pairs (Nil+Cil, No2+Co2, etc.; Fig. 2; ref. 20). As shown in Fig. 3, coexpression of three of the six polypeptide pairs (Ni2+Ci2, No3+Co3, and Ni3+Ci3, respectively) led to the appearance of a significant number of specific high-affinity [3H]NMS binding sites. These data clearly indicated that covalent connections between TM III and IV, TM IV and V, and TM V and VI, respectively, are not essential
34
n3
;llular .
v
Figure 2. The rat m3 mAChR was "split" at the indicated positions (arrows), thus generating six polypeptide pairs [20]. for proper receptor folding. However, it was also noted that the the Ni2+Ci2 and No3+Co3 polypeptide complexes showed significant decreases in binding affinities for most ligands tested (including acetylcholine; Fig. 3), suggesting that the i2 and o3 loops exert indirect conformational effects on the proper arrangement of the TM helical bundle predicted to contain the primary ligand binding site. To investigate whether the Ni2+Ci2, No3+Co3, and Ni3+Ci3 polypeptide complexes were still capable of activating G proteins, their ability to mediate carbachol-induced phosphoinositide (PI) hydrolysis was examined [20]. As shown in Fig. 4, the No3+Co3 and Ni3+Ci3 complexes were able to stimulate PI hydrolysis to a similar maximum extent as the wild type m3 mAChR. In contrast, the Ni2+Ci2 complex displayed only residual functional activity, indicating that the structural integrity of the i2 loop is essential for efficient G protein coupling [20]. Immunocytochemical and ELISA studies showed that all N- and C-terminal receptor fragments capable of forming functional receptor complexes in the cotransfection experiments (see above) were stably inserted into lipid bilayers, even when expressed alone. Similar to the wild type m3 mAChR, the various receptor polypeptides were found both intraceUularly (ER/Golgi complex) as well as in the plasma membrane [20]. These observations suggest that the presence of the full-length receptor protein is not essential for membrane insertion and proper intracellular tr~fScking. Taken together, these results strongly support the notion that mAChRs and, most likely, other GPCRs [21] are composed of multiple (rather than only two) individually stable, autonomous folding domains. The folding and assembly of GPCRs therefore appears to occur via a two-step mechanism, similar to that previously described for the bacterial membrane protein, bacteriorhodopsin [22]. In step I, individually stable folding units are established across the lipid bilayer; in step II, these domains interact with each other to form functional receptor complexes.
35 [3H]NMS
m3 (wt)
Ni2 ~
Jill
No3
N i 3 ~
!111Oq + fi::!::iii::!ii!illi||ill::i::iii::i::itCi2 i~llh qlll I
:::::::::::::::::::::::::::::::::::::::::::::::::::: Co3
+
,-il--
+
I::i::i::i::~i::i:J Ci3
Bmax (fmol/mg) KD (pM)
4-DAMP K i (nM)
Carbachol K i (~M)
Acetylcholine K i (~M)
960+120
12+-3
0.47 + 0.03
7.28+0.88
1.40+0.17
44+3
28+6
1.43+0.13
35.0 + 3.7
11.0+2.1
52 + 16
43 + 7
9.20 + 0.48
33.9 +__3.6
15.4 + 0.8
22+__2
0.92+0.08
6.94+0.44
1.16 + 0.04
122+7
Figure 3. Ligand binding properties of polypeptide complexes formed by coexpressed m3 mAChR fragments. All studies were carried out in transiently transfected COS-7 cells (data taken from ref. 20).
100 -
I--
12.
._= ~
80~
o ._ r
60-
40"~"
200
%
%
x
%
%
•
%
Figure 4. Functional properties of polypeptide complexes formed by coexpressed m3 mAChR fragments. The ability of the wild type m3 mAChR (Emax = 100%) and three polypeptide complexes (for fragment structures, see Fig. 3) to mediate carbachol-induced PI hydrolysis was studied in transiently transfected COS-7 cells. For these experiments, the wild type m3 mAChR was expressed at Bmax levels similar to those found with the coexpressed polypeptides (data taken from ref. 20).
36
RECEPTOPJG PROTEIN COUPLING Characteristically, each GPCR can couple only to a limited set of the many structurally similar G proteins expressed in a cell [23]. The same is also true for the individual members of the mAChR family: the ml, m3, and m5 mAChRs are known to preferentially couple to G proteins of the Gq/ll family [2,25,27] which mediate the activation of different isoforms of PLC~ (biochemical response: PI hydrolysis) [24,26,27]. The m2 and m4 mAChRs, on the other hand, are selectively linked to G proteins of the Gi]o class [25,28] which, at a biochemical level, mediate the inhibition of adenylyl cyclase [27].
Receptor residues/domalns critical for G protein coupling selectivity Studies with chimeric m2/m3 mAChRs have shown that the N-terminal portion of the i3 loop is intimately involved in G protein recognition [29-31]. When this receptor segment was exchanged between the m2 and m3 mAChRs, the resultant hybrid receptors gained the ability to interact with the same type of G proteins as the wild type receptor from which this short sequence was derived [29-31], although with clearly reduced efficiency [30]. It was also noted that a m u t a n t m2 mAChR which contained m3 mAChR sequence at the beginning of the i3 loop still retained the ability to inhibit adenylyl cyclase [30], suggesting that the N-terminal segment of the i3 loop is an important but clearly not the sole structural element determining the selectivity of mAChR/G protein interactions (see below). Computational methods [32] as well as site-directed mutagenesis studies [3338] suggest that the N-terminal portion of the i3 loop of mAChRs and other GPCRs forms an amphipathic a-helical extension of TM V (shown for the rat m3 mAChR in Fig. 5) and that the noncharged side of this helical segment represents an important G protein binding surface. Detailed mutational analysis of this segment of the m3 mAChR showed that a Tyr residue (which is conserved among the m l , m3, and m5 mAChRs; m3Tyr254 in Fig. 5) is of particular importance for efficient Gq recognition [35,36]. We could show, for example, that substitution of Tyr254 into a mutant m3 mAChR in which the first 16 ~mino acids of the i3 loop were replaced with the corresponding m2 mAChR sequence (CR1; Fig. 6) was able to confer on this mutant receptor (which does not couple to Gq) the ability to efficiently activate the PI pathway (Fig. 6; ref. 36). However, when Tyr254 was substituted into the homologous position of the wild type m2 mAChR, the resulting m2(Ser210->Tyr) did not gain the ability to efficiently couple to Gq (Fig. 6; ref. 36), thus providing additional support for the notion that other intracellular domains, besides the N-terminus of the i3 loop, also contribute to the specificity of mAChR]G protein interactions. To identify such regions, we systematically substituted distinct intracellular domains of the m3 receptor into the m2(Ser210->Tyr) mutant receptor (= m2Y) and into the wild type m2 receptor [39]. As shown in Fig. 7, introduction of the i l loop or the C-terminal tail of the m3 receptor into the wild type m2 or the m2Y mutant receptor did not lead to a significant stimulation of PLC activity (PI hydrolysis). In contrast, substitution of the i2 loop of the m3 mAChR into the wild type m2 receptor (as well as into m2-Y) led to a hybrid receptor which could stimulate PI hydrolysis in a fashion similar to a mutant m2 receptor (m2-Ni3) in which the
37 K262 K255 E2~
259
A265
R252 E263
T261
E256 i/01
L264 1253
, i--~0
Figure 5. Helical wheel projection of the N-terminal segment of the i3 loop of the rat m3 mAChR (Arg252-Ala265), viewed from the N- to the C-terminus. This model predicts that one side of this putative helix consists of exclusively charged residues (plain), whereas the other side is formed by exclusively noncharged residues (bold). Tyr254 (boxed) is conserved among all PI-coupled mAChRs (ml, m3, and m5) and is required for efficient activation of G proteins of the Gq/ll family [35,36]. N-terminal 21 ~mino acids of the i3 loop were replaced with m3 sequence (Emax ca. 40% of wild type m3; Fig. 7). This finding clearly indicates that the i2 loop also makes a critical contribution to proper recognition of Gq/ll proteins. Introduction of the C-terminal 30 amino acids of the i3 loop of the m3 receptor (Lys464-Ser493) into the wild type m2 mAChR (resulting in m2-Ci3) did not improve coupling to Gq/ll proteins. However, introduction of the additional Ser210->Tyr point mutation into this chimeric construct (resulting in m2-Y-Ci3) led to a mutant receptor which, similar to m2-i2 and m2-Ni3, gained coupling to PI turnover (Fig. 7). This result suggested that the N-terminus of the i3 loop (where m3Tyr254 is located) acts in a cooperative fashion with a region at the C-terminus of the same loop to allow proper recognition of Gq/ll proteins. Following these initial results, detailed site-directed mutagenesis studies were carried out to identify individual amino acids within the i2 loop and the C-terminal segment of the i3 loop of the m3 mAChR required for efficient coupling to Gq [39]. We found that substitution of Ser168, Argl71, Arg176, and Arg183 (residues located in the i2 loop of the m3 receptor; Fig. 8A) into the wild type m2 mAChR yielded a mutant receptor that showed functional properties very similar to the m2-i2 mutant receptor (Fig. 8B). These four amitm acids therefore fully account for the contribution of the i2 loop of the m3 mAChR to Gq coupling efficiency. Single amino acid substitutions showed that m3Arg176 which is located in the middle of the i2 loop is of particular importance for Gq coupling (Fig. 8B). However, experiments with double and triple point mutants (data not shown) suggested that all four m3 receptor residues are required for optimum coupling to Gq [39].
38 I
I m2
CR1
~
m3
CR1-Y
m2(Ser210->Tyr)
12o- I 9 m3(wild type) [] CR1-Y
E-Q 80 (D (D
>
(!) (~ f~
co~
o m2(wiid type) 9 m2(Ser210->Tyr) .. CR1
40
-8
-7
-6
-5
-4
-3
-2
Carbachol, log M
Figure 6. Importance of Tyr254 (rat m3 receptor sequence) for mAChR -mediated stimulation of PI hydrolysis [35,36]. All studies were carried out with transiently transfected COS-7 cells. In CR1, the first 16 amino acids of the i3 loop of the r a t m3 mAChR (RIYKETEKRTKELAGL) were replaced with the corresponding human m2 receptor sequence (HISRASKSRIKKDKKE). In CR1-Y and m2(Ser210->Tyr), a Tyr residue (corresponding to Tyr254 in the rat m3 mAChR) replaces Ser210 located at the N-terminus of the i3 loop of the m2 mAChR. The maximum stimulation of PI hydrolysis mediated by the wild type m3 receptor was set at 100%. The C-terminal segment of the i3 loop was analyzed in a similar fashion to identify additional amino acids required for efficient coupling to Gq. Substitution of Ala488, Ala489, Leu492, and Ser493 into m2(Ser210->Tyr) resulted in a m u t a n t receptor which quantitatively mimicked the PI response mediated by the m2-Y-Ci3 hybrid receptor (Figs. 7, 9). More detailed mutagenesis studies (Fig. 9B; ref. 39) showed that all four residues (which are located at the i3 loop/ TM VI junction) are required for optimum Gq activation. Secondary structure prediction programs [32] suggest that the region at the i3 loopfrM VI junction of mAChRs and other GPCRs is a-helically arranged. If this is correct, m3Ala488, m3Ala489, m3Leu492, and m3Ser493 are located on one side of an a-helical receptor domain (Fig. 10A). Based on this notion, there are two patches of primarily hydrophobic amino acids located at the N- and Ctermini of the i3 loop, respectively. Given the predicted spatial proximity of the N- and C-terminal segments of the i3 loop, it is therefore likely that these two
39
il I
i2 II
III
Max.
i4 IV
i n c r e a s e in I P1 (% above basal)
VI VII
v
m
m
+++
m3 m2
.
m2-Y
f
..,
~ ,
--{ -
rn2-il m2-Y-il m2-i2
..~ i
.: ~
:'i' I
~
.
.;
.u
i
m2-Y-i2
~
m2-Ni3
~
m2-Ci3 m2-Y-Ci3
~ ,
m2-i4
~
m2-Y-i4
~
|
,_
I
(+) (+) (+) (+) ++ ++ ++
I ,L
Y
I
(+) ++
(+) (+)
Y (+) + + +++
< 15 % 40-60 % 100%
Figure 7. Carbachol-induced PI hydrolysis mediated by hybrid m2/m3 mAChRs transiently expressed in COS-7 cells. The Tyr residue (Y) present at the beginning of the i3 loop in several of the chimeric constructs corresponds to m3Tyr254 and replaces m2Ser210 (data taken from ref. 39). receptor sites form a contiguous hydrophobic binding surface critical for G protein recognition and activation. Additional mutagenesis studies [39] with mutant m2 mAChRs containing m3 receptor sequences in multiple intracellular domains showed that the residues important for Gq coupling (located in the i2 loop and at the N- and C-termin_i of the i3 loop of the m3 mAChR; see above) act in a cooperative fashion to allow efficient Gq activation. It was found that Ser168, Argl71, Arg176, Arg183 (present in the i2 loop of the m3 mAChR), together with Ala488, Ala489, Leu492, and Ser493 (present at the i3 loopfrM VI junction of the m3 mAChR) and the Nterminal segment of the i3 loop, quantitatively account for the G protein coupling preference of the m3 mAChR (Fig. 11). These residues are conserved among all Gq-coupled mAChRs (ml, m3, and m5; Figs. 8A, 9A) but replaced with different residues in the m2 and m4 receptors. It will be of interest to examine whether amino acids at the corresponding positions in other GPCRs have a similar
40
A
i2
ml m5 mj
DRY~LSY~RTP~ DRY~S~~LT~KRTP~ DRY~~LTY~AKRTT~
m4
120]
I"
1 00 -=
e-
"7~
r
o e-
~ v
t-~
171
176
183
127
132
1~
801 / 9
r
168
124
DRY~LT~RRTT~ ****# *#** *#
B n
164 120
60
**
#
m3 residues substituted into the m2 mAChR
s=serlosR i2 = Asp164-Arg 183
R. = Arg171 = Arg176 R** = Arg 183
40 20
Figure 8. Four 3mino acids in the i2 loop of the m3 mAChR are critical for receptor-mediated PI hydrolysis [39]. (A) Amino acid sequences of the i2 loop of the ml-m5 mAChRs (rat=human). *, Positions at which all five receptors have identical residues. #, Positions at which the ml, m3, and m5 receptors have identical residues (boxed) which differ from those present in the m2 and m4 receptors. Numbers refer to amino acid positions in the human m2 and rat m3 mAChRs, respectively [49]. (B) The ability of the wild type m2 and m3 mAChRs (wild type m3: Emax = 100%) and a series of m u t a n t m2/m3 receptors to mediate carbachol-induced increases in IP1 levels was studied in transiently transfected COS-7 cells [39].
41
A
VI
ml
I-T-1 I-T--I R A G K G Q K P R O K E Q L A K R K T F S a V E m K ~ k ~ L : ~ . T ~ ~z~.I a n iii iii
m3
L~R-
m2
V A R K - IVKMTK- Q PAKKKP- P P S R E K K ~ ~ T ~
m5
m4
B
TM
Ci3
FALKTRSQ I TKRK~'VlSL mK E K ~ i Q ~ I ~ ~ A r LL
I I I
V~K-
I I
AILL
FAS I~e~QVRKKRQM- ~ R E R ~ I R T i Z l ~ A I EL * *# #* * # # * # # * * * *
120 -]
m3 residues substituted into the m2 mAChR
1
100 c- ~
a~ ~ m o -g ~ o~ ~'~
"
Y = Tyr254
8060 40
" .
~
-T~ ~
--]~
Ci3 = A A*= L = S =
Lys464-Ser493 Ala488 Ala489 Leu492 Ser493
20
Figure 9. Four amino acids at the C-terminus of the i3 loop of the m3 mAChR play an important role in receptor-mediated PI hydrolysis [39]. (A) Amino acid sequences of the C-terminal portion of the i3 loop of the ml-m5 mAChRs (except for m3 [rat], human sequences are shown). Boxed residues were targeted by sitedirected mutagenesis [39]. For additional explanations, see legend to Fig. 8. (B) see Fig. 8B (data taken from ref. 39).
42
494 "k41~7 490
B
4~l.k
.384
~
)2 "k
4t
O
0
488
493
O 489
385
4~6~r 492
390
O 386
393 ~'L
389
Figure 10. Helical wheel models of the m3 (A) and m2 mAChR (B) sequences located at the i3 loopfrM VI junction (for linear sequences, see Fig. 9A). The direction of view is from the N- to the C-terminus. Residues highlighted in black are thought to be critically involved in recognition of Gq/ll (m3 mAChR; ref. 39) and Gi/o (m2 mAChR; ref. 46), respectively. *, Positions at which all five mAChRs have identical residues.
I
A F K V N K Q L
II
N N V T K
III
y F
IV
RD
(~K 183 T T
. E:
1~O
IT
171
V
O 172
IR 9
T
254
48~(i~
o A K Te 9
P L T Y 176
i2
VI
K'. EL
FT
E
i3 eOeeooee272 S
K
N
RTTFKT
K
E.
R~ T 9
VII
K I L
AG L Q A S GT - " 2 0 8 a a
LCQ LL 25 aa
i4
OI A
,,~ (COoH)
Cytoplasm
Figure 11. Residues in the m3 mAChR (rat) critically involved in selective recognition of Gq/ll [39]. Only the intracellular receptor regions and a few ~mino acids of TM I-VII are shown. Besides the nine ~mino acids highlighted in black, optimum activation of Gq/ll proteins appears to require one or more additional residues in the N-terminal portion of the i3 loop (stippled sequence; ref. 39).
43 functional role in determining the selectivity of G protein recognition as described here for a Gq-coupled mAChR. I d e n t i f i c a t i o n of a receptor/G p r o t e i n c o n t a c t site To better understand how receptors and G proteins interact with each other at a molecular level, the regions on the G proteins with which the various intracellular receptor domains (see above) can functionally interact need to be identified. Biochemical studies suggest that at least three distinct regions on the GTPase domain of the G protein a-subunits, including the N- and the C-terminus, can directly contact the receptor [40,41]. These regions clearly differ from those predicted to be involved in the activation of effector enzymes such as adenylyl cyclase [42] or phospholipase C [43]. The C-terminus of the G protein a-subunits (Ga) is known to play a key role in receptor/G protein interactions [40,41,44]. Con]din et al. [45] recently showed that m u t a n t versions of Gaq in which the last five amino acids of aq were replaced with the corresponding sequences derived from different members of the Gai/o protein family (ai2, ao, or az) allowed stimulation of PLC by receptors (A1 adenosine and D2 dopamine receptors) that otherwise are exclusively coupled to G proteins of the Gi]o class which cannot activate PLC efficiently. Recently, we obtained essentially similar results with the m2 mAChR (Fig. 12; ref. 46). In COS-7 cells, the m2 mAChR, a prototypical Gi/o-coupled receptor, stimulates PI hydrolysis only poorly, even in the presence of overexpressed wild type Gaq (q(wt); Figs. 12, 13). However, coexpression of the m2 receptor with qi5 or qo5 (mutant Gaq-subunits in which the last five amino acids of q(wt) were replaced with the corresponding ai2 or ao sequences, respectively) led to a pronounced stimulation (4-8-fold) of PLC activity (Figs. 12, 13; ref. 46). The specificity of this interaction could be demonstrated by the relative lack of PLC activity observed after coexpression of the m2 receptor with a mutant aq-subunit (qs5) that contained five amino acids of as sequence at its C-terminus [46]. Taken together, these results suggest that the m2 mAChR as well as other Gi/o-coupled receptors contain a structural motif that can specifically recognize the C-terminus of Gai/o-subunits and that this interaction is critical for the specificity of receptor/ G protein interactions and G protein activation. To identify this receptor site, a series of mutant m2 receptors were created in which distinct intracellular segments were systematically replaced with the corresponding sequences derived from receptors (m3 receptor, Gq-coupled; ~2adrenergic receptor, Gs-coupled) that couple to G proteins different from Gi/o. It seemed reasonable to assume that structural modification of the m2 receptor region(s) that can contact the C-terminus of Gai/o-subunits should prevent m2 receptor-mediated activation of m u t a n t aq-subunits such as qo5 or qi5. We found [46] that the ability of the m2 receptor to productively interact with qo5 or qi5 was not affected by substitutions involving the il and the i2 loops, the Nterminal segment of the i3 domain, or the C-terminal tail (i4). In contrast, replacement of the C-terminal portion of the i3 loop of the m2 receptor (residues 361-390) with the homologous m3 or ~2-adrenergic receptor sequence almost completely abolished qo5/qi5-mediated PLC stimulation [46]. This observation suggested that the C-terminal segment of the i3 loop of the m2 mAChR plays a specific role in the recognition of the C-terminus of Gai/o-subunits. To further test this hypothesis, a series of gain-of-function mutagenesis
44
Receptor
Max. increase in IP1 (above basal)
G protein a-subunit
m2(wt)
+
I
m2(wt)
+
I
II
G a i 2 or G a o sequence
G a q (=
I
q(wt))
< 2-fold
i
qi5 or qo5
4-8-fold
Figure 12. Functional interaction of the wild type m2 mAChR with mutant G protein aq-subunits [46]. COS-7 cells were cotransfected with expression plasraids coding for the wild type m2 mAChR and wild type Gaq (q(wt)) or mutant Gaq-subunits in which the C-terminal five amino acids of Gaq (EYNLV) were replaced with the corresponding sequences from Gai2 (qi5; DCGLF) or Gao (qo5; GCGLY). Cells were then stimulated with the agonist carbachol (1 raM), and increases in intracellular IP1 levels were determined [46].
m2 i i m2 (wt) CR2
'
CR13 CR14 CR15
, , i
CR16
I
Max. increase in IP1 (above basal)
m3 r-~ tl
ill
iv
v
I
'
'
+
i '
qo5 +-I-++
+
+
+
++++
~.
+
++++
l,~, VT~IL
+
+-I--I-+
.,j);
VT CR17
q(wt)
vt vtt
,
~..
+
+
+
+
IL.
Figure 13. Functional interaction of mutant m2/m3 mAChRs with wild type Gaq (q(wt)) or the mutant Gaq-subunit, qo5. COS-7 cells were cotransfected with the indicated mutant receptors and wild type Gaq (q(wt)) or a mutant Gaq-subunit in which the last five amino acids of Gaq were replaced with the corresponding sequence from Gao (qo5; see Fig. 12). Cells were then stimulated with the agonist carbachol (1 raM), and increases in intracellular IP1 levels were determined [46]. The degree of PLC stimulation mediated by the various mutant receptors upon coexpression with q(wt) or qo5 is denoted by + (~2-fold) and ++++ (4-8-fold). In CR15, Va1385, Thr386, Ile389, and Leu390 (m2 receptor sequence) were substituted into CR2 (thus replacing Ala488, Ala489, Leu492, and Ser493, respectively; m3 receptor sequence).
45 studies were designed. Initially, the C-terminal segment of the i3 loop of the m2 mAChR (residues 359-390) was substituted into a mutant m3 receptor, CR2 (Fig. 13), to study whether the resulting hybrid receptor (CR13; Fig. 13) gained the ability to productively interact with qo5. Whereas CR2 was unable to efficiently stimulate PI hydrolysis when coexpressed with either q(wt) or qo5, CR13 gained the ability to productively interact with qo5 (Fig. 13). Further mutational modification of CR13 showed that substitution into CR2 of only four m2 receptor residues (Va1385, Thr386, Ile389, and Leu390), predicted to be located at the i3 loopfrM VI junction, was sufficient to confer on the resulting mutant receptor (CR15) the ability to potently stimulate PLC activity when coexpressed with qo5 (Fig. 13). Coexpression of CR15 with qi5, but not coexpression with q(wt) or qs5, also resulted in a pronounced activation of the PLC pathway [46]. Substitution into CR2 of m2Va1385Thr386 or m2Ile389Leu390 alone resulted in m u t a n t receptors (CR16 and CR17, respectively) which failed to productively interact with qo5 (Fig. 13). Introduction of the %VrIL motif' (m2Va1385, m2Thr386, m2Ile389, and m2Leu390) directly into the wild type m3 receptor (thus replacing m3 receptor residues Ala488, Ala489, Leu492, and Ser493, respectively) yielded a hybrid receptor (m3(AALS->VTIL))which was able to stimulate PI hydrolysis with high efficiency when coexpressed with qo5 or qi5 (5.5_+0.3-fold stimulation of PLC activity; carbachol EC50 = 0.42+0.07 ~M; Fig. 14). In contrast, the interaction of this mutant receptor with q(wt) was clearly less efficient (3.8_+0.3-fold stimulation of PLC activity; carbachol EC50 = 4.7+_1.5 ~M). This finding is consistent with the observation that the "AALS" motif at the C-terminus of the i3 loop of the m3 mAChR is required for efficient coupling to Gq (see above; ref. 39). Moreover, coexpression experiments with wild type Gai2 showed that the m3(AALS->VTIL) m u t a n t receptor also gained the ability to inhibit adenylyl cyclase (29+3% inhibition of stimulated cAMP levels; wild type m2: 41_+4% inhibition) (Fig. 15; ref. 46). Consistent with these gain-of-function experiments, replacement of the '~VTIL motif' in the wild type m2 mAChR with the corresponding m3 receptor residues resulted in a mutant receptor (m2(VTIL->AALS)) that almost completely lost the ability to functionally interact with qo5 (Fig. 16) or to mediate inhibition of adenylyl cyclase (Fig. 15; ref. 46). Taken together, these results strongly support the concept that the '%ZTIL motif' (formed by m2 receptor residues Va1385, Thr386, Ile389, and Leu390) can specifically recognize the C-terminus of Gai/o-subunits and that this interaction is required and sufficient for Gi/o activation. Except for an m2Leu390->Phe substitution, these four amino acids are also present in the Gi/o-coupled m4 mAChR (but not in the Gq-coupled mAChRs; Fig. 9A). In analogy to the proposed arrangement of the corresponding residues in the m3 mAChR (see previous section), Va1385, Thr386, Ile389, and Leu390 (m2 mAChR sequence) are predicted to be located at the i3 loop/TM VI junction on one side of an a-helical receptor segment (Fig. 10B). It is therefore conceivable that these four amino acids form a receptor surface which can recognize the Cterminus of Gai/o-subunits, most likely via hydrophobic interactions. This interaction is likely to induce conformational changes in the C-terminus of the Gai/osubunits which eventually lead to GDP release and G protein activation. This concept is supported by NMR studies on a C-terminal peptide derived from
46 750 " (D (D
m3(AALS->VTIL)
qo5 + qi5
9 + 9
500 12..Q
o
.=_ ~>
+
q(~)
o
250 -
(D L,_
f: v I
-10
I
-8
I
I
-6
I
I
-4
I
I
-2
Carbachol, log (M)
Figure 14. Functional interaction of the m3 (AALS->VTIL) mutant receptor with wild type Gaq (q(wt)) and mutant Gaq-subunits, qo5 or qi5 (see Fig. 12). In the m3 (AALS->VTIL) mutant receptor, Ala488, Ala489, Leu492, and Ser493 (m3 receptor sequence) were replaced with the corresponding residues present in the m2 mAChR (Va1385, Thr386, Ile389, and Leu390, respectively). COS-7 cells coexpressing the m3 (AALS->VTIL) mutant receptor and wild type Gaq (q(wt)) or mutant Gaq-subunits, qi5 or qo5 (see Fig. 12), were incubated with the indicated concentrations of carbachol, and increases in intracellular IP1 levels were determined. Data are taken from a representative experiment [46]. 5O "~
40
~~
3o
8-~ 20 ~AALS) ->VTIL)
Figure 15. Inhibition of adenylyl cyclase by wild type and mutant m2/m3 mAChRs. COS-7 cells cotransfected with mAChR DNA and plasmids coding for the V2 vasopressin receptor and wild type Gai2 were studied for their ability to mediate carbachol-induced (0.1 raM) inhibition of [Arg]vasopressin-stimulated cAMP levels [46]. The maximum stimulation of cAMP levels induced by [Arg]vasopressin (0.5 nM) amounted to 13+3-fold above basal levels (100 %). In the m2(VTIL->AALS) and m3(AALS->VTIL) mutant receptors, Va1385, Thr386, Ile389, and Leu390 (m2 receptor sequence) and Ala488, Ala489, Leu492, and Ser493 (m3 receptor sequence) were exchanged between the two wild type receptors. Data are taken from a representative experiment [46].
47 IP1 levels 16
[-7
-
Control (no carbachol) + 1 mM Carbachol
|
o12 X
8
E d. d
4-
0
"
m2(wt) + qo5
m2(VTIL->AALS) + qo5
Figure 16. Functional interaction of the wild type m2 mAChR and the m2(VTIL>AALS) m u t a n t receptor with the mutant Gaq-subunit, qo5 (see Fig. 12). In the m2(VTIL->AALS) m u t a n t receptor, Va1385, Thr386, Ile389, and Leu390 (m2 mAChR sequence) were replaced with the corresponding residues present in the m3 mAChR (Ala488, Ala489, Leu492, and Ser493, respectively). COS-7 cells coexpressing the m2(VTIL->AALS) mutant receptor and the m u t a n t Gaq-subunit, qo5 (see Fig. 12), were incubated with 1 mM carbachol, and increases in intracellular IP1 levels were determined. Data are taken from a representative experiment [46]. at (transducin; another member of the Gai/o protein family) demonstrating that the last four amino acids of Gai/o-subunits form a type II' ~-turn (centered around a glycine residue at position -3 which is specific for all members of the Gai/o family) which is broken upon interaction with the ligand-occupied receptor [47]. In conclusion, this is the first report describing a specific, functionally relevant site of interaction between a GPCR and its cognate Ga-subunits. The receptor/G protein contact site described here may serve as a useful anchoring point for the delineation of three-dimensional models of the receptor/G protein interface. It should be borne in mind, however, that optimum_ receptor/G protein coupling is thought to involve several intracellular receptor domains (see above) and at least three sites on the G protein a-subunits (including the C-terminus; refs. 40,41,44). It is therefore likely that additional interactions, besides the one described above, modulate the specificity of receptor/G protein coupling and further increase the efficiency of G protein activation. It will be interesting to examine whether GPCRs that couple to G proteins different from Gi/o operate through a molecular mechanism similar to that described here for the m2 mAChR.
48 REFERENCES
1 Hulme EC, Birdsall NJM, Buckley NJ. Annu Rev Pharmacol Toxicol 1990; 30:633-673. 2 Wess J. Trends Pharmacol Sci 1993; 14:308-313. 3 Schertler GFX, Villa C, Henderson R. Nature 1993; 362:770-772. 4 Baldwin JM. EMBO J 1993; 12:1693-1703. 5 Baldwin JM. Curr Opin Cell Biol 1994; 6:180-190. 6 Schwartz TW. Curr Opin Biotechnol 1994; 5:434-444. 7 Fraser CM, Wang C-D, Robinson DA, Gocayne JD, et al. Mol Pharmacol 1989; 36:840-847. 8 Wess J, Gdula D, Brann, MR. EMBO J 1991; 10:3729-34. 9 Curtis CAM, Wheatley M, Bansal S, Birdsall NJM, et al. J Biol Chem 1989; 264:489-495. 10 Spalding TA, Birdsall NJM, Curtis CAM, Hulme EC. J Biol Chem 1994; 269:4092-4097. 11 Suryanarayana S, von Zastrow M, Kobilka BK. J Biol Chem 1992; 267:21991-1994. 12 Pittel Z, Wess J. Mol Pharmacol 1994; 45:61-64. 13 Zhou W, Flanagan C, Ballesteros JA, Konvicka K, et al. Mol Pharmacol 1994; 45:165-170. 14 Rao VR, Cohen GB, Oprian DD. Nature 1994; 367:639-642. 15 Elling CE, Nielsen SM, Schwartz TW. Nature 1995; 374:74-77. 16 Liu J, Sch~ineberg T, van Rhee M, Wess J. J Biol Chem 1995; 270:1953219539. 17 Kobilka BK, Kobilka TS, Daniel K, Regan JW, et al. Science 1988; 240:1310-1316. 18 Maggio R, Vogel, Z, Wess J. FEBS Lett 1993; 319:195-200. 19 Maggio R, Vogel, Z, Wess J. Proc Natl Acad Sci USA 1993; 90:3103-3107. 20 Sch/ineberg T, Liu J, Wess J. J Biol Chem 1995; 270:18000-18006. 21 Ridge KD, Lee SSJ, Yao LL. Proc Natl Acad Sci USA 1995; 92:3204-3208. 22 Popot J-L, Engelman DM. Biochemistry 1990; 29:4031-4037. 23 Hedin, KE, Duerson, K, Clapham, DE. Cell Sig 1993; 5:505-518. 24 Berstein G, Blank JL, Smrcka AV, Higashijima T, et al. J Biol Chem 1992; 267:8081-8088. 25 Offermanns S, Wieland T, Homann D, Sandmann J, et al. Mol Pharmacol 1994; 45:890-898. 26 Smrcka AV, Hepler JR, Brown KO, Sternweis PC. Science 1991; 251:804807. 27 Peralta EG, Ashkenazi A, Winslow JW, Ramachandran J, et al. Nature 1988; 334:434-437. 28 Parker EM, Kameyama K, Higashijima T, Ross EM. J Biol Chem 1991; 266:519-527. 29 Wess J, Brann MR, Bonner TI. FEBS Lett 1989; 258:133-136. 30 Wess J, Bonner TI, D6oe F, Brann MR. Mol Pharmacol 1990; 38:517-523. 31 Lechleiter J, Hellmiss R, Duerson K, Ennulat D, et al. EMBO J 1990; 9:4381-4390. 32 Strader CD, Sigal IS, Dixon RAF. FASEB J 1989; 3:1825-1832.
49 33 Cheung AH, Huang R-RC, Strader CD. Mol Pharmacol 1992; 41:10611065. 34 Arden JR, Nagata O, Shockley MS, Philip M, et al. Biochem Biophys Res Commun 1992; 188:1111-1115. 35 Blfiml K, Mutschler E, Wess J. J Biol Chem 1994; 269:402-405. 36 Blfiml K, Mutschler E, Wess J. J Biol Chem 1994; 269:11537-11541. 37 Bliiml K, Mutschler E, Wess J. Proc Natl Acad Sci USA 1994; 91:79807984. 38 HSgger P, Shockley MS, Lameh J, Sadee W. J Biol Chem 1995; 270:74057410. 39 Blin N, Yun J, Wess J. J Biol Chem 1995; 270:17741-17748. 40 Harem HE, Deretik D, Arendt A, Hargrave PA, et al. Science 1988; 241:832-835. 41 Co~klin BR, Bourne HR. Cell 1993; 73:631-641. 42 Berlot CH, Bourne HR. Cell 1992; 68:911-922. 43 Arkinstall S, Chabert C, Maundrell K, Peitsch M. FEBS Lett 1995; 364:45-50. 44 Rasenick MM, Watanabe M, Lazarevic MB, Hatta S, et al. J Biol Chem 1994; 269:21519-21525. 45 Conklin BR, Farfel Z, Lustig KD, Julius D, Bourne HR. Nature 1993; 363:274-276. 46 Liu J, Conklin BR, Blin N, Yun J, Wess J. Proc Natl Acad Sci USA (in press). 47 Dratz EA, Furstenau JE, Lambert CG, Thireault DL, et al. Nature 1993; 363:276-281. 48 Matsui H, Lazareno S, BirdsaU NJM. Mol Pharmacol 1995; 47:88-98. 49 Bonner TI, Buckley NJ, Young AC, Brann MR. Science 1987; 237:527-532.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardin~, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
51
Muscarinic Receptor Subtypes - Search for Selective Agonists and Antagonists
E. Mutschler a, H. A. Ensingerb, J. Gross a, A. Leis c, K. Mendla b, U. Moser a, O. Pfai~, D. Reichel d, K. ROhlmannc, R. Tacke d, M. Waelbroecke, J. Wehrle a and G. Lambrecht a aDepartment of Pharmacology, Biocentre Niederursel, University of Frankfiart, Marie-CurieStr. 9, D-60439 Frankfurt/Main, Germany bBoehringer Ingelheim KG, Binger Str., D-55216 Ingelheim, Germany Clnstitute of Inorganic Chemistry, Technical University of Dresden, Mommsen-Str. 13, DO1069 Dresden, Germany dInstitute of Inorganic Chemistry, University of Wiarzburg, Am Hubland, D-97074 W0rzburg, Germany eDepartment of Biochemistry and Nutrition, Medical School, Free University of Brussels, B1070 Brussels, Belgium ABSTRACT Since the late eighties five muscarinic receptor subtypes (ml - m5) have been cloned and four of them (M 1 - M4) have also been pharmacologically characterized. However, there is still a lack of potent muscarinic agonists and antagonists, which are highly selective for one muscarinic receptor subtype over all other subtypes. For the treatment of Alzheimer's disease, Ml-selective agonists capable of penetrating into the CNS are needed. It is hypothezised that such substances would not only improve memory and cognitive ability, but also delay the progression of the disease. In our laboratory, the functionally M 1-selective quaternary ammonium compound McN-A-343 has been used as a starting point for the design of such CNS active muscarinic ligands. Structure-activity relationship studies led to the tertiary amine 4-(4-fluorophenylcarbamoyloxy)-2-butynylpyrrolidine (4-FPyMcN), which was found to stimulate M 1 receptors with some functional selectivity. In order to increase the potency and selectivity of 4-F-PyMcN several new derivatives were synthezised and pharmacologically characterized in different functional assays as well as in binding and biochemical (PI turnover) studies. The most promising results were obtained with (S)4-(4-fluorophenylcarbamoyloxy)-l-methyl-2-butynylpyrrolidine (4-F-MePyMcN). Due to its potent partial agonistic activity at M 1 receptors and its M2-antagonistic properties leading to an increase of acetylcholine release by blockade of M2 autoreceptors, this compound may be considered as an important tool for future drug research of cognitive disorders. M 2 receptor antagonists may also be used for the treatment of Alzheimer's disease, furthermore in the therapy of supraventricular bradycardia and for quantifying M2 receptors in the CNS with PET imaging. In the search for antagonists which clearly differentiate M 2 from other muscarinic receptors, we investigated the two enantiomers of the widely used Hi-antihistaminic drug dimethindene. (S)-Dimethindene proved to be a potent M2-selective antagonist with lower affinities for the M1, M 3 and M4 receptors. In addition, the (S)-enantiomer
52 was more potent than the (R)-enantiomer in all muscarinic assays. Interestingly, the stereoselectivity was inverse at histamine H1 receptors, the (R)-enantiomer being the eutomer. M 3 receptor antagonists may be useful in the treatment of spastic disorders of the gastrointestinal, urogenital and respiratory tract as well as for the relief of glandular hypersecretion. In previous studies, hexahydro-difenidol (HHD) and its sila-analogue, hexahydro-sila-difenidol (I-~SiD), as well as the antiparkinsonian drug trihexyphenidyl (THP) were found to be valuable tools for the discrimination of M 3 and M 2 receptors. In order to further assess the structural requirements (including stereochemical aspects) of the above-mentioned compounds for potency and selectivity, a series of ttHD and THP analogues as well as of the corresponding silicon and germanium derivatives (sila- and germa-substitution) were studied. The (R)-enantiomers displayed higher affinities and selectivities than the corresponding (S)-isomers. The enantioselectivity of some of these analogues is best explained by the concept of the fourbinding-subsite model suggesting that the differences in affinity of the (R)- and (S)-enantiomers at muscarinic receptors are due to opposite binding of the phenyl and the cyclohexyl ring to the preferring subsites. Surprisingly, there was no significant difference between the Si and Ge analogues indicating a strongly pronounced Si/Ge bioisosterism in this series of compounds. The related carbon derivatives, however, showed higher receptor affinities as well as greater stereoselectivities at all muscarinic receptors studied compared with the silicon and germanium analogues. INTRODUCTION Four muscarinic receptor subtypes (M 1 - M4) have been defined by pharmacological studies. On the other hand, receptor cloning studies revealed the existence of five different but highly homologous muscarinic receptor gene products (ml - m5). The pharmacological classification of muscarinic receptor subtypes is mainly based on the relative sensitivities of the receptor subtypes to key muscarinic antagonists such as pirenzepine (M1 > M4 > M3 > M2), 11-[[2-[(diethylamino)methyl]- 1-piperidinyl]acetyl]-5,11-dihydro6H-pyrido-[2,3-b][1,4]-benzodiazepin-6-one (AF-DX 116) and related compounds, methoctramine and himbacine (M2 > M 4 > M 1 > M3) as well as hexahydro-sila-difenidol and its pfluoro derivative (M 3 > M 1 > M 4 > M2) [for recent reviews see 1-4]. In addition, muscarinic receptors may be differentiated on the basis of their stereoselectivity to chiral antagonists [57]. However, it should be noted that in contrast to antagonists of other neurotransmitters, e.g. of histamine or serotonin, most of these antimuscarinic agents are not highly selective for one receptor subtype over all other subtypes. Thus, the search for subtype-selective muscarinic and antimuscarinic ligands is still of great importance. Ml-selective agonists capable of penetrating into the CNS may be important for the treatment of Alzheimer's disease [8-10]. It is hypothezised that such substances would not only improve memory and cognitive ability, but also delay the progression of the disease by enhancing the formation of the neuroprotective secreted amyloid precursor protein (APPs) and by inhibiting the production of the neurotoxic amyloid B-protein [11-13]. M 2 receptor antagonists may be applied also for the treatment of Alzheimer's disease [9, 14-16], furthermore in the therapy of supraventricular bradycardia [17-19] and for quanti~ing M2 receptors in the CNS with PET imaging [20].
53
M 3 receptor antagonists may be useful in the treatment of spastic disorders of the gastrointestinal, urogenital and respiratory tract as well as for the relief of glandular hypersecretion [21-241. In the following we will concentrate on Ml-selective agonists as well as on M 2- or M3-selective antagonists, respectively. METHODS
Agonism and antagonism of the compounds for muscarinic receptors have been measured in functional and binding experiments using the preparations listed below:
Functional experiments (pD 2 and pA2 values) M1 :Rabbit vas deferens (RVD) [25] Rat duodenum (RD) [26] M2: Guinea-pig left atria (GPA) Rabbit vas deferens [27] M3: Guinea-pig ileal longitudinal muscle (GPI) Guinea-pig trachea (GPT) [28]
Binding experiments (pK i values) Binding affinities were determined in NB-OK 1 cells (M 1 receptors) and rat heart (M2 receptors), pancreas (M 3 receptors) and striatum (M4 receptors) homogenates using [~H]-Nmethylscopolamine as radioligand [29]. Binding affinities of the compound (S)-F-MePyMcN were measured in membrane preparations from CHO-cells transfected with human ml to m4 receptors using [3H]-N-methylscopolamine as radioligand, too. The assay was carried out according to [7].
Biochemical experiments PI turnover was measured in CHO-cells transfected with human ml receptors [30]. RESULTS AND DISCUSSION
M 1 agonists with additional M2-antagonistic properties~ M1 receptors are particularly enriched in two regions of the central nervous system, the cortex and the hippocampus, known to play an important role in learning and memory processes. Activation of these postsynaptically located muscarinic receptors causes cellular excitation. In contrast, M 2 receptors appear to be most often expressed as presynaptic autoreceptors mediating the inhibition of acetylcholine release [31 ]. Based on this knowledge, an ideal compound for the treatment of Alzheimer's disease would act as a selective postsynaptic M1 agonist and as a selective presynaptic M 2 antagonist. The functional Ml-selective agonist McN-A-343 (Figure 1) has been used in our laboratory as a starting point for the design of CNS-active muscarinic ligands, the primary objective being
54 to replace the quaternary ammonium group of McN-A-343 by a tertiary amino moiety. The chemical structures of the new compounds are given in Figure 1.
X , ~ H
0 II
R1 I
N-C-O-C-C-C-CH2-Am I
H
R1 = H, CH 3, C2Hs
X 3-CI, -F
Am (R2= H, CH3) *NR2(CH3)2 /\
4-Br, -CI, -F
+NR2CH3C2Hs
3,4-dichloro
*NR2(C2Hs)2
Figure 1. Chemical structure of tertiary and quaternary analogues of McN-A-343 [X = 3-C1, Am = +N(CH3)3, R1 = H], 4-F-MePyMcN +" X = 4-F, Am = N-methylpyrroli-dinium, R 1 = CH3; 4-F-MePyMcN: X = 4-F, Am = pyrrolidino, R 1 = CH3; 4-CI-McN-A-343" X = 4-C1, Am = +N(CH3)3, R 1 = H; 4-F-PyMcN: X = 4-F, Am = pyrrolidino, R 1 = H; 4-F-PyMcN+: X = 4-F, Am = N-methylpyrrolidinium, R1 = H. The functional investigations of these compounds revealed that all agonist responses were muscafinic in nature in that their activities were blocked by: pirenzepine (0.1 ~tM; pA 2 = 8.14 8.51) in rabbit vas deferens (M1), AQ-RA 741 (100 - 300 riM; pA 2 = 8.31 - 8.49) in guineapig atria (M2) and p-F-HHSiD (0.5 ~tM, pA 2 = 7.51 - 7.91) in guinea-pig ileum (M3). Tetrodotoxin (0.1 ~tM) and hexamethonium (100 ~tM) did not block agonist activities in guinea-pig atria and ileum. The potency (pEC50), apparent efficacy (a.e.) and affinity (PA2) for McN-A343 and some of its analogues are shown in Table 1. From these data it becomes evident that the effects of McN-A-343 and of most of its analogues differ clearly in the three functional assays. McN-A-343 and its 4-chloro derivative had comparable potency in rabbit vas deferens, whereas in both atria and ileum they behaved as partial or full agonists, being more potent and more efficacious in the M 3 assay. The incorporation of chirality into this series of acetylenic McN-A-343 derivatives had a beneficial effect upon the potency (affinity) and muscafinic receptor subtype-selectivity, the (S)-enantiomers consistently showing higher activity than the corresponding (g)-isomers.
55 Table 1
In vitro functional activity of McN-A-343 analogues at M 1 receptors in rabbit vas deferens (RVD), M 2 receptors in guinea-pig atria (GPA) and M 3 receptors in guinea-pig ileum (GPI). Abbreviations for the muscarinic agems are explained in the legend of Figure 1 RVD/M 1 pEC50 a.e. a
pA 2
GPA/M 2
GPI/M 3
pECs0 a.e.b-- pA 2 '
pEC50 a.e.--c
pA2
5.51g 5.71 . -
5.76_h 6.42_.h_h 5.29 h 6.82h
i
McN-A-343 d 4-CI-McN-A-343 d (R)-a-F-MePyMcN + (S)-4-F-MePyMcN + (R)-4-F-MePyMcN (S)-4-F-MePyMcN
6.77 e 7.13 6.37 7.25 7.22
1.00 1.00 0.27 0.25 0.83
-
5.65 h 7.26_.h_h 5.41 h 7.11h
4.87-I5.26 -
0.49 0.77 _ _ -
5.90 h 6.33 h 5.60_h 7.39h--
0.83 1.00 _ _ -
a,b_,_c Apparent efficacy: the maximum response to McN-A-343 a and to arecaidine propargyl esterb-,-c = 1.00. d Data taken from Lambrecht et al. [32]. e pK A (= 5.17) was estimated using irreversible antagonism [33]. _f,g pK A values determined by the technique of Waud [34] and using arecaidine propargyl ester as full agonist, were: M 2 = 4.79, M 3 = 5.17. h Slopes of Schild plots were not significantly different from unity (P>0.05). Among the amino terminally modified analogues of McN-A-343, the tertiary amine, (S)-4F-MePyMcN, was a potent partial agonist at M 1 receptors in rabbit vas deferens. In contrast, at M 2 receptors in atria and at ileal M 3 receptors (S)-4-F-MePyMcN lacked efficacy and was shown to act as a competitive antagonist with pA 2 values of 7.4 and 6.8, respectively. Unexpectedly, the corresponding quaternary analogue, (S)-4-F-MePyMcN +, was found to be a relatively potent M 1-selective antagonist. The (R)-enantiomers of 4-F-MePyMcN and 4-FMePyMcN + were weak non-selective muscarinic antagonists (pA 2 values = 5.3 - 5.9). The stereoselectivity ratios (= antilogs of the difference of respective pA 2 values) for 4-FMePyMcN at M 1, M 2 and M 3 receptors were very similar [(S)/(R) = 50, 63 and 32, respectively]. In contrast, these ratios of the quaternary analogue, 4-F-MePyMcN +, differed in the three functional pharmacological assays. They were very low at M 2 and M 3 [(S)/(R) = 2.5 and 4, respectively], but high at M 1 receptors [(S)/(R) = 40]. This implies that the stereochemical requirements of the muscarinic receptor subtypes are different for the enantiomers of 4-F-MePyMcN +, being most stringent at M 1 receptors. The most promising compound (S)-4-F-MePyMcN was further assessed for its affinity to muscarinic receptor subtypes, m l - m4, in binding experiments as well as for its agonistic properties at ml receptors expressed in CHO-K1 cells by measuring the stimulation of the PI turnover. The binding affinities (pK i values: ml = 7.16, m2 = 7.21, m3 = 6.71, and m4 = 7.04) were very similar to the affinities obtained in functional studies (Table 1). The muscarinic receptor-mediated PI turnover in CHO-hml cells of this compound was found to be 40 % of the maximum effect evoked by carbachol, the EC50 value being in the lower nanomolar range. Thus, the high potency and functional M 1 selectivity of (S)-4-F-MePyMcN make this compound suitable for the study of muscarinic receptor mechanisms. Due to its M l-agonistic and M2-antagonistic properties (S)-4-F-MePyMcN may be considered as an important tool for future drug research in the field of cognitive disorders. Its partial agonistic nature may be ad-
56 vantageous in therapeutic applications, as chronic administration of the drug would be less likely to result in receptor desensitization.
M 2 receptor antagonists. Muscarinic M 2 receptors have been reported to be depleted in post mortem brains from patients with Alzheimer's disease [9]. This has raised the interest of developing M2-selective muscarinic receptor antagonists, capable of penetrating into the central nervous system, that could be useful both as diagnostic tools for quantif~ng the loss of muscarinic M 2 receptors with positron emission tomography imaging [20], and as therapeutic agents (see introduction). Most of the substances described in the literature as M2-selective muscarinic antagonists, however, cannot distinguish between M 2 and M 4 receptors. In the search for antagonists which clearly differentiate M 2 from M1, M 3 and M 4 receptors we investigated the two enantiomers of the widely used Hi-antihistaminic drug dimethindene (Figure 2) [35, 36]. It has been found that the enantiomers of dimethindene behaved as simple competitive muscarinic antagonists. Functional (pA 2 values) and binding (pK i values) affinities for the two enantiomers are shown in Table 2 and 3. In general, (S)-dimethindene was more potent than the (R)-enantiomer in all muscarinic assays. However, the stereoselectivity ratios were found to be different at the four muscarinic receptor subtypes, being greatest at M 2 receptors (32- to 41-fold). In contrast, the stereoselectivity was inverse at histamine H I receptors, (R)-dimethindene being the eutomer (Table 2). The pA 2 values for (S)-dimethindene at M 1 receptors in rat duodenum and rabbit vas deferens were not significantly different (P>0.05) either from each other or from pA 2 values determined at M 3 receptors mediating contractions of the guinea-pig ileum and trachea or from the pK i value determined at M 4 receptors in rat striatum. However, the pA 2 (PKi) values for (S)-dimethindene at muscarinic M 2 receptors were significantly (P0.05). Thus, pA2 values were calculated from regression lines whose slopes were constrained to 1.00. b.c,d,e,_f, Agonists used: 4-CI-McN-A-343 b (Figure 1), 4-F-PyMcN+-c (Figure 1), arecaidine propargyl esterd-, carbachol -e, histaminef. In conclusion, the experiments demonstrate that (S)-dimethindene is a novel M2-selective muscarinic antagonist. Since studies in man showed that (S)-dimethindene penetrates readily into the brain [35-37], this compound could be a valuable tool to test the hypothesis that lipophilic M2-selective antagonists show beneficial effects in the treatment of Alzheimer's disease. (S)-Dimethindene might also become the starting point for the development of appropriate PET ligands useful as antemortem diagnostic tools for quantifying the loss of M 2 receptors in the brain of Alzheimer's patients. One interesting aspect of using chiral antagonists, such as dimethindene, in PET studies is the availability of the less potent optical isomer, which can be used to measure nonspecific binding under the imaging condition. Table 3
Apparent affinities (pK i values) for (R)- and (S)-dimethindene at muscarinic binding sites in various tissues and cell lines pKi values Subtype
Tissue/Cell Line
(R)-Dimethindene
(S)-Dimethindene
M1 M2 M3 M4
NB-OK 1 cells Rat heart Rat pancreas Rat striatum
5.6 6.3 5.6 6.0
7.1 7.8 6.7 7.0
Muscarinic binding sites were labelled using [3H]N-methylscopolamine. Hill coefficients of the competition curves were not significantly different from unity (P>0.05).
58
Antagonists with high affinity for M 3 receptors. The muscarinic antagonist (R)-hexahydro-difenidol [(R)-I: HHD] and especially its racemic sila-analogue hexahydro-sila-difenidol (2: HHSiD) are widely used as selective antagonists for the classification of muscarinic receptor subtypes. Both compounds exhibit up to 50-fold higher affinity to M1 and M 3 (as well as to M4) receptors than to M 2 receptors (Figure 3) [38, 39]. In addition, the configurationally stable enantiomers of HHD display high stereoselectivity at M 1 and M 3 receptors (up to 500fold), but ~'ather low stereoselectivity at M 2 receptors (16-fold)[38]. Another compound structurally related to this series is the aminoethyl analogue of (K)HHD, i.e. the (K)-enantiomer of the antiparkinsonian drug trihexyphenidyl (7, THP). (R)-THP proved to be an Ml-selective muscarinic antagonist with intermediate affinity at M 3 and at M 4 and low affinity at M 2 receptors. However, its stereoselectivity profile 0US) of MI=M3=M4 > M 2 is veq' similar to that of HHD (Figure 4) [39]. In contrast to the enantiomers of the carbinols, e.g. HHD or THP, the enantiomers of silanols, e.~. sila-procyclidin, racemize quickly in aqueous solution [40] and are therefore unsuitable for the pharmacological investigation of enantioselectivity. Thus, we wanted to further assess the structural requirements of this kind of compounds for affinity and selectivity on muscarinic receptor subtypes. Accordingly, we replaced the OH group of (R)- and ( S ) - ~ as well as of (R)- and (S)-1vd:P at the central atom by a CH2OH (3, 8), a COOH (5) or CONH 2 (6, 11) group and investigated the influence of this structural change on antimuscarinic potency and stereoselectivity. Furthermore, the exchange of the central carbon atom in the hydroxymethyl analogue o f ~ by a silicon atom (--> 4) as well as of THP by a silicon (-> 9) or a germanium atom (--> 10) led to enantiomers, which were expected to be configurationally stable and therefore valuable for the stereochemical characterization of the interaction of antagonists at muscarinic receptors (Affinity data of compounds 4a, 9 and 9a have already been published [41, 42]). From previous investigations it was postulated, that muscarinic receptors possess four different subsites for antagonist binding, shown e.g. for the carbinols procyclidine and hexahydro-difenidol [43, 44]. Therefore, another aim of this study was to compare the antimuscarinic properties of the enantiomers of the hydroxymethyl-derivatives described above (3, 4, 8, 9) with their related diphenyl- (3a, 4a, $a, 9a) as well as their dicyclohexyl analogues (3b, 4b, $b, 9b) in order to interpret their stereoselective interaction with the different muscarinic receptor subtypes. The following results were obtained: In all cases where the absolute configuration was determined the (R)-enantiomers (eutomers) exhibited higher affinities and stereoselectivities at muscarinic receptor subtypes than the corresponding (S)-enantiomers (distomers) (Figure 3, 4). For the compounds 8, 10 and 11 the configuration is not yet known. However, with respect of the high structural homology and the unique enantioselectivity data [(R) over (S) at muscarinic receptors] found for all compounds in this study, we hypothesize that the absolute configurations of the eutomers of 8, 10 and 11 is (R), too. In the following, eutomers and distomers of these compounds will be given the prefix I and U, respectively. Substitution of the OH group by a CH2OH, a COOH or a CONI-I2 group in the eutomers of HHD and THP resulted in a decrease of affinity, receptor selectivity and stereoselectivity (except stereoselectivity of compound 5) (Figure 5). Nevertheless, all eutomers of compounds 3 - 6 and $ - 12 displayed a selectivity profile of M 1 = M 3 > M 2. In addition, most of these enantiomers exhibited also a stereoselectivity profile of M 1 = M 3 > M 2. Compound 5 with a
59 COOH group - instead of a OH group - at the central carbon atom displayed a reduced affinity at all three receptors, but the enantioselectivity ratios were slightly increased, compared to the parent compound HHD. These results are not in agreement with Pfeiffer's role. Additionally, the eutomer of this carboxylic acid (1-5) displayed higher affinities than its amide 6, and the stereoseleetivity ratios were higher for 5 than for 6. Shortening of the aminopropyl to an aminoethyl chain in the eutomers of the HHD-derivatives (3---)11, 4---)9, 6 ~ 1 1 ) did not change significantly the affinities. In contrast, the stereoselectivity ratios for the enantiomers of these aminopropyVaminoethyl pairs were changed. It was found, that the stereoseleetivity ratios for 3 (aminopropyl series) were higher than those for the related compound in the aminoethyl series (8). Vice versa, the amide of the HHD type (6) exhibited lower eudismic ratios than the amide of the THP type (11).
* /
El
CH2.--CH2.--CH2--N
4
\
5
6
pA2 9.0-
8.0
8.7
r---i (R)-isomer
8.0
7.9
-
E! C Si C Si C C
Compd HHD'I HHSiD: 2
R
8.2
L ~ racemate
II
8.2
R OH OH CH20H CH20H COOH CONH 2
(s)-isomer 7.9
7.7 .,.....,-.
7~ 7,.0
x x
-
7.3
3.7
7.0
7.0
K~
6,,0
5.0
K~
-
K
•
~
x
51.11
-
,cx
~
9 x
4.0
C~
-
M1 M2 M3 1 !
121131
I M1 M2 M3 M1 M2 M3 M1 M2 M3 ! 4 II S !1 6 1
Figure 3. Structures and affinity profiles (pA2 values) of (R)- and (S)-enantiomers of compounds 1 - 6 at muscarinic M 1 (rabbit vas deferens), M 2 (guinea-pig atria) and M 3 receptors (guinea-pig ileum); *center of chirality.
60 Compd R2
./
PA2 10.0 -
,,
R1 9.6
El
THP: 7 8
/
\
CH2_CH2.__ N \ "
-
8~
-
/
6.0
-
5.0
-
Com -I
10 11 Fenpipramide: 12
R2 OH CH20H CH20H CH20H CONH 2 CONH2
~
distomer -b
8.2
IK~
73 9
7.1
7.3 --
7.1
7.2
ii
M1 M2 M3 M1 M2 M3 M1 M2 M3 M1 M2 M3 M1 M2 M3
7
1 l 8
9.1
8.: ~>
7.4 -
9
racemate
I eutomer -a
8.2
7.0
\
R1 c-Hex c-Hex c-Hex c-Hex c-Hex Ph
9.4
I 9.0
,
El C C Si Ge C C
I !
'
I I
10
I
I
11
!
K~x
Ig>
KY
Ix> lg>
g:~ K)
K)
KX
!~
Kx
IX'>
~-)
4) and THP (8->9) lowered atfmity and stereoselectivity, whereas the receptor selectivity for compounds 3 and 4 was unchanged. In contrast, the corresponding (R)- and (S)-enantiomers of the Si/Ge analogues exhibited very similar affinities to the different muscarinic receptors indicating a strongly pronounced Si/Ge bioisosterism. Replacement of the cyclohexyl by a phenyl ring (-~ diphenyl analogues: 3a, 4a, 8a, 9a) or of the phenyl by a cyclohexyl ring (--> dicyclohexyl analogues: 3b, 4b, 8b, 9b) of the hydroxymethyl C/Si analogues of HIK) (3, 4) and THP (8, 9) diminished affinity to all muscarinic receptors studied compared to the corresponding eutomers (Figures 5 and 6).
61
R1 x
R2"
C
/ \
CH=--CH2--CH2"-'N /
pA2 8.5-
R1
Compd
CH2-OH
\
8.2
'~)
/
V-q
R2
Ph c - H e x (R)-3 Ph Ph 3a 3b c - H e x c-Hex Ph 1s1-3 c - H e x 8.2
8.07.5-
7.1
7.06.56.0-
ApA2
5.5 1.1 1.4 2.1 2.5
AI" [(R)-31 - [3al A 2 : [ ( R ) - 3 1 - [3b] E.I.: [[R)-3] - [(S)-3] A I + A 2 = expected E.I.:
RI
X
CH2-OH
/
,, Si \
R2
CH2--CH2--CH2--N
pA 2 7.5
I~
/
Compd
RI
R2
(R)-4 4a 4b
Ph
c-Hex
Ph Ph c-Hex c-Hex
I---! (s)-4 c-Hex
\
7.2
0.9 1.3 2-2 2-2
0.4 0.9 1.1 1.3
Ph
7.3
7.0 6.5
6.2
6.05.5-
ApA 2
5.0 -
A 1" [(R)-4] - [4a] A2: [(R)-4] - [4b] EJ.: [(R)-4] - [(S)-41 A I + A 2 = expected E.I.:
0.9 0.8 1.0
0.2 0.6 0.8
0.5 0.8 1.0
1.7
0.8
1.3
Figure 5. pA 2 and ApA2 values of (R)- and (S)-isomers of compounds 3 and 4 as well as of related achiral compounds 3a, 3b, 4a and 4b at muscarinic M 1 (rabbit vas deferens), M 2 (guinea-pig atria) and M 3 receptors (guinea-pig ileum); Ph = Phenyl; c-Hex = Cylohexyl; E.I. = Eudismic index.
62 R1
\
CH2-OH
/
II
/ CX
R2
fl)-8
8a 8b I----1 (11)-8
CH 2-- CH2 --N/~
pA 2 8 . 5 -
Compd
8.2
R1
R2
Ph
c-Hex
Ph
Ph
c-Hex c-Hex c-Hex Ph 8.2
8.0--
7.5
7.2 7.2
7.06.56.0-
APA2 A 1" [(!)-8] -
5.5 [8a]
R1 \
R2 pA2/pKi
1.5 1.6
/
/
0.0 0.7 0.7 0.7
0.6 1.0
A2: [(I)-8] - [8b] E.I.: [(I)-8] - [(11)-8] AI+A2 = expected E.I.:
SiX
2.0
CH2-OH I~
CH2--CH2 - N
7.4
7.5
0.9 1.1 1.6
7.3
7.0
Compd
R1
R2
(R)-9
Ph
c-Hex
9a
Ph
Ph
9b c-Hex c-Hex r---I (s}-9 c-Hex Ph 7.2
6.8 ~ 7
6.5
'
6.0 5~
APA2/PKi
5.0
A 1" [(R)-9] -
[9a]
-
A2: [[R)-9] - [9b] E.I.: [(R)-9] - [ ( S ) - 9 ] • 1 + A 2 = expected E.I.:
0.4 0.6 0.9
0.1 0.6 0.5
0.5 0.5 1.1
0.4 0.4 0.8
1.0
0.7
1.0
0.8
Figure 6. pA 2 (PKi) and ApA2 (ApKi) values of the enantiomers of compounds 8 and 9 as well as of related achiral compounds 8a, 8b, 9a and 9b at muscarinic M 1 (rabbit vas deferens), M 2 (guinea-pig atria) and M 3 receptors (guinea-pig ileum); Ph = Phenyl; c-Hex = Cylohexyl; E.I. = Eudismic index.
63 However, these achiral analogues displayed still higher affinities to the muscarinic receptors than the corresponding distomers. In contrast to these results, the diphenyl derivative with a CONH 2 moiety (fenpipramide, 12) showed up to 220-fold higher atY~ties than the related eutomer I - l l at the muscarinic receptors. According to the concept of the four-binding-site model [43, 44], it is suggested that muscarinic antagonists of the given structure (except 11) might be recognized by muscarinic receptors with subsites for the ammonium group, the hydroxyl group and the phenyl and cyclohexyl moiety (Figure 7). As can be seen in Figure 5 and 6, the experimentally obtained eudismic indices for 3, 4, 8 and 9 [pA2(eutomer ) - pA2(distomer)] were in most cases very similar to the expected eudismic indices {[PA2(eutomer) - PA2(diphenyl analogue)] + [PA2(eutomer) - PA2(dicyclohexyl analogue)]} at all muscarinic receptors. Only the expected and the observed eudismic index for compound 4 at M 1 receptors showed a remarkable difference. These results suggest that in most cases the stereoselective interaction of the two enantiomers of these antagonists with muscarinic receptors is based on opposite binding of the phenyl and cyclohexyl ring to site 1 and site 2 as well as on equal binding of the hydroxyl and the ammonium group to site 3 and site 4, respectively (Figure 7). Thus, the stereoselectivity ratios of these compounds are best explained by weaker binding of the phenyl and the cyclohexyl ring of the distomers.
Site I
Site 3
Site I
Site 3
8+...X
.~1~~ CH=-OH
H
CH2-CH=-CH::~Nx - - +,_._., ~ X"
Site 2
_ ~'I"'"ElXcH=-CH=-cH=H~,N+~
LR
f
~lUet4
"~
Figure 7. Interaction of the protonated (R)- and (S)-isomers of compounds 3 (El = C) and 4 (El = Si) with four subsites of muscarinic receptors. Site 1: phenyl-preferring hydrophobic subsite; Site 2: cyclohexyl-preferring hydrophobic subsite; Site 3: polar subsite for the hydroxyl group; Site 4: ionic subsite for the protonated amino group; *center of chiraliV. In contrast to these results, the enantiomers of the amide 11 exhibited an aJLtagonistic binding behaviour which could not be interpreted by the concept of the four-binding-site model. It is therefore suggested that the amides 11 and 12 may interact with other ~;ubsites of muscarinic receptors than the related chiral compounds 3, 4, 8 and 9. In conclusion, the substitution of the hydroxyl moiety in HHD and HHSiD as well as in THP by a CH2OH or a CONH2 group decreased affinity, subtype-selectivity and stereoselectivity. The enantiomers of the carboxylic acid derivative 5 showed the highest sterecselectivity of the compounds tested in this study. However, all these derivatives are able to differentiate M1 and M 3 receptors from M 2 receptors. In addition, most of the stereoselectivirf ratios of
64 these compounds follow the same profile of MI=M3>M2, and may therefore used as an additional parameter to characterize muscafinic receptor subtypes. Their enantioselectivities are best explained by the four-binding-subsite model suggesting that the differences in affinity of (R)- and (S)-enantiomers at muscafinic receptors are due to opposite binding of the phenyl and the cyclohexyl ring to the preferring subsites. ACKNOWLEDGEMENTS The authors thank the Fonds der Chemischen Industrie (Germany), the Deutsche Forschungsgemeinschafl, the Boehringer Ingelheim KG, Ingelheim, and the Fonds for Medical Scientific Research (Belgium) for financial support and gratefully acknowledge the donation of drugs [(R)- and (S)-dimethindene: Zyma GmbH, Munich; (R)- and (S)-trihexyphenidyl: Dr. A. Aasen, Oslo]. REFERENCES
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21
Hulme EC, Birdsall NJM, Bucldey NJ. Annu Rev Pharmacol Toxicol 1990; 30: 633-673. Levine RR, Birdsall NJM. Life Sci 1993; 52: 405-597. Levine RR, BirdsaU NJM. Life Sci 1993; 56: 801-1053. CaulfieldMP. Pharmac Ther 1993; 58: 319-379. Feifel R, Wagner-R6der M, Strohmann C, Tacke R, et al. G Br J Pharmacol 1990; 99: 455-460. Waelbroeck M, Camus J, Tastenoy M, Mutschler E, et al. Eur J Pharmacol - Mol Pharmacol Sect 1992; 227: 33-42. D6rje F, Wess J, Lambrecht G, Tacke R, et al. J Pharmacol Exp Ther 1991; 256: 727733. Baker R, MacLeod AM. In: Kozikowski AP, eds. Drug Design for Neuroscience, New York: Raven, 1993; 61-85. Quirion R, Aubert I, Lapchak PA, Schaum RP, et al. Trends Pharmacol Sci 1989; 10 (Suppl.): 80-84. Bartus RT, Dean RL, Beer B, Lippa AS. Science 1982; 217: 408-417. Nitsch KM, Wurtmann RJ, G-rowdon JH. Arzneim -Forsch/Drug Res 1995; 3a (Suppl): 435-438. Gandy S, Greengard P. Trends Pharrnacol Sci 1992; 13" 108-112. CordeUB. Annu Rev Pharmacol Toxicol 1994; 34: 69-89. Aubert I, Araujo DM, Cecyre D, Robitaille Y, et al. J Neurochem 1992; 58: 529-541. Gitler MS, Reba RC, Cohen VI, Rzeszotarski WJ, et al. Brain Res 1992; 582: 253-260. Doods H, Entzeroth M, Ziegler I-I, Schiavi G, et al. Eur J Pharrnacol 1993; 242: 23-30. Schulte B, Volz-Zang C, Mutschler E, Home C, et al. Clin Pharrnacol Ther 1991; 50: 372-378. Goyal RK. New Engl J Med 1989; 321: 1022-1029. Mutschler E, Feifel R, Moser U, Tacke R, et al. Eur J Pharmacol 1990; 183" 117-119. Dewey SL, Volkow ND, Logan J, MacGregor KR, et al. J Neurosci Res 1990; 27: 569575. Barnes PJ. Life Sci 1993; 52 521-527.
65 22 Barnes PJ, Belvisi MG, Mak JCW, Haddad E-B, O'Connor B. Life Sci 1995; 56: 853859. 23 Bungardt E, Mutschler E. Spasmolytics. In: Ullmann's Encyclopedia of Industrial Chemistry, 1993; Vol. A 24. VCH Publishers, Inc., Weinheim, New York, pp 515-528. 24 Minette PA, Barnes PJ. Am Rev Respir Dis 1990; 141: S 162-S 165. 25 Eltze M, Gmelin G, Wess J, Strohmann C, et al. Eur J Pharmacol 1988; 158: 233-242. 26 PfaffO, Gross J, Waelbroeck M, Mutschler E, Lambrecht G. Life Sci 1995; 56: 1038. 27 Eltze M. Eur J Pharmacol 1988; 151: 205-221. 28 Emmerson J, Mackay D. J Pharm Pharmacol 1979; 31: 798. 29 Waelbroeck M, Tastenoy M, Camus J, Christophe J. Mol Pharmacol 1990; 38: 267-273. 30 Ensinger HA, Doods HN, Immel-Sehr H ~ Kuhn FJ, et al. Life Sci 1993; 52: 473-480. 31 Doods HN. Drugs ofthe Future 1995; 20: 157-164. 32 Lambrecht G, Moser U, ~ U, PfaffO, et al. Life Sci 1993; 52: 481-488. 33 Micheletti R, Schiavone A. J Pharmacol Exp Ther 1990; 253: 310-314. 34 Waud DR. J Pharmacol Exp Ther 1969; 170:117-122. 35 Cicurel L, P6chadre J-C, Grandjean E, Duch~ne-Marullaz P. Agents Actions 1992; SPC: C440-C443. 36 Casy AF, Drake AF, Ganellin CR, Mercer AD, Upton C. Chirality 1992; 4: 356-366. 37 Nicholson AN, Pascoe PA, Turner C, Ganellin CR, et al. Br J Pharmacol 1991; 104: 270276. 38 Lambrecht G, Feifel R, Moser U, Wagner-ROder M, et al. Trends Pharmacol Sci 1989; 10 (Suppl.):60-64. 39 Waelbroeck M, Camus J, Tastenoy M, Mutschler E, et al. Eur J Pharmacol Mol Pharmacol Sect 1992; 227: 33-42. 40 Tacke R, Linoh H, Ernst L, Moser U, et al. Chem Ber 1987; 120: 1229-1237. 41 Tacke R, Reichel D, Kropfgans M, Jones PG, et al. J Organomet Chem 1995; 14: 251262. 42 Tacke R, Kropfgans M, Tafel A, Wiesenberger F, et al. Z Naturforsch 1994; 49B: 898910. 43 Waelbroeck M, Camus J, Tastenoy M, Lambrecht G, et al. Eur J Pharmacol Mol Pharmacol Sect 1990; 189:135-142. 44 Waelbroeck M, Camus J, Tastenoy M, Mutschler E, et al. Chirality 1991; 3:118-123.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardinh, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
67
Perspectives in the design and application of dopamine receptor agonists Cesare Casagrande, Giorgio Bertolini Department of Organic and Industrial Chemistry, University of Milan Italfarmaco Research Centre, Milan Introduction
The era of dopamine receptor pharmacology began around 1960, heralded by the discovery of the neurotransmitter role of dopamine, previously regarded only as a metabolic precursor of noradrenaline. In the early years fundamental discoveries were made, such as the dopamine defect as a cause of Parkinson disease and the substitutive therapeutic value of levodopa, the involvement of dopamine receptors in the mode of action of existing neuroleptics, e.g., chlorpromazine, leading to the hypothesis that schizofrenia may originate from a disfunction of neuronal dopaminergic pathways, and the unique vasodilator and natriuretic properties of dopamine with respect to the other catecholamines. These discoveries were made possible by the use of the few available drugs 9in addition to dopamine itself, applicable "in vivo" only by intravenous infusion and not entering the CNS upon peripheral administration, and to levodopa, actively transported into the CNS and converted into dopamine, apomorphine (!) and some ergot derivatives, such as bromocriptine 2(2) and codergocrine were soon recognized to exert some of the actions of dopamine and became useful tools for pharmacological investigations. These drugs were also the leads of medicinal chemistry, either indicating the way to new prodrugs of dopamine as carriers to the cardiovascular-renal system or to the CNS, or being suggestive of new active molecules by the recognition of a dopamine-like moiety buried in the rigid structures of aporphines and ergolines. Indeed, all dopaminergic pharmacology has been imprinted by these early findings: as a matter of fact, Parkinson disease, psychotic disorders, and cardiovascular disease, such as congestive heart failure and hypertension, remain the main therapeutic targets, and medicinal chemists have elaborated most of their efforts to develop new agonists, and some of the antagonists, on structural relationships with dopamine. Only one drug of this "first generation", piribedil (3) [1], developed for its vasodilator properties and somewhat later shown to act by stimulation of dopamine receptors, indicated the possibility to work out new leads for agonists not related to dopamine. Investigation of dopamine receptors in the central nervous system led to the identification of two subtypes, D 1 and D 2 showing distinctive ligand binding profiles [2]; at the same time, two similar subtypes DA 1 and DA 2 were characterized in peripheral tissues, mostly by "in vivo" pharmacological responses, and were kept on a separate denomination, by suggestion of L.I. Goldberg [3], until an unequivocal demonstration of correspondence could be achieved.*
* Investigation of D 1 vs DA1, and D 2 vs DA 2 receptors later reconciled their belonging to the same subtype, and the denomination D l_like and D2.1ike became comprehensive of these receptors and of the new subtypes (])3, D4, D5). For the sake of simplicity, in the following we shall use the D1-D 2 denomination for discussion of literature prior to the discovery of the latter subtypes, and the D 1 to D 5 denomination in the section "Investigation of new subtypes" and in the following ones when referring to recent literature making relevant distinction of the subtypes.
68 O HN H I
1
CH3 O CH(CI'I~)2 F-----I
H
"
%o
"NH'I ~I u
"CH3
!
I0, X : OH
3
'~ #c,,
H
Ho.
CH2CH(CH3)2" ~ /
;, U
A
~
I~
"~
4, R=H 5, R =nPr
NR2 ~
N
HO- y OH
, ~ Z
~,x=oso~cF, I- I1
8, X=H 9, X= OH
HO
2 Br
r
N ~
12, X = SO2CH3
R
2
H
v 6, R=H 7, R=nPr
~ d,~H Nit
NO X
13, X = Y = Z = R = H 14, X= CIY = OHZ= R = H 15, X= CIY= HZ= R = CH3
These findings opened the way in the late 1970 to the design of a second generation of agonists and to the definition of structure-activity relationships including selectivity for the receptor subtypes. The progress registered in the following decade has been extensively reviewed [ 1, 3-6]; we shall point out here the main pharmacological and clinical achievements, as well as the essential results of the search of pharmacophores and of tentative receptor models, in order to provide the basis for discussing the ongoing efforts towards a "third generation" of dopamine receptor agonists.
The "second generation" of dopamine receptor agonists Simple modifications of the dopamine structure showed that N-substituents, apart from N-methyl, abolished D 1 activity. D 2 activity also decreased by mono-substitution but was preserved or increased in disubstituted compounds, when one of the substituents was a small alkyl, optimally n-propyl; activity markedly decreased with n-butyl, or with branched groups. A modest D 1 activity (twenty times less than dopamine) reappeared in N,N-di-n-propyl and Nn-propyl-N-n-butyldopamine. At the same time, it was shown that D 2 activity was preserved in compounds having only the m-hydroxy group in the aromatic ring, while D 1 activity required the unchanged catechol system. Several groups become engaged in the synthesis of rigid dopamine analogues, focussing on the peculiar pharmacological properties of 6,7-ADTN and 5,6-ADTN (racemic _4 and 6_., respectively), whereby it was confirmed that one hydroxy group was required for D 2 activity, respectively in 7 or 5 position, in both cases coincident with the m-position of dopamine structure, while two hydroxy groups were required for D 1 activity. In the two isomeric series, the dopamine side chain was fixed in two different rotameric conformations, respectively denominated 13 and oL While the D 2 activity increased in both isomeric series from the primary amines to the N,N-di-n-propyl derivatives (racemic _5 and 7_), and D 1 activity was significantly expressed only in the N-unsubstituted 6,7-ADTN, it was surprisingly observed that in the 5,6-ADTN series D 1 activity was elicited only by the N,N-din-propyl derivative 7(].) and not by the primary amine. These observations were in good part made rational by an investigation of McDermed et al. [7], revealing that in the 6,7- and 5,6ADTN series, as well as in their monophenolic analogues, the pharmacological activity was respectively expressed by the (R)- and (S)-enantiomers (respectively 4_, _5 and _6, 7_); they
69
C HO
\ H~
~-----~~ "~ /
\
i~/"=-FN OH3
Fig. 1. Receptor models by McDermed [7] ( A red, 4-5_.; black, 6-7; B " red, (+)isoapomorphine; black, 1_) and Kaiser [4,5] (C, 13 with phenyl group in pseudoequatorial or pseudoaxial (dotted lines) conformation). keenly proposed the first model of the dopamine receptor (Fig. 1A), accomodating in an almost superimposable way the active enantiomers, thus accounting for the activity of both ctand 13-rotameric forms of dopamine. Furthemore, the model provided an explanation of the activity of apomorphine and the inactivity of (RS)-isoapomorphine (9,10-dihydroxyaporphine) (Fig. 1B). The latter was related to a steric impairement in the bottom of the receptor cleft, rather than to its 13-rotameric conformation, as previously suggested. The model could not account for the peculiarity of the activity of N,N-di-n-propyl-5,6-ADTN with respect to the D 1 receptor. Investigation of the role of the catechol system, using in part previous experience on agonists of 13-adrenergic receptors, pointed out, among various cyclic and non cyclic analogues carrying NH acidic groups in lieu of the m-hydroxy group of dopamine, the indolone SK&F 89124 ~ as the most potent and selective D 2 agonist [8]. The analogue devoid of the paralike hydroxy group (ropirinole, 8_), although markedly less potent, showed a good bioavailability and pharmacological activity in vivo. The latter may be due to its metabolical conversion into 9. Ropirinole has been initially investigated as an antihypertensive agent, and later on in Parkinson disease. Clinical studies are currently in progress; ropirinole has recently been shown to be also active on D 3 receptors. Swedish academic and industrial investigators [9] decided to exploit the new information on the role of presynaptically located neurotransmitter autoreceptors, including those of dopamine, in controlling, by a feed-back mechanism, the release of the neurotransmitter in the synapse. They suggested that a selective agonist of the presynaptic dopamine autoreceptor could provide a pharmacological alternative to the blockade of postsynaptic receptors in the treatment of psychotic conditions. Their prototype D 2 presynaptic agonist, preclamol 0 ~ , was preferred to the corresponding racemate, which had generated some confounding results, and it was promoted to clinical investigations, which have not yet reached a full conclusion, but have stimulated new research in this direction. The discovery of the selective vasodilation produced by dopamine in the renal circulation and of its natriuretic effects, both mediated by D 1 receptors, respectively in renal arteries and in renal tubules, prompted research of new D 1 selective and peripherally acting agonists, potentially useful in the treatment of congestive heart failure and of hypertension. In the most severe forms of the former clinical condition, dopamine is used by intravenous infusion, taking
70 advantage of dopamine receptor mediated effects at low doses, as well as of cardiac stimulation via 13-adrenergic receptor stimulation at high doses. The interest in the exploitation of the renal effect of dopamine in hypertension is linked to the complex control exerted by the kidney on the circulation [ 10, 11 ]. Derivatives of 7,8-dihydroxy-3-benzazepine, which enclose a constrained, but not rigid, dopamine-like structure, show only modest dopamine-like activity. Addition of a phenyl ring in the 1-position afforded SK&F 38393 (racemic 1_3), showing a remarkably selective activity at the D 1 receptor. A large number of" analogues was investigated [4, 12], pointing out that appropriate structural modifications can increase the intrinsic activity, producing full agonists, while (+)-13 shows a partial agonist profile in some assays, or can decrease selectivity and specificity by inducing either D 2 agonistic activity or affinity for adrenergic or serotoninergic receptors. Modification of the benzazepines has also produced a class of D 1 selective antagonists, the prototype of which is SCH23390. Among the derivatives, fenoldopam, the racemic form of the 6-chloro-4'-hydroxy analogue ( ! ~ , showing predominant distribution in peripheral tissues, was selected for in-depth investigation and became a unique tool for studying the cardiovascular implication of stimulation of D 1 receptor in the kidney and the circulation. Clinical investigations were promoted in congestive heart failure and hypertension [ 13 ]. Development was restricted to short-term intravenous administration in the treatment of hypertensive crises. The oral use in chronic treatment was discouraged in part by the low bioavailability and short duration of action, and in part by the observation that the efficacy rapidly decreased due to reflex sympathetic activation, consequent to the potent vasodilator effects of the drug, and also due to increase of plasma renin activity. The latter has been linked to an increase in renin secretion dependent upon the stimulation of specific D 1 receptors located in the juxtaglomerular cells in the kidney. The interest in the use of dopamine agonist in cardiovascular therapy was also the basis of investigation of dopexamine ~ at Fisons laboratories [14]. Starting from the biscatecholamine structure of hexoprenaline, a tocolytic 132-agonist, a pair of dopamine agonistic structures were joined by variable length links between the amino groups. It was shown, as could be expected, that only one moiety carrying the essential features for dopamine receptor stimulation was sufficient for the activity, but it was also shown that a six-atom link was optimal, and that the basicity of the second amino group and the second aromatic ring were essential for D 1 receptor stimulation. The selected compound, dopexamine, expressed a marked activity at D 1 receptors, unusual in N-mono-substituted dopamines, suggesting that useful interactions involving counterparts of the second amino group and aromatic ring in a region of the receptor far away from the points of interaction of dopamine may be involved. A lesser (1/10) activity at D2 receptors and a 132-adrenergic agonistic action were shown to be present. The latter was considered a useful complement of the pharmacological profile, providing additional vasodilation and possibly a positive inotropic effect. Dopexamine has been clinically compared with i.v. dopamine, showing a dose-dependent vasodilator effect, at difference from that of dopamine which is limited, at very high doses, by the counteraction of the ot-adrenergic vasoconstrictor activity. The intravenous infusion of dopexamine is in clinical use in some countries in conditions of low cardiac output [ 13 ]. As in the case of fenoldopam, the effects of a prolonged administration, such as an infusion prolonged up to 24-48 hours, tended to wear off, because of reflex sympathetic activation, which was amplified in this case by a specific ability of the drug to block the uptake of norepinephrine, thus sustaining the high circulating level produced by the synaptic spillover. Elaborations of the ergoline structural lead were prevailingly addressed to the development of new antiparkinsonian agents, since bromocriptine, acting as a D 2 agonist, had
71
xN~HN
H N!
~
R 16, % - nPr, Rsb - CH2SCH 3
17, R6 = allyl, R8b = N-- CONHEt |
(CH2)3N(CH3)2
18
~
X~ ~ ~ ~ , v ~ , s 19
N
20, X - H
21, X OH 22, X = COOH
shown its clinical utility, either in delaying the initiation of levodopa therapy, or in complementing the efficacy of moderate doses of levodopa. Levodopa, although markedly effective, had shown undesirable side effect in a number of patients, and frequently a decrease of efficacy after some years of therapy, along with the induction of "on-off' fluctuations of effects and of motory disturbances. Bromocriptine interacts with adrenergic and serotoninergic receptors. The pharmacological profile of pergolide ~ [15] showed increased, although not complete, specificity for dopamine receptors, and also involved both D 1 and D 2 agonistic action. Cabergoline (17.) [16] showed increased specificity for the dopamine receptors, marked D 2 agonistic activity and long duration of action. Owing to its scarce entrance in the CNS, its clinical application has been preferentially addressed to the reduction of hyperprolactinemia, an effect elicited via the D 2 receptors located in the anterior pituitary, which is not protected by the blood-brain barrier. High prolactin secretion has clinical consequences, such as galactorrhea, amenorrhea and increased risk of breast cancer relapse. In the ergoline structure, two blocked heteroaryl-ethylamine moieties can be assumed to have relevance in dopamine receptor interaction : a 4-indoleethylamine, in which the NHindole group may surrogate the m-hydroxy group of dopamine in donating ahydrogen bond, or a 3-pyrroleethylamine also containing the NH-group suitable for a similar interaction. This ambiguity has stimulated different approaches of molecular dissection, such as the one represented by N,N-dipropylindoleethylamine ( ~ ) [1], which was shown to be significantly active (1/10 of pergolide) at D 2 receptors, and the other one by quinpirole (racemic 1_.9), a pyrazoloquinoline endowed with marked D 2 activity, in common with the pyrrolo-quinoline analogue directly derived from ergoline structure modification. Quinpirole, originally investigated as an antihypertensive agent, showed some paradoxical hypertensive effects in normal volunteers; its pharmacological profile is being reconsidered in view of its D 3 agonistic activity [9]. Active D 2 agonists, having an indolebutylamine structure not strictly related with the ergolines, were discovered when compound EMI) 23348 2(!0.) was submitted, in a frame of pharmacological screening, to the Ungerstedt test of controlateral rotation in rats with unilateral nigrostriatal lesions. Roxindole ~ and carmoxirole ~ were generated from this new lead and respectively investigated as potential antiparkinson and antihypertensive agents, in the latter case because of the exclusively peripheral distribution of carmoxirole [ 17]. Intensive discussion of structure-activity relationships and features of pharmacophores and receptor models accompanied the development of the "second generation" of dopamine agonists. There was agreement in pointing out, as the key features of the pharmacophores, the distance between the m-hydroxy group and the amine nitrogen, corresponding to a fully extended conformation of the side chain of dopamine, and the directionality of the lone pair orbital of the nitrogen. Among various hypotheses, the model of Kaiser [4,5] shown in Fig. 1C implemented the basic concepts of McDermed [7] and included all possible, and yet not fully defined, interactions of the D 1 and D 2 agonists. There were considered : 1) an anionic site
72 (COO-), binding the amino nitrogen; 2) the M-site, binding the m-hydroxy group; 3) the Psite, binding the p-hydroxy group; 4) an N-site, located at about 2.0 A distance from the Psite, interacting with the NH group of the pyrrole ring in the ergoline structure; 5) a I'I 1 site providing a rI-rI interaction with the aromatic ring of the catecholamine/ergoline structure; 6) a I-I2 site, at the bottom of the receptor pocket, allowing a 1-I-rI interaction of the pseudoaxial conformation of (R)-I3 (dotted-line phenyl), or the alternative site 1-I2.. The latter appeared as most likely in view of the pseudoequatorial conformation of (R)-fenoldopam ~ derived from X-ray diffraction [12]; 7) a I"I3 site, corresponding to the second aromatic ring of apomorphine; 8) a C3-site, as a lipophilic cavity suitable to fit a N-substituent not larger than n-propyl; 9) a large lipophilic site (LL) suitable to enclose a larger N-substituem.
Dopamine prodrugs Levodopa holds the main role in the therapy of Parkinson disease, complemented by bromocriptine and the new ergolines. The use as a prodrug of this endogenous precursor of dopamine showed, in addition to side effects, drawbacks dependent upon irregular absorption and peripheral decarboxylation. The latter problem was effectively overcome by the coadministration of inhibitors of levodopa decarboxylase having distribution limited to peripheral tissues, such as benserazide and carbidopa, allowing an increased and more regular access of the prodrug to the CNS. So far attempts to design prodrugs of dopamine or levodopa, which could effectively improve the delivery of the active agent in the central nervous system, were not successful. Analyses by various kinetic models of the factor governing the selective delivery of a drug in a target compartment by a suitably designed prodrug [18], applicable to the CNS and the kidney, brought into light the main determinants of site-specificity: a small volume of distribution of the prodrug in the central and other non-target compartments, an active uptake of the prodrug into the target compartment, a fast conversion of the prodrug into the drug in the target compartment, and the retention there of the drug. Retention or binding of the prodrug in the target compartment may also contribute, as far as the conversion to the drug is not hindered. The fast diffusion of the prodrug to the target compartment gives only modest contribution, since it produces similarly fast distribution towards non-target compartment and tends to decrease selectivity. The key advantage of levodopa as a prodrug is represented by its transport via the physiological mechanism of aromatic aminoacids uptake into the brain, particularly when peripheral metabolization/activation is drastically reduced. Up to now no practical progress with new prodrugs has been achieved; use of soluble prodrugs of levodopa has been suggested as a means of continuous administration, approaching a zero-order rate of absorption, in order to limit the incidence of kinetical factors on-off phenomena in severe patients. A device for retention of a dopamine prodrug into the central nervous system has been designed by the synthesis on N-methyl-dihydronicotinoyl amides of dopamine 3,4-0-diesters, such as 23, which are converted into a quaternary pyridinium derivative by oxidation, and thereby retained inside the blood-brain barrier [19]. Only modest dopamine-like activity was observed, and this may due to a slow rate of enzymatic hydrolysis of the amide bond joining the dopamine and the pyridinium moieties. A new intriguing approach to a prodrug retained by quatemarization in the CNS has been reported recently [20]. Exploiting a mechanism previously used in the vitamin prodrug thiamine disulphide, lipophilic compound 2_4,4has been synthetized by coupling a 3,4-0-diacyl ester of levodopa with the open-ring precursor of a thiazolium compound. In this case, the quaternary derivative is generated by a reductive cleavage of the disulphide, involving
73
~
H~J~
U9
--N--
AH~
23
N CH3
I I
0
I.,
~O~
L~"
O" ~(.~ I~
CH3 OH3.N~ ~ . / O , .,~ I
~
/I
NH~ ,~'1"1 ~" L~x I ~
Oy~y~'~,~. NHCH3HO~ ~ ~ ,,,~/
24
~
,
~~L. ~
"~
O
-o
O O / 1 ~Y
0
@H3
,
,~
"1.
o-
T
27
HO~
p -0 I' o
NHCH3 26
~ -NH ,
~
H
--
T/
I~ ..~
HO~ ' ~
IT v
" ~og.
"C,OOH
2s
H NHCOCH3
O rO y'vNHC' 'SOH O O "~ /
O"~'O
28
O
glutathione; thereafter, the release of levodopa appears to be fast, and produces by i.v. administration in rats brain level AUC's 30 times larger than an equimolecular dose of levodopa. At difference from the pyridinium amides, an easily hydrolyzed ester bond joins levodopa with the quaternary moiety. It has however to be investigated whether this prodrug can overcome first-pass modification by the glutathione pool in the liver, and which consequences can derive from the burden of the thiazolium compound in the brain. Specific delivery of dopamine to the kidney represents an interesting pharmacological target in hypertension and in congestive heart failure. Investigations in this direction have been inspired by the presence of dopamine receptors in tubular cells, in which both active transport of organic anions and high local concentrations of enzymes amenable to prodrug activation are available. Gludopa (2~ has been assumed to be taken up, hydrolyzed to levodopa by Tglutamyltranspeptidase, and then decarboxylated "in situ" to dopamine. A group of phosphoric esters of dopamine, including as the most active compound fosopamine (N-methyldopamine 40-phosphate) (~6.), were investigated assuming uptake and dephosphorylation by alkaline phosphatase in the brush border membrane of the tubular cell [ 18]. In laboratory investigation 26 showed a potent, dose-related renal vasodilator effect in dogs on oral administration even in low doses of 0.6 mg/kg, but no dopamine-like effect was observed in preliminary studies in humans. It has to be concluded that the prodrug, which appeared to reach the kidney for local activation in significant amounts in laboratory animals, was most probably completely dephosphorylated in the gut wall in humans. Gludopa, although not absorbed orally, was investigated in a number of studies in healthy volunteers and in selected hypertensive patients by i.v. infusion, indicating that it can correct the fault of sodium excretion, and improve renal function without modifying renin plasma levels [21 ]. Its effects appeared to be dependent both on D 1 and D 2 receptor stimulation. The dose required was high (10-50 times the dose of i.v. dopamine). It has recently been shown in animal experiments [22] that the effects of 25 are not selective for the renal district; this may be due to extensive metabolization in other tissues, particularly in the intestinal wall. Thus, both attempts failed because of scarce organ selectivity in the metabolic activation step, and the quest of a renal selective dopaminergic drug, or prodrug, remains unsolved. At difference from these results, the design of prodrugs suitable to produce by oral administration all the cardiovascular and renal effects of dopamine met success with ibopamine 2(~7.) and with docarpamine 2(2~. Ibopamine, a diester of N-methyldopamine, appeared to be orally absorbed and rapidly activated in peripheral tissues, but did not induce central dopaminergic effect in laboratory animals, at difference from levodopa. It was compared with
74 dopamine i.v. infusion in short term studies, showing similar hemodynamic and renal effects, and it was thereafter investigated in the chronic treatment of congestive heart failure [ 13]. In long term studies, its efficacy appeared to be dependent not only upon D 1 receptor related effects, but also on effects mediated by the stimulation of peripheral D 2 receptors, in particular decreased noradrenaline release via presynaptic receptors, reduction of overstimulated aldosterone release via D 2 receptors located in the adrenal zona glomerulosa, and lack of increase of renin plasma level, since D 2 presynaptic action counteracts the release induced via D 1 receptors. This pattern of neurohumoral effects has clinical relevance in congestive heart failure, particularly for sustained therapeutical efficacy. Docarpamine was also shown to be an orally effective peripheral dopamine prodrug, at doses higher than ibopamine [13, 23], and it is used in Japan for weaning patients from i.v. dopamine infusion in severe heart failure.
New dopamine agonists in cardiovascular therapy The therapeutic properties of ibopamine appear to be dependent upon its combined effects on D 1 and D 2 receptors, avoiding unwanted neurohumoral activation and development of tolerance, at difference from selective D 1 agonists. This suggests that an advancement in cardiovascular and renal applications of peripherally acting dopamine agonists may be represented by balanced D1-D 2 activity in a drug devoid of the ot-adrenergic and 13-adrenergic effects that dopamine and N-methyldopamine can elicit in high doses. In our approach to such a goal, we were attracted by the potent dopaminergic activity of the isomeric ADTN and by the possibility of modulating it, as well as the adrenergic activity present in these compounds [4], by structural modifications of the nitrogen substituents. In particular, we considered the use of ortho-substituted-aryloxyethyl groups [24, 25], which could revert the a-adrenergic action into an antagonistic effect this result was indeed achieved by the synthesis of N-2,6dichlorophenoxyethyl-N-n-propyl-5,6-ADTN ~ , which showed antihypertensive properties on oral administration in spontaneous hypertensive rats. Investigation of the enantiomeric pairs of 33 and of the monosubstituted analogue 32 showed, as it was expected, that only the (S)-enantiomers were endowed with D 2 agonistic activity, but also pointed out a new, interesting feature whereas the N-propyl derivatives 33 was endowed with agonistic activity, 32 showed D 1 antagonist properties [26]. It appears that in the 5,6-ADTN series the receptor interaction provided by the N-propyl cleft is not only compatible with, but required for D 1 agonistic activity, confirming that (S)-5,6-ADTN and (R)-6,7-ADTN, although similar as shown in the model of McDermed, still behave very differently in the interaction of the Npropyl substituents with the D 1 receptor. We and others [27] thereafter applied similar structural modification in a series of dopexamine analogues, were significant D 1 activity had been previously reported [14], and achieved the synthesis of Z 1046 ~ . Compound 34 appeared to be endowed with D 1 activity comparable to dopexamine and with a marked D 2 activity, as well as with c~1-adrenergic antagonistic activity and with a moderate ot2-agonistic action, while it was devoid of any affinity for the 13-adrenergic receptors [28]. Vasodilatory and antihypertensive effects were observed on oral administration in rats, dogs, and monkeys [29], and were shown to depend on both D 1 and D 2 receptor stimulation, since they were fully antagonized only by the combination of SCH 23390 and domperidone. Thus ~ showing the desired pharmacological profile, may represent an interesting candidate for clinical investigation in congestive heart failure and in hypertension. Other groups have been continuing the pursuit of D 1 selective agonists as cardiovascular drugs, aiming at new compounds having full agonistic properties and increased potency with
75 OH
OH
HO
"~
HO
R
29, R = (CH2)6NH(CH2)2Ph, R 1 = H
31, R
~"/~~..'/N
CI ' ~
,
34
(CH2)6NH(CH2)2Ph' R1 =
OH
H~O
HO-
v
NH2
H
36, R = phenyi 37, R = 1-adamantyl
HO
HO-
v
v
HO HO
38, X = H 39, X = OH
-OH
35
NH
HO ~
~/CI-I ~ , '~,s-- N H ' ~
/(CH2)e
x
HO~,IH
|
k,,,,,,,,/
32, R = H 33, R = nPr
.~cH~
30, R = H, R 1 = ~
I. IJ
0~
H,
HO - v
v 40
H H
HO C ~ C l ~ . HO- v
'NH
41
respect to fenoldopam. A research programme at Abbott laboratories exploited new methods for 3D-database search [30]. At first a common pharmacophore for a number of D 1 and D 2 agonists was defined, relating the positions of the amino nitrogen and the catechol oxygen atoms, substantially in accordance with previous findings, then an aromatic ring was added to define a region accessible to D 1 agonists, but not to D 2 agonists, as suggested by the results of fenoldopam and related benzazepines. Among various attempts, a very active and selective benzopyran derivative, A-68930 ~ , was obtained by a relatively simple synthesis [3i]. Unexpectedly, the substitution of the phenyl ring by a branched alkyl substituent was shown to maintain activity, and the 1-adamantyl derivative, A-77636 ~ , showed potent activity and selectivity [3 2]. Other attempts to achieve D 1 selectivity by adding an aromatic ring to a flexible or rigid dopamine structure led to the synthesis of30 [33], 40 (dihydrexidine) [34], and 41 [35], while previous observations concerning the 3',4'-dihydroxy derivative of nomifensine were brought to a rational definition by the synthesis of 38 [6] and 39 (YM 43 5) [36]. Further modifications of the dopexamine structure led to FPL-63012 ~ [37], a selective D 1 agonist, and RS45496 35 [38], a mixed D1-D 2 agonist. The results of molecular modelling studies of these selective agonists are altogether consistent with the revised model of Kaiser (Fig. 1C), as far as lipophilic interactions, rather than H-H-stacking, are considered to be involved in the D 1-selecting site. The catechol moiety is preserved in all selective and non selective D 1 agonists considered so far, with the exception of pergolide ~ and some related ergolines. Compound 43, a monophenolic benzo(g)quinoline analogue of 7, was also shown to act as a D 1 and D 2 agonist, with 50% intrinsic efficacy ad D 1 receptors, by the work of group of investigators at Sandoz. This group set to prepare hybrids of dopamine and ergoline structures [6, 39], in the search of a common pharmacophore. The actions of 43 was totally shifted to D 2 selectivity by the insertion of the side chain of etisulergine 4(~), resulting in quinagolide ~ clinically effective as a prolactin release inhibitor. On the other hand, the addition of a fused aromatic ring to the ergoline structure resulted in compound CY 208-243 ~ , which was shown to be the first selective non-catechol D 1 agonist. Among these new D 1 agonists, clinical development in cardiovascular medicine is reported only for 39, while 37 and 45 are investigated for their potential antiparkinsonian properties.
76 X
.,, NHSO2NE
HN
H
H I CH3
H
42
H2N
I
//NH~ 48
H
H I CH3 45, X = H 46, x = OH
_
43, X= H 44, X = NHSO2NEt2 N
47
H
~X
r ~
H2N
CH3 49
50
H%~"
Investigation of new receptor subtypes and of their pharmacological relevance Starting from 1990, cloning of dopamine receptors of human and animal tissues, their expression in cultured cell lines, the analysis of their structure, as well as the availability of techniques of receptor imaging, boosted a new wave of investigation of dopamine pharmacology. The attention is now focussed on two subtypes of the D l_like family, namely D 1 and D 5, and three subtypes of the D2_like family, namely D2, D3, and D4, while other subtypes are on the horizon [40, 41]. The search for selective ligands, both agonists and antagonists, is inspired by different anatomical locations of the subtypes and by different pharmacological responses. The understanding of the latters is made complex by the uncommon wealth of transduction mechanisms that the dopamine receptors appear to be endowed with [42]. Hopefully, a "third generation" of precisely targeted and therapeutically useful dopaminergic agents shall arise from current research efforts. A first glimpse to the interaction of the main classes of dopaminergic agonists and antagonists with the new subtypes is provided by the extensive tabulation of affinity data reported by Seeman and van Tol [43], some of which are shown in Table 1. No selectivity was detected between D 1 and D 5 in these studies, but an over-simplified description of the Dl_like family appears undue. For instance, the selective benzazepine SK&F 83959 ~ showed high affinity, but variable action (from weakly agonistic to antagonistic) on adenylate cyclase stimulation in D 1 receptor preparations from different brain areas, and still potently induced Dl_like behavioural effects in laboratory animals [44, 45]. Interesting differences emerged in the ratio D2/D 3 and D3/D 4, both for agonists and antagonists. As for the latter, current interest for antipsychotic agents is directed to selective D 3 or D 4 agents, for different reasons. D 4 receptor density has been found to be markedly increased in postmortem examination of the brain of schizophrenic patients, and clozapine, an atypical neuroleptic having reduced risk of side effects, shows a good degree of selectivity for D 4 receptor, suggesting that at therapeutic levels in tissues (10-20 nM) it may act almost exclusively on this subtype [43]. D 3 receptors, on the other hand, are specifically located in the mesolimbic area of the brain, presiding to motivation, emotion, and cognition, whereas D 2 and D 4 are more evenly distributed and are present in striatal areas, presiding to locomotion [46]. Thus selective D 3 antagonists should be less liable to produce locomotor side effects, as well as disturbing side effects in female patients, such as amenorrhea and galactorrhea. The latters depend upon the inhibition of specific D 2 receptors in the anterior pituitary. As for agonists, compounds selective for the the various subtypes are investigated as potential antiparkinsonian drugs. For instance, D 1 agonists could be preferable, since they should not induce nausea and emesis. Still, in order to really ascertain the therapeutic value of
77
T a b l e 1. D i s s o c i a t i o n c o n s t a n t (K i, nM) f o r a g o n i s t s (A) a n d a n t a g o n i s t s (B). All v a l u e s refer to t h e h i g h - a f f i n i t y state, e x c e p t f o r D 3. M o d i f i e d with p e r m i s s i o n f r o m ref. 43
Receptor subtype A
B
Dopamine Apomorphine Bromocriptine Pergolide SKF 38393 7-OH-DPAT Chlorpromazine Haloperidol Clozapine (S)-Sulpiride SCH 23390
Dl-like
D2-1ike
D1
D5
D2
D3
D4
0.9 -- 0.7 - 440 0.8 1 ~ 5000
< 0.9
~ 7 ~ 0.7 ~ 8 - 0.8 - 150 10
~ 4 ~ 32 ~ 5 ~ 1.5 ~ 5000 ~ 1
~ 30 ~ 4 - 290 - 1000 650
3 1.2 ~ 230 -- 15 - 1100
4 - 7 ~ 170 ~ 13 ~ 800
35 2.3 21 1000 ~ 3000
~ 90 ~ 80 - 170 ~ 45 000 ~ 0.2
~ 450 -- 0.5 ~ 130 ~ 100 - 330 77 0 0 0 0.3
selective agonists, either alone or in combination with levodopa, a large effort in clinical trials appears mandative. On the side of psychopharmacology, the hypothesis of employing a selective agonist of the central presynaptic D 2 autoreceptors, such as preclamol (1..O), as an antipsychotic agent, is currently under new focus. Although it is now recognized that presynaptic and postsynaptic D 2 receptors are not structurally different, the presynaptic "autoreceptors" appear to be more sensitive to low concentration of agonists, and particularly of partial agonists. It has been proposed that the responsiveness to partial agonists depends on a considerably high reserve of the autoreceptors. Considerable interest is raised by the observation that presynaptic receptors in the mesolimbic area of the brain pertain to the D 3 subtype [47], and that preclamol ~ is equally active on this subtype. Analogues, such as the triflate 11 and the sulphoxide 12 [48] were found to act as antagonists, and to be somewhat selective for the D 3 with respect to the D 2 subtype, indicating that modifications of the phenolic group of 1..9.0 suppress its partial agonist character. On the contrary, it was unexpectedly shown that marked agonistic activity was preserved in the triflate ester of the full agonist (S)-5-hydroxy-N,N-di-n-propyl-2-aminotetralin [49]. Other structural modifications of the aminotetralins produced antagonists endowed with D 3 vs D 2 selectivity, such as (+)-UH 232 (53) [50] and (+)-S 14297 ~ [51], the latter with 301 ratio. On the other hand, the 13rotameric (R)-(+)-7-OH-DPAT ~ [46] and its rigid analogue PD 198907 (5~ [52] did show a similarly high selectivity, acting as agonists.
HO. N HOrN i/~
51
~,
52
~H~
OCH 3
~
53
54
Other previously "known D 2 agonists, such 8_, 16, ~ 21, and 48 [53] were also found to bind with high affinity to D 3 receptors. The last compound, pramipexole, had been structurally derived from talipexole ( 4 ~ [1], a D 2 agonist discovered within a series of ot2-adrenergic agonists. The stereochemistry of 4...S8and 19 suggests that they may fit the receptor in a similar way of (S)-5,6-ADTN (Fig. 1A) but the different activity of (+)- and (-)-49 [54], having
78 unsolved absolute configuration and respectively behaving as an autoreceptor agonist and a postsynaptic antagonist, raises new questions. A broad programme of investigation of potential D3/D 2 autoreceptor agonists at Parke Davis laboratories followed leads unrelated to dopamine, including piribedil (3), 20, ~ and features of antagonists of the butyrophenone and benzamide classes. PD 143188 (~3 emerged from these studies as a candidate antipsychotic agent [55]. The lack of hydrogen bond donor group in 50, as well as in the triflate of (S)-5-hydroxy-N,N-di-n-propyl-2-aminotetralin cast doubt on the requirement of such bonds for agonistic activity.
Progress in dopamine receptor models A new avenue of investigation of the interactions of agonistic and antagonistic ligands with receptor models directly derived from the structure of the protein is opened by the recent advancements. However, although the amino acidic sequences have been defined, tridimensional structures are not available for any of the members of the G-protein coupled family, to which dopamine receptors pertain. As a consequence, direct models built using theoretical methods of protein modelling and/or criteria of analogy are not exempt from criticism and controversy. Thus, progress in indirect modelling, based on steric and electronic properties of the ligands, plays an important role not only in the design of new active compounds, but also in the improvement and the validation of the direct models. In the search of indirect (ligand-based) models no substantial progress has been recorded for D 2 receptors with respect to the proposals of McDermed [7] and Kaiser [4, 5]. There is agreement among several investigators [56] on the main features, such as a plane defined by the oxygen atoms and the aromatic ring of dopamine, the protonated amine nitrogen approximately on the same plane, with its salt bridge to the binding site directed backwards and its two substituents extending far from the aromatic ring and slightly in front of the plane, respectively towards the "n-propyl cleft" and towards a region which can accomodate large groups. The distance from the m-oxygen and the nitrogen atom ranges between 6.2 and 7.3 A. In these models, it has been generally accepted that at least one phenolic or equivalent function, such as the indole NH group of the ergolines, acts as a hydrogen bond donor with respect to a site of the receptor. The recent discovery of potent dopamine agonists devoid of such functions suggests that recognition modes other than hydrogen bonding may be involved in the corresponding region of the receptor. Interesting considerations have been derived from molecular electrostatic potential (MEP) studies of the ligand. Subsequently to studies focusing on sets of structurally homogeneous ligands, a recent comparison of agonists pertaining to different classes [57] pointed out, in addition to the global minimum corresponding to the amino group, a second minimum located in the region of the aromatic/heteroaromatic moiety. The good matching of both minima in these agonists supports a role in the interaction with the receptor. In the case of the aromatic/heteroaromatic moieties, their I-I or lone-pair electron dense regions could interact either with a proton-donor group or with a H-stacking heterocyclic ring in the receptor. A suggestion that the phenyl ring of PD 143188, or the 2-pyridyl and p-hydroxyphenyl ring of two related, and somewhat more active analogues [58], interact in this way with Ser-194 and Ser-197 appears consistent with the hypothesis. The proposal of Kaiser [4, 5] for 1-arylbenzazepines, revised by using the most likely equatorial conformation (Fig. 1C) established the basis for a number of investigations of D 1 iigand-based, indirect models, analysing the position of the aryl substituent, considered as the selectivity-inducing feature, excluded by a steric barrier from interaction with the D 2
79 receptor. A recent comparison of the D 1 agonists of different structural classes [59] used quantum-mechanical methods and superimposed the lone pair of the amine nitrogen and the catechol ring. A site for the aryl ring could not be precisely defined, leading to the assumption that it is probably involved in a directionally non critical hydrophobic interaction in a lipophilic pocket, rather than a specific rI-rI stacking. The assumption is supported by the finding of selective agonists carrying an adamantyl, such as 37, or cyclohexyl moiety [31]. The close matching of the dipole vectors of structurally diverse agonists indicates a significant role of the electronic distribution in the recognition process [59]. An analysis of the MEP o f D 1 and D 2 selective and non-selective agonists [60] showed similarity of main features, regarded as primary requirements for binding, in agreement with Kocjan et al. [57]; it was pointed out that potentials in the proximity of the catechol ring are consistently lower in D2-selective than in D 1-selective compounds, suggesting that the difference may have some relevance in modulating selectivity. Accomodation of ergoline derivatives in D 1 and D 2 indirect models raised questions about the positioning of their heteroaromatic system, related both to the lack of phenolic groups and the ambiguity of the pyrrolethylamine/phenethylamine moiety participating in the pharmacophore. Seiler et al. [6, 39] proposed a superimposition of the ergolines, different from that provided by the model of Kaiser (Fig. 1C). In their model the NH group lays between the m- and p-phenolic groups not only in D 2 selective and D1-D 2 non selective ergolines, but also in the D 1-selective benzergoline 45. The hydroxy derivative 4_..66,devoid of selectivity, being 10 times more effective as a D 2 than a D l-agonist, was suggested to assume a position similar to that predicted by the Kaiser model. It is interesting to recall that 13hydroxy-lergotrile, a metabolite having a phenolic group in a position equivalent to 4...66was shown to be 10 times more active than its parent compound in the inhibition of prolactin release, a D 2 mediated effect [61 ]. On the contrary, the results of Alkorta and Villar [59] for the benzergoline 45 were in accordance with the model of Kaiser. Our analysis of six D 1 selective agonists, 14, 30, 3..66,38,
Fig. 2. D 1 receptor pharmacophore (amine nitrogen, blue; m-oxygen, red; centroid of the selectivity inducing aromatic ring, grey) with superimposed agonists. A 93..88,blue; 40, grey; 41, green; B " ~ blue; 36, grey; 30, green. A and B 945, red. See text for explanations.
80 40, and 41 by a systematic constrained search with Tripos' program RECEPTOR led to similar conclusions. The compounds were superimposed (Fig. 2) on a three-centres pharmacophore including the amine nitrogen, the oxygen atom corresponding to the m-position of dopamine and the centroid of the selectivity inducing aromatic ring. They fitted together in an almost identical way, both when equal geometrical weight was given to the three centres (as shown in the Fig. 2 A and B) and when the weight of the oxygen atom was decreased to 1/100. When the indole NH of 45 was assumed to be equivalent to the m-oxygen atom, and given equal weight, it was superimposed in accordance to Seiler et al. [6, 39], but the aromatic ring protruded out of the common aryl region (Fig. 2 B). On the other hand, when NH was released by assigning predominant weight to the other two centres, 45 was superimposed (Fig. 2 A) in accordance to Kaiser and to Alkorta and Villar. Direct (receptor-based) models of dopamine receptors, as well as of other G-protein coupled receptors, have been built on the analogy with bacteriorhodopsin, an evolutionarily related protein, whose tridimensional structure is known, although with moderate refinement. Notwithstanding the scarce sequence homology, the similarity of the topology of the seven transmembrane helices (TM1-7) was the starting point for the development of a number of Gprotein coupled receptors [62]. Models of dopamine receptors were built using primary sequence analysis, site-directed mutagenesis, and ligand structure-activity relationships [6265], frequently taking advantage of the similarity with the 132-adrenergic receptor. A main difference in the modelling approach was the use of the structural data of bacteriorhodopsin, either employed only as a 3D template for building the transmembrane helices from helical sequences otherwise assigned [62-64, 66], or also taken as the basis for the identification of the helical sequences [65]. Site-directed mutagenesis indicated that the aspartate residue in TM3 (Asp-114 in D 2 *) is essential for binding of both agonists and antagonists [67]. All models provide for a ionic interaction between Asp-114 and the charged amino group of the ligand. Most models also involve hydrogen bonding of the catechol system of dopamine and of related functional group of other agonists to Ser-193 (or Ser-194) and Ser-197 in TM5 [62-65], as in the examples of D 2 receptors shown in Fig. 3, B and C. In the model ofMalmberg et al. [64], shown in Fig. 3 B with 7_ as the ligand, the ionic interaction at Asp-114 is reinforced by surronding aromatic residues, as previously suggested [62], and Ser-193 is involved in the hydrogen bonding interaction with the 5-hydroxy group. Aromatic residues enclosing the binding pocket are Phe390, Phe-389, and Trp-386 at the bottom. The n-propyl cleft is defined by the residues Leu113 and Met-117 in TM3, Ph-389 in TM6, and Tyr-408 and Thr-412 in TM7. The aromatic moieties of larger aralkyl substituents, e.g. in compound 33, pointing in the extracellular direction can favourably interact with the aromatic moieties of Phe-110 in TM3 and Tyr-408 in TM7. The comparison of this model with the analogous model built on the D 3 sequence failed to point out important differences; it was suggested that the distance between Asp-110 and Ser192, being 1 A larger than the corresponding distance of Asp- 114 and Ser- 193 in D 2, may be more suitable to accomodate (R)-7-OH-DPAT, a D 3 70-fold selective agonist 5(5~, which shows a 0-7 to N distance of 7.4 A, vs 6.6 A of the somewhat less selective (S)-5-OH-DPAT. In the model of Teeter et al. [65] (Fig.3 C), similar features were pointed out; the hydrogen bonds were suggested to involve Ser-194 and Ser-197 in TM5, and the aromatic residues
* In the following and in Fig. 3 residues are numbered according to the human D 2 long isoform sequence (443 residues), unless otherwise annotated.
81
A
B
EXTRACELLULARSPACE
~.NH2
1M4 lU51
TM4
TM2
~'~
TM7 I Vd91
TU6
Asp114 Ile391 Pro388 Leu387 /'~ 11e384 Cys282 _..~
11tl !
Va187 ,Lcu4-1ql /./~o' 'Phi/3191 os
c~4151 kspso ~181
I
Asn124 Asr~221 : Set75
'"-~-
~
m5
Ash,52 I
3
~
~s
"x~ 'lrl~
o rpN6
/ /
,._
AspS0
.......
I
Fig. 3. Schematic representation of direct receptor models by 9A) Hutchins [66] Malmberg et al [64] C) Teeter et al [65], respectively holding compounds 36, 7, and 1.
B)
Trp-160 in TM4, Phe-389, Phe-390, and Trp-386 to build the flanks and bottom of the binding pocket. An ancillary site, flanked by Phe-110 in TM3 and Tyr-408 in TM7, suitable to accomodate large N-substituents, such as phenethyl, projecting upward and parallel to the helical axes, was similarly suggested. On the contrary, substituents longer than n-propyl projecting perpendicular to the axes appeared not to be accepted due to the hindrance by Thr412 in TM7. The aspartate residue in TM2 (Asp-80) is considered to be crucial for receptor activation and triggering of signal transduction [68], but its function at the molecular level is not clear. It was considered by Teeter et al. [65] to be involved in receptor regulation by Na +, along with Ser-121 and Asn-124 in TM3, and Ser-419 and Asn-422 in TM7, forming a square piramidal coordination site, directed from the middle to the intracellular face of the membrane. Hutchins [66] took a completely different approach, suggesting that Asp-80 actually interacts with the catechol system of dopamine (Fig. 3 A). In this way, the interaction of the catechol system with the serine residues on TM5 was disregarded, pointing out the scarce consistency of mutagenesis studies of these residues, but, on the other hand, a mechanism for receptor activation was hypothesized. Hydrogen bonding of the m-hydroxy group, or a functionally equivalent group, to Asp-80 would release some existing interactions of the latter with a set of residues, such as Asn-52 in TM1, Ser-75 in TM2, Asn-124 in TM3, and Asn-422 in TM7, located near to the intraceliular side of the membrane. This could start a conformational change relaying a signal transduction message to the G-protein. In this model the n-propyl site has been identified in a region projecting between TM3 and TM7 towards Val-87 and Val-91 on TM2. D 1 selectivity was analyzed, pointing out an additional pocket between TM6 and
82 TM7 accomodating the phenyl substituent. As indicated for 36 in Fig. 3 A, in the D 1 receptor, differing from D 2 for Cys-283 instead of Ile-384, and Phe-319 instead of Leu-414, the pocket has a suitably open shape, fixed by hydrogen bond linking of Cys-283 to the asparagine residue corresponding to Asn-418 in D 2. It has to be noticed that these models are limited to the transmembrane domains, necessarily neglecting the intracellular loops, which can anyhow exert an important role in binding and in signal transduction. For instance, chimeric modifications of the intracellular loop between TM5 and TM6 have shown its relevance in modulating agonist affinity and selectivity for D 2 and D 3 receptors (Robinson, 1994). Some remarks, and new questions
In the last few years significant progress has been recorded in the investigation of structural requirements for agonist selectivity on the new receptor subtypes, although there are yet no hints for selectivity at D 5 vs D1, and D 4 vs D 2 or D 3 receptors. As for D4, (+)-apomorphine has unexpectedly been shown to act as a selective antagonist [70], pointing out the interest of a new search for agonists in the previously fertile field of the aporphines. Selective D 3 agonists have been identified and are on the way to clinical investigation, particularly because of their action on mesolimbic autoreceptors. In the analysis of the interaction of dopamine agonists with the receptors old dogmas have been gradually dismantled, including the one of hydrogen bond donation, at least in D 2 and D 3 receptors. A recent report [71] also suggested, according to results of affinity modulation by GTP, that 13-(3-pyridyl)-N,N-di-n-propylphenethylamine, which is devoid of hydrogen donor groups and has nanomolar affinity for D 1 receptors, may behave as an agonist. Functional response data are desirable in order to confirm this interesting observation. The finding that recognition can involve electron rich, or polarizable, regions of agonist molecules widens the space of controversy and of further investigation on direct and indirect receptor models, including the possibility that either different sites or different conformations of receptors may fit different classes of agonists. In the case of the D 1 receptor, the most selective feature for recognition appears to be the occupation of a lipophilic site, forbidden for D 2 activity, near to the "mouth" of the receptor pocket, but other features also appear to favour D 1 activity : 1) the catechol system, although no longer considered as essential; 2) a far-away site in the receptor, accomodating an aryl group and a second amine nitrogen at two-three atom distance, in compounds such as dopexamine 2(~), 31, 34, and 35; and 3) a specific role of the n-propyl group in N,Ndisustituted derivatives of 5,6-ADTN such as 7, 33, and 43, which is supported by the observation that ~ the N-despropyl analogue of 33, behaves as an antagonist. These features frequently appear to cooperate in increasing the activity, suggesting that multiple interactions may help in a mutual conformational accomodation of some ligands and the D 1 receptor. The existence of different modes of interaction for different classes of agonists appears as a likely explanation in the case of the above 5,6-ADTN derivatives with respect to the primary amine 6,7-ADTN (4). The therapeutic targets of the development of a new generation of dopaminergic agonists remain mainly those that were identified for the first generation, where there is still a medical need of improved drugs. We can anyhow expect that the availability of potent and selective agonists for pharmacological and clinical investigations will lead to the discovery of new applications. Some suggestions, by way of example, may be derived by recent reports: a D 3 receptor agonist showed gastroprotective and antisecretory effects in rats [72]; dopamine
83 agonists may find application in conditions of alcohol [73] and opiate [74] dependence; D 4 receptors are expressed in relatively high density in cardiac tissues [75, 76], and their function awaits to be elucidated.
Acknowledgment : We are indebted to Dr. Andrea Zaliani for molecular modelling studies and for useful discussion.
REFERENCES 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31
Cannon JP. In: Jucker E, ed. Progress in Drug Research. Basel: Birkh~iuser Verlag 1985; 29: 303-412 Seeman P. Biochem Pharmacol 1982; 31:2563-2568 Goldberg LI, Kohli JD. In: Casagrande C, ed. Experimental Cardiology: Linking Laboratory and Clinical Research. Cormano - Milan: CNM 1993; 81-100 Kaiser C, Jain T. Med Res Rev 1985; 5:145-229 Katermopoulos HE, Schuster DI. Drugs of Future 1987; 12:223-253 Seiler MP, B61sterli JJ, Floersheim P, et al. In: Testa B, Kyburr E, Fuhrer W, Giger R, eds. Perspectives in Medicinal Chemistry. Basel: Verlag Helvetica Chimica Acta 1993; 221-237 McDermed JD, Freeman HS. In: Kohsaka M, Shohmoki T, Tsukada Y, Woodruff GN, eds. Advances in Dopamme Research. Oxford: Pergamon Press 1982; 179-187 De Marmis R, Hieble JP. Drags of Future 1989; 14:781-797 Wikstrom H. In: Ellis GP, Luskombe DK, eds. Progress in Medicinal Chemistry. Amsterdam: Elsevier 1992; 185-216 Soares-da-Silva P, ed. Cardiovascular and renal actions of dopamme. Oxford: Pergamon Press 1993 Casagrande C. Herz 1991; 16:102-115 Weinstock J, Hieble JP, Wilson III J. Drugs of Future 1985; 10:646-697 Casagrande C. In: Barnett DB, Pouleur H, Francis GS, eds. Congestive heart failure: pathophysiology and treatment. New York: M Dekker Inc 1993; 251-286 Brown RA, Brown RC, Hall JC, et al. In: Lambert RW, ed. Third SCI-RSC Medicinal Chemistry, Symposium. London: Royal Society of Chemistry 1985; 169-192 Fuller RW, Clemens JA. Life Sci 1991; 49:925-930 Pontiroli AE, Cammelli L, Baroldi P, Pozza G. J Clin Endocrmol Metab 1987; 65:1057-1059 Seyfried CA, Boettcher H. Drugs of Future 1990; 15:819-832 Casagrande C. In: ref. 10; 203-216 Simpkins JW, Bodor N, Enz A. J Pharm Sci 1985; 74:1033-1036 Ishikura T, Senou T, Ishidara H, et al. Int J Pharm 1995; 116:51-63 Lee MR. Triangle 1987; 26:11-21 Drieman JC, van Kaan FJPM, Thijsen HHW, et al. Brit J Pharmacol 1994; 111:1117-1122 Nishiyama D, Yoshikawa M, Yamaguchi I. Cardiovasc Drag Rev 1992; 10:101-116 Augstein J, Austin WC, Boscott RJ, et al. J Med Chem 1965; 8:365-371 Casagrande C, Galli A, Femni R, Miragoli G. Farmaco Ed Sci 1972; 27:445-470 Casagmnde C, Bertolini G, Montanafi S, Santangelo F. Colloque de Chimie Organique Fine, ERAI, Lyon 1991; 11 Santangelo F, Bertolmi G, Casagrande C, et al. U.S. Pat. 5.407956 Pocchiari F, AUievi L, Zanzottera D, et al. Can J Physiol Pharmacol 1994; 72 (suppl 1):132 Abs. P.1.9.61 Marchini F, Pradella L, Miragoli G, Semeraro C. Can J Physiol Pharmacol 1994; 72 (suppl 1): 131 Abs. P1.9.60 Martin Y. J Med Chem 1992; 35:2145-2154 DeNinno MP, Schoenleber R, Pemer RJ, et al. J Med Chem 1991; 34:2561-2569
84 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76
Kebabian JW, Briton DR, DeNinno MP, et al. Eur J Pharmacol 1992; 229:203-209 Springthorpe B, Dixon J., Inee F, et al. 12th Int Syrup Med Chem. Basel 1992; P-144-A Brewster WK, Nichols DE, Riggs RM, et al. J Med Chem 1990; 33:1756-1764 Mori N, Ishihara H, Matsuoka T, et al. Eur J Pharmacol 1990; 183:1049 Anan H, Tanaka A, Tsuzuki R, et al. Chem Pharm Bull 1991; 39:2910-2914 Smith GW, Farmer JB, Ince F, et al. Br J Pharmacol 1990; 100:295-300 McClelland DL, Lakatos I, Rosenkranz RP. Faseb J 1990; 4: A608, Abs. 1981 Seiler MP, Floersheim P, Markstein R, Widmer A. J Med Chem 1993; 36:977-984 Civelli O, Bunrow JR, Grandy DK. Ann Rev Pharmacol Toxieol 1993; 32:281-307 Jackson DM. Pharrnaeol Ther 1994; 64:261-369 Memo M. Mol Neurobiol 1990; 4:181-195 Seeman P, van Tol HHM., Trends Pharmacol Sci 1994; 15:264-270 Arnt J, Hyttel J, Sgmchez C. Eur J Pharmacol 1992; 213:259-267 Gnanalingham KK, Hunter AJ, Jenner P, Marsden CD. Biochem Pharmacol 1995; 49: 11851193 Levesque D, Diaz J, Pilon C, et al. Proc Natl Acad Sci USA 1992; 89:8155-8159 Murray AM, Ryoo HL, Gurevich E, Joyce JN. Proc Natl Acad Sci USA 1994; 91:11271-1"1275 Sonesson C, Lin CH, Hansson L, et al. J Med Chem 1994; 37:2735-2753 Sonesson C, Boije M, Svensson K, et al. J Med Chem 1993; 36:3409-3416 Haadsma-Svensson SR, Smith MW, Lin CH, et al. Bioorg Med Chem Letters 1994; 4:689-694 Rivet JM, Audinot V, Gobert A, et al. Eur J Pharmaeol 1994;265:175-177 Dijkstm D, Damsma G, Wikstr6m H, et al. 12th Int Symp Med Chem. Paris, 1994; P302 Lecrubier Y. Neuropsychopharmacol 1994; 10: (suppl 3) Abs. S- 16-91 Meltzer LT, Caprathe BW, Christoffersen CL, et al. J Pharmacol Exp Ther 1994; 266:11771189 Wright JL, Caprathe BW, Downing DM, et al. J Med Chem 1994; 37:3523-3533 For a review, see Johansson AM, Grol CJ, Karlen A, Hacksell U. Drug Design and Discovery 1994; 11:159-174 Kocjan D. J Med Chem 1994; 37:2851-2855 Downing DM, Wright JL, Mirzadegan T, et al. 206th ACS National Meeting, Chicago, August 1993, Abstract MEDI 172 Alkorta I, Villar HO. J Computer-Aided Molecular Design 1993; 7:659-670 Alkorta I, Villar HO. J Med Chem 1994; 37:210-213 Parli CJ, Schmidt B, Shaar CJ. Biochem Pharmacol 1978; 26:1405-1408 Trumpp-Kallmeyer S, Hoflack J, Bruinvels A, Hibert M. J Med Chem 1992; 35:3448-3462 Livingstone CD, Strange PG, Naylor LH. Biochem J 1992; 287:277-282 Malmberg A, Nordvall G, Johansson AM, et al. Mol Pharmacol 1994; 46:299-312 Teeter MM, Froimowitz M, Stec B, DuRand CJ. J Med Chem 1994; 37:2874-2888 Hutchins C. Endocr J 1994; 2:7-23 Mansour A, Meng F, Meador-Woodruff JH, et al. Eur J Pharmacol 1992; 227:205-214 Neve KA, Cox BA, Hermmgsen RA, Spanoyannis A, Neve RL. Mol Pharmacol 1991; 39: 733739 Robinson SW, Jarrie KR, Caron MG. Molecular Pharmacol 1994; 46:352-356 Seeman P, Van Tol HHM. Eur J Pharmacol 1993; 233:173-174 Claudi F, Cingolani GM, Giorgioni G et al. Eur J Med Chem 1995; 30:415-421 Glavin GB. Life Sci 1995; 56:287-293 Lawford BR, Young RMcD, Rowell JA, et al. Nature Med 1995; 1:337-341 Defonseca FR, Rubio P, Martmcalderon JL, et al. Eur J Pharmacol 1995; 274:1-3 O'Malley KL, Harmon S, Tang L, Todd RD. New Biol 1992; 4:137-146 Amenta F, Ricci A. 5th Int Conf Peripheral Dopamine, Kyoto, 1994; pg 22, Abs L-4
Perspective in Receptor Research
D. Giardin~, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All fights reserved.
85
Serotonin Receptor Complexity: Relationships and Roles T.A. Branchek Department of Pharmacology, Synaptic Pharmaceutical Corporation, 215 College Road, Paramus NJ 07652, United States of America
INTRODUCTION The serotonergic innervation of the brain arises primarily from a small and discreet set of raphe nuclei which send diffuse afferents to innervate virtually all levels of the CNS. This distribution suggests that 5-HT can influence neural activity in a wide variety of brain regions and may exert modulatory or integrative functions. In addition to its actions in the CNS, 5-HT also acts on cells and tissues throughout the rest of the body including smooth muscle, endothelia, and neurons. This heterogeneity of action appears to be served through a complementary complexity of receptor subtypes via which 5-HT exerts these actions. The rules for relating receptor subtypes to roles in serotonergic fimction remain elusive. Although the ubiquity of serotonin responses has been apparent for several decades, the precise definition of the molecular substrates underlying this apparent complexity is a recent achievement. At present genes for at least fourteen distinct serotonin receptor subtypes have been cloned and their expressed proteins evaluated by a variety of techniques. Progress has been made in the understanding of the structural relationships of these subtypes, residues in the amino acid sequences that are important for ligand recognition, their localization in the body, their signal transduction mechanisms, and some of their possible roles in physiology and pathophysiology. However, simple generalizations about this receptor system cannot be made. For example, there is no definitive evidence that points to any receptor subtype as having an exclusively CNS or peripheral location or action. For a given preparation whether it be a single enteric neuron, raphe neuron, or smooth muscle cell, there is not a single receptor subtype but often sets of 4-5 different ones. How signals mediated through these diverse subtypes are integrated with each other as well as with those from receptors which converge onto the same signaling pathways remains an active area of investigation. The more global understanding of the exact function of each of these subtypes awaits the discovery of selective chemical and molecular tools with which to refine our hypotheses.
STRUCTURAL RELATIONSHIPS INTRACELLULAR SIGNALING
OF
5-HT
RECEPTORS
AND
THEIR
Receptor subtypes for serotonin have been divided into seven distinct classes based on operational, structural and transductional properties [1]. There are five 5-HT~ receptor subtypes, three 5-HT 2 subtypes; one 5-HT 3 receptor, one 5-HT 4 receptor, two 5-HT 5 subtypes, one 5-HT6 receptor and one 5-HT7 receptor. Splice variants or additional
86 subtypes are likely for the 5-HT2c [2] and 5-HT2A[3] and have already been demonstrated for the 5-HT3 [4] and 5-HT 4 [5] receptors. With the exception of the 5-HT3 receptor which forms a ligand-gated ion channel [6], all other cloned 5-HT receptors are members of the G-protein coupled receptor superfamily. The ligand binding and activation or "operational" properties of these receptors has been extensively reviewed [1,7]. In general, the task to create sets of selective agonists and antagonists for each subtype remains a significant challenge for the future. The salient features of the categorization of 5-HT receptors based on commonality in structure and signalling are addressed below.
5-HT 1 Receptors
The five 5-HT~ receptors are termed 5-HTIA, 5-HTI o~, 5"HT~o~, 5-HT~E and 5-HT1F [1,7]. They each contain similar numbers of amino acids as follows: 421,377, 390, 365, 366, respectively for the human homologues. Each receptor is encoded by an intronless gene. At the structural level these proteins all share a "homology cluster" wherein there is mutual conservation of greater than half of their amino acid residues in the transmembrane spanning region which contains the ligand binding pocket for many biogenic amine receptors [8,9]. In contrast, the extracellular amino termini and loop regions of these seven helix transmembrane spanning proteins as well as their intracellular loops and carboxy termini lack observable homology that would facilitate categorization or prediction of their common intracellular coupling. Each of these 5-HT~ receptors couples to G-proteins which leads to the inhibition of adenylate cyclase activity and thus cAMP production [1]. The 5-HT~A [10], 5-HT~D~ [11], 5-HTIDp [11], and 5-HT~F [12] subtypes, also modulate intracellular Ca** through PTX-sensitive G-proteins. In cases where it has been evaluated, the 5-HTzE receptor has not been shown to do so [13]. In addition to these pathways, each of the 5-HT I receptors, with the exception of 5-HT1E [13] has been shown to increase the turnover of inositol phosphates [11,12,14,15]. Physiologically, the initial mediator of the action of these receptors may be through Gprotein-gated channels such as KGA [16]. Thus far, coupling of cloned 5-HT~ receptors to G-protein-gated K§ or Ca** channels has only been demonstrated for the 5-HTIA receptor [17]. Coupling of the 5-HT~A [14], 5-HT1D~ [18,19], and 5-HT~E [13] receptors to the stimulation of adenylate cyclase has also been demonstrated. However, this signaling pathway appears to be detected when relatively high levels of expression are attained relative to levels which can elicit a cyclase inhibition in the same cell host, or in the absence of a cyclase inhibitory response in a particular host. Therefore, it is tempting to speculate that these ancillary coupling pathways may result from a simple law of mass action in which all of the relevant Gi alpha subunits for a given receptor subtypes are "unavailable" and thus there is overflow to alternative pathways. Such a speculation is consistent with observation of preferential receptor-G-protein interaction when assayed using immunoprecipitation [20]. Future directions for the study of 5-HTI receptor signal transduction include quantitative analysis of subtype-G-protein interactions in reconstituted systems which do not rely on the variability of G-protein expression in host cells selected
87 for transfection based on incidental properties. In addition, coupling of the remaining cloned 5-HT I receptor subtypes to channels requires investigation.
5-HT 2 Receptors The 5-HT 2 receptors include 5-HT2A, 5-HTzB and 5-HT2c [ 1]. The proteins are similar in length, containing 471, 479, and 458 amino acids, respectively, for the human homologues. These subtypes possess a high degree of conservation in their amino acid sequences, contain over 70% identical amino acids in their transmembrane regions, and show highly conserved intron-exon boundaries [21]. All 5-HT 2 receptors couple to Gproteins which elicit phospholipase C activation and Ca++ mobilization. However, it appears that there may be some differences in the efficiency of coupling of these subtypes. Whereas both the 5-HTzA and 5-HTzc receptors show large stimulation of PI hydrolysis in many types of host cell, the 5-HTzB receptor gives a weaker response under similar conditions [21]. This recently cloned 5-HTzB receptor can also be observed in native systems such as the rat stomach fundus where it was originally described [22], and more recently in a teratocarcinoma cell line [23]. In this cell line, the 5-h 22B subtype couples to PI hydrolysis via a Gq protein. Coupling to both adenylate cyclase activation and inhibition has been reported for the 5-HT2c subtype [24] at high expression levels only. Interestingly, coupling to PI hydrolysis in the rat stomach fundus has never been demonstrated, although in this tissue it is clear that the receptor couples to Ca *+ influx through voltage sensitive Ca ++ channels, intracellular Ca+* release, and activation of PLC [25].
5-HT3 Receptors The 5-HT 3 receptor is a ligand-gated ion channel. Although there are differences in the literature in the apparent operational properties of this receptor in native preparations, they appear to be related to species differences. There has been no report of novel subtypes of 5-HT 3 receptor despite searching by many excellent investigators. The 5-HT 3 receptor was originally cloned from the mouse and the sequence predicts a protein of 487 amino acids [6]. This molecular mass is consistent with that reported for the solubilized receptorchannel complex [26]. An interesting structural property of the 5-HT 3 receptor is that it appears to be a homo-oligomeric receptor which distinguishes it from other ligand-gated ion channels such as those for acetylcholine or glutamate which are heteropentameric. The 5-HT3 receptor has also been cloned from the human and contains 478 amino acids [27]; however, no other characterization is available.
88 5-HT 4 Receptors
Cloning of the 5-HT4 receptor has finally been reported [5,28]. So far no subtypes of this receptor have been identified. However, a splice variant has been found in the rat which differs only in the carboxy terminus. The variants contain 387 and 406 amino acids for the rat receptors. No obvious operational differences have been detected in binding studies. Functionally, the cloned 5-HT4 receptor couples to the stimulation of adenylate cyclase activity. The two isoforms differ in their rates of desensitization, consistent with differences in phosphorylation sites in the carboxy termini [29]. No coupling to cyclase inhibition or to PI turnover has been detected.
5-HT 5 Receptors
The 5-HT5 receptors include 5-HT5A and 5-HT5B in rodents [1, 30, 31]. The rat receptors are proteins containing 357 and 371 amino acids. They share >75% amino acid identity in the transmembrane regions. In the human, the 5-HT5A has been cloned [32] and it has 84% overall amino acid identify to its murine homologue. Only an untranscribed fragment (one of the two exons) of the human 5-HT5B receptor has been detected [32] . If the absence of a full length cDNA for the human 5-HT5B subtype can be proven, it would be the first G-protein coupled receptor found in lower species but absent in man. Quite recently, a different group has reported the second exon of the human sequence and has found that it is interrupted by several stop codons [33]. Both Southern blot and PCR experiments indicate that this is the only homologue of the rodent 5-HTsB gene and thus, there will be no functional 5-HT5B receptor in the human. Since the function of the 5-HT 5 receptor in rodents is unknown, it is unwise to speculate on the significance of its possible loss in evolution. Although the signal transduction pathway of the vast majority of Gprotein coupled receptors is demonstrated readily after transfection into several host cells, coupling of the 5-HT 5 subtypes has remained somewhat mysterious until recently. Evidence now indicates that these subtypes may couple to the inhibition of adenylate cyclase activity [34]. Details of the response are not yet available. However, if the cyclase coupling is a robust and primary response, it is likely that the receptor will also couple to mobilization of intracellular Ca++.
5-HT 6 Receptors
The rat 5-HT 6 receptor is a protein of 436 amino acids [35, 36]. The human homologue contains 440 amino acids [36a]. This sequence contains at least one intron, located in the third intracellular loop. However, no isoforms have been cloned. Like the 5-HT4 receptor,
89 5-HT 6 receptor also couples to adenylate cyclase stimulation. Despite this similarity in coupling, however, there is no significant homology in the amino acid sequence of these two subtypes [5]. A detailed functional analysis has not been reported for the cloned 5HT 6 receptor. However, a native receptor which is detected in N 18TG2 cells, displays a rank order of agonist potency in cAMP assays which is similar to the affinity values for these compounds determined by radioligand binding in the same cell line [37]. It is likely that this site is similar or identical to the 5-HT 6 receptor. A detailed analysis of the N18TG2 cells using molecular techniques has not been reported.
5-HT 7 Receptors
The 5-HT 7 receptor is a third adenylate cyclase stimulatory subtype [1]. The human sequence encodes a protein of 445 amino acids [38]. The sequence appears to contain an intron in the carboxy terminus. Like the 5-HT 6 receptor, the 5-HT 7 subtypes lacks any homology to the other cloned 5-HT stimulatory receptors [5]. No inhibition of adenylate cyclase activity or coupling to PLC has been demonstrated for this subtype.
FUNCTIONS OF 5-HT RECEPTORS: STUDIES, TRANSGENIC ANIMALS
SELECTIVE LIGANDS, ANTISENSE
Although the majority of 5-HT receptor subtypes have been cloned, the task ahead remains to discover their distinct and/or interacting roles in physiology and pathophysiology. In order to do so, three main methods are under investigation. The classical approach is the development of subtype selective ligands with which to probe receptor function. The fastest approach is to employ antisense methodology to "knockdown" expression a discreet subtype of receptor. The third approach is to manipulate gene expression by homologous recombination and thus to "knock-out" or overexpress receptors.
USE OF SELECTIVE LIGANDS TO REVEAL RECEPTOR FUNCTION: 5-HT l
The clearest demonstration of 5-HT, receptor function which can be linked to therapeutic potential has come from the development of drugs which target the 5-HT~A and 5-HT~D receptors. The 5-HT~A partial agonists such as buspirone are clinically useful in the treatment of anxiety [39]. In addition, it has been suggested that antagonists of the 5-HTIA receptor, such as pindolol, may lead to a more rapid onset of antidepressant activity when combined with a selective serotonin reuptake inhibitor [40]. Although this particular
90 compound has pharmacological actions at receptors other than the 5-HT~A site, preliminary clinical observations indicate that the approach may hold promise [41]. Agonists of the 5-HT~D receptor subtypes have been developed for the acute treatment of migraine. Experimental data indicate that drugs such as sumatriptan may act on extracranial blood vessels to reduce vasodilation and/or on the terminals of the trigeminal-vascular afferent s to inhibit the release of peptides which mediate the sterile inflammatory response [42]. Clinically, sumatriptan is useful in the relief of acute migraine. Possible functions of the 5-HT~ and 5-HTlr receptor await the development of selective compounds or other approaches.
USE OF SELECTIVE LIGANDS TO REVEAL RECEPTOR FUNCTION:
5-HT 2
Much evidence has accumulated to link activation of the 5-HT2A receptor to hallucinations [43]. Agonists of this receptor subtype correlate best with the potency of several classes of hallucinogenic compounds including tryptamines and phenylalkyamines to induce hallucinations in people. Since this is not a clinically useful profile, focus has been placed on the development of antagonists for the 5-HTEA receptor. Many such compounds have been developed but their benefit has been elusive. Ketanserin, once thought to be a new antihypertensive agent via a 5-HT2A receptor mechanism, is now thought to exert that action via an alpha-1 receptor subtype. Several compounds with affinity for 5-HT2 receptors are now under investigation for a collection of CNS disorders including anxiety, depression, sleep disorders and drug abuse [44]. The 5-HTEAantagonists risperidone, sertindole, and amperozide all have been tested clinically as atypical antipsychotics [44]. These compounds combine 5-HT2A antagonist with D2 antagonist action and are reported to have a benefit for the negative symptoms of schizophrenia as well as a lower rate of extrapyramidal side effects. Drugs such as methysergide, used in the prophylactic treatment of migraine, are now speculated to act on 5-HT2s receptors [45]. However, there are many compounds which have mixed 5-HT2B and 5-HT2c activity and thus it is still difficult to assign a single subtype to the observed activities of these compounds. Potential therapies mediated by one or both subtypes include anxiety, depression, sleep disorders, and feeding disorders.
USE OF SELECTIVE LIGANDS TO REVEAL RECEPTOR FUNCTION: 5-HT 3
The 5-HT 3 antagonists, such as ondansetron and granisetron, have found clinical utility in the treatment of emesis, particularly for radiation or chemotherapy-induced cases [46]. Application to post-surgical and other forms of emesis are less well documented. 5-HT 3 receptors have also been speculated to be involved in cognition, anxiety, migraine, and pain. However, clinical proof of their role in these states is still lacking.
91 USE OF SELECTIVE LIGANDS TO REVEAL RECEPTOR FUNCTION: 5-HT4 Possible functions of the 5-HT4 receptor have been recemly reviewed [47,48,49]. In the CNS, 5=HT4 activation increases cAMP in the hippocampus [50]. This observation has led to behavioral studies using cognition models. 5-HT4 receptor activation induces relaxation of the esophagus [51,52] and increases cAMP accumulation in this tissue [53]. These data indicate a possible role for the 5-HT4 receptor in GERD. A selective 5-HT4 partial agonist, cisapdde, has been used clinically for gastroprokinesis for many years prior to the elucidation of its molecular site of action. Functional 5 - H T 4 receptors have also been demonstrated in the guinea pig ileum [54] and in the cardiac atrium, in which increases in cAMP accumulation can also be detected [55]. Such functions indicate possible roles of the 5-HT4 receptor in irritable bowel syndrome and in arrhythmia.
USE OF SELECTIVE LIGANDS TO REVEAL RECEPTOR FUNCTION: 5-HT 5, AND $-HT6 At present there are no selective ligands to evaluate 5-HT5 function. Possible i.n vivo roles of the 5-HT6 receptor may be related to neuropsychiatrie functions, based primarily on the high affinity of atypical antipsyehotic drugs and for several tricyclic antidepressants for this receptor [56]. Thus, it may modulate affective state. The localization of mRNA for the 5=HT6 receptor in the limbic system is consistent with this speculation.
USE OF SELECTIVE LIGANDS TO REVEAL RECEPTOR FUNCTION: 5-HT 7 There are no selective ligands available to study the 5-HT7 receptor. However, based on the activity profiles of a set of known compounds (which have additional receptor activity), the 5-HT7 receptor may play a role in neuropsychiatric processes. Like the 5-HT6 receptor, the 5=HT7 subtype has high affinity for several antipsychotic compounds and antidepressants [56]. In addition to these actions, many smooth muscle responses to 5-HT which result in relaxation [57,58,59] may be mediated by 5=HT7 receptors. In several tissues, positive linkage to cyclase stimulation has been detected. A recent study of relaxant responses of the ileum has [60] revealed pharmacological properties which are nearly identical to those of the cloned guinea pig 5=HT7 receptor [61]. Thus the 5-HT7 receptor may play a role in smooth muscle relaxation in irritable bowel syndrome or angina.
92 The possible roles of the 5-HT7 receptor in circadian phase shifts [62] has been hampered by the lack of selective compounds. However, based on the observation that 5CT, DPAT and 5-HT, compounds with affinity for both 5-HT~A and 5-HT7 receptor subtypes, can phase advance neuronal firing activity in hypothalamic slices in culture [63], and that a 5-HT~A antagonist was unable to block this response [20], it remains a plausible hypothesis. Whole cell voltage clamp recordings from rat suprachiasmatic nuclei neurons [64] also support a role for the 5-HT 7 receptor in circadian function. Proof of the role of 5-HT 7 receptors in such processes awaits the development of subtype selective tools.
USE OF ANTISENSE AND TRANSGENIC RECEPTOR FUNCTION: 5-HT 1 RECEPTORS
APPROACHES
TO
REVEAL
Antisense oligonucleotides can be administered in vivo in an attempt to uncover the function of receptors when their sequences first become available. As such, it is an attractive approach which has been applied to the study of G-protein coupled receptors over the last several years [65]. Employing such a methodology, there is no time lag for selective ligands or for the production of transgenic animals, both of which can be quite lengthy processes. The application of this technology to the study of 5-HT ! receptors has only come quite recently. In a preliminary report, antisense oligos directed against the 5HT~A receptor have been injected into rats in order to evaluate the role of this subtype in anxiety [66]. Behavioral data are not yet available. For the 5-HT~w~Da receptor, a transgenic knockout mouse has been produced and partially characterized at the behavioral level [67]. These animals display an increase in aggressive behavior in an intruder paradigm. Many other studies employing these animals are underway.
USE OF ANTISENSE AND TRANSGENIC RECEPTOR FUNCTION: 5-HT 2 RECEPTORS
APPROACHES
TO
REVEAL
Antisense oligonucleotides have been used to disrupt the function of 5-HT2Areceptors [68]. In a learned helplessness model for depression, treatment of animals with such tools led to a significant difference in the treatment vs the control group, indicating a possible role for 5-HT2A antagonists in depression. Transgenic animals have also been constructed which knock out the 5-HT2A receptor [69]. Unfortunately, this knock out leads to disruptions in cranial facial structure and results in lethality. Behavioral analysis of the heterozygotes was not complete due to these malformation. The future use of inducible knockouts may facilitate the study of the function of this receptor subtype using the heterologous recombination technology. More success has been obtained in animals with targeted disruptions of the 5-HT2c receptor [70]. These animals display profound disruptions of brain function. They are prone to sudden death as a result of seizure activity
93 and also display a significantly greater body weight than their wild type litter mates. Such an observation is consistent with previous pharmacological data linking 5-HT2c antagonists to reduction in food intake [70].
USE OF ANTISENSE AND TRANSGENIC APPROACHES RECEPTOR FUNCTION: 5-HT 5 AND 5-HT 6 RECEPTORS
TO
REVEAL
Antisense oligonucleotides targeted to the 5-HT 6 receptor subtype have been administered to rats and evaluated behaviorally [71 ]. In these studies, the animal exhibited a behavioral phenotype consisting of an increase in the number of yawns and stretches. This behavior was blocked by atropine, suggesting a role of the 5-HT6 receptor in the control of cholinergic neurotransmission. For the 5-HT 5 receptor, knockout mice are in progress but no behavioral data have been reported [33].
SUMMARY Many advances have been made in understanding the receptor substrates for the multiple actions of 5-HT. However, many fruitful areas of exploration remain ahead. The generation of receptor subtype selective approaches including ligands, radioligands, antibodies, antisense oligonucleotides, and transgenic animals has only begun. Even "wellcharacterized" receptors may hold new surprises. Further investigation of the molecular basis of ligand recognition are largely unexplored. Only a rudimentary understanding of signal transduction of these subtypes is extant. The next decade will likely see enormous progress in the understanding of this complex signaling system.
ACKNOWLEDGMENTS Many thanks to my colleagues at Synaptic Pharmaceutical Corporation and on the Serotonin Nomenclature Committee for helpful discussions and sharing preliminary information. Thanks to Ms. Lisa Gonzalez and Elinor Bernstein for preparation of the manuscript.
94 REFERENCES
Hoyer D, Clarke DE, Fozard JR, Hartig PR, et al. Pharmacol Rev 1994; 46(2): 157-204. 11,
Bonhaus DW, Bach C, DeSouza A, Salazar RFH, et al. 1995; 115: 622-628.
Brit J Pharmacol
Xie E, Bartkowski H, Chang L-S 1995; Soc Neurosci Abstr 21,774. ,
Hope AG, Downie DL, Sutherland L, Lambert JL, et al. Eur J Pharrnacol 1993; 245: 187-192. Gerald C, Adham N, Kao HT, Olsen MA, et al. EMBO J 1995; 14(12): 28062815. Maricq AV, Peterson AS, Brake AT, Myers RM, et al. Science 1991; 254: 432-437. Boess FG, Martin IL. Neuropharm 1994; 33: 275-317. Kobilka B. A Rev Neurosci 1992; 15" 87-114. Branehek TA. Med Chem Res 1994; 3: 287-296.
10.
Liu YF, Albert PR, J Biol Chem 1991; 266: 23689-23697.
11.
Zgombick JM, Borden LA, Cochran TL, Kucharewicz SA, et al. Mol Pharm 1993; 44: 575-582.
12.
Adham N, Borden LA, Schechter LE, Gustafson EL, et al. Arch of Pharmacol 1993; 348: 566-575.
13.
Adham N, Vaysse P J-J, Weinshank RL, Branchek TA. Neuropharmacol 1994; 33: 403-410.
14.
Fargin A, Raymond JR, Regen JW, Cotecchia S, et al. J Biol Chem 1989; 264: 14848-14852.
15.
Raymond JR, Albers FJ, Middleton JP, Lefkowicz RJ, et al. J Biol Chem 1991; 266: 372-379.
16.
Dascal N, Schreibmayer W, Lim NF, Wang W, et al. Proc Natl Acad Sci USA 1993; 90: 10235-10239.
95 17.
Karschin A, Ho BY, Labarca C, Elroy-Stein O, et al. Proc Natl Acad Sci USA 1991; 88:5694-5698.
18.
Maenhaut C, Van Sande J, Massart C, Dinsart C, et al. Biochem Biophys Res Commun 1991; 180:1460-1468.
19.
Van Sande J, Allgeier A, Massart C, Czemilofsky A, et al. Eur J Pharmacol 1993; 247: 177-184.
20.
Raymond JR, Olsen CL, Gettys TW. Biochemistry 1993; 32:11064-11073.
21.
Schmuck K, Ullmer C, Enges P, Lubbert H. FEBS 1994; 342" 85-90.
22.
Cohen ML, Fludzinski LA. J Pharmacol Exp Ther 1987; 243" 264-269.
23.
Loric S, Maroteaux L, Kellermann O, Launay J-M. Mol Pharmacol 1995; 47: 458466.
24.
Lucaites VL, Nelson DL, Yu L, Baez M. J Cell Biochem 1992(supp 16E); 228.
25.
Cox DA, Cohen ML. J Pharmacol Exp Ther 1995; 272: 143-150.
26.
McKeman RM, Biggs CS, Gillard N, Quirk K, et al. Biochem J 1990; 269: 623628.
27.
Miyake A, Mochizuki S, Akttzawa S, Kon G. DDBJ 1995; database submission, unpublished.
28.
Adham N, Gerald C, Vaysse PJJ, Weinshank RL, et al. Soc Neurosci Abstr 1994; 20:1266.
29.
Adham N, Gerald C, Vaysse PJJ, Weinshank ILL, et al. Soc Neurosci Abstr 1995; 21" 773.
30.
Matthes H, Boschert U, Amlaiky N, Grailhe R, et al. Mol Pharmacol 1992; 43" 313-319.
31.
Erlander MG, Lovenberg TW, Baron BM, De Lecea L, et al. Proc Nati Acad Sci 1993; 90: 3452-3456.
32.
Rees S, den Daas I, Foord S, Goodson S, et al. FEBS 1994; 355" 242-246.
33.
Grailhe R, Ramboz S, Boschert U, Hen R. Soc Neurosci Abstr 1995; 21" 1856.
96 34.
Ronken E, van Duk R, van Oostenbugge M, Kodden L, et al. Soc Neurosci Abstr 1995; 21: 1364.
35.
Monsma FJ, Shen Y, Ward RP, Hamblin W, et al. Mol Pharmacol 1993; 43 320327.
36.
Ruat M, Traiffor E, Arrang JM, Tardivellacombe J, et al. Biochem Biophys Res Commun 1993; 193: 268-276.
36a.
Hamblin M, 1995; personal communication.
37.
Unsworth CD, Molinoff PB. JPET 1994; 269: 246-255.
38.
Bard JA, Zgombick JM, Adham N, Vaysse PJJ, et al. J Biol Chem 1993; 268: 23422-23426.
39.
Traber J, Glasner T. TIPS 1987; 8: 432-437.
40.
Blier P, Montigny C. TIPS 1994; 15: 220-226.
41.
Artigas F, Perez V, Alvarez E. Arch Gen Psychiatry 1994; 51: 248-251.
42.
Moskowitz MA. TIPS 1992; 13:307-311.
43.
Glennon RM, Teitler M, McKenney JD. Life Sci 1984; 35:2505-2511.
44.
Baxter G, Kennett G, Blaney F, Blackburn T. TIPS 1995; 16(3): 105-110.
45.
Kalkman H. Life Sci 1994; 54: 641-644.
46.
Saxena PR. Pharmac Ther 1995; 66: 339-368.
47.
Bockaert J, Fozard JR, Dumuis A, Clarke DE. TIPS 1992; 13:141-145.
48.
Ford APDW, Clarke DE. Med Res Rev 1993; 13: 633-662.
49.
Kaumann AJ. TIPS 1993; 15: 451-455.
50.
Dumuis A, Bouhelal R, Sebben M, Cory R, et al. Mol Pharmacol 1988; 34: 880887.
51.
Baxter GS, Craig DA, Clarke DE. Arch Pharmacol 1991; 343: 439-450.
52.
Reeves JJ, Bunce KT, Humphrey PPA. Br Pharmaeol 1991; 103: 1067-1072.
97 53.
Moummi C, Yang DC, Gullikson GW. Eur J Pharmacol 1992; 216: 47-52.
54.
Craig DA, Clarke DE. J Pharmacol Exp Ther 1990; 252" 1378-1386.
55.
Kaumann AJ, Sanders L, Brown AM, Murray KJ, et al. Br Pharmacol 1990; 100: 251-258.
56.
Roth BL, Craigo SC, Choudhary MS, Uluer A, et al. J Pharmacol Exp Ther 1994; 268(3): 1403-1410.
57.
Trevethick MA, Feniuk W, Humphrey PPA. Life Sci 1984; 35: 477-486.
58.
Trevethick MA, Feniuk W, Humphrey PPA. Life Science 1986; 1521-1528.
59.
Fenuik W, Humphrey PPA, Watts AD. Eur J Pharmacol 1983; 96" 71-78.
60.
Eglen RM, Champney M, Carter D. 3RD IUPHAR Satellite Meeting on Serotonin 1994.
61.
Tsou AP, Kosaka A, Bach C, Zuppan P, et al. J Neurochem 1994; 63(2): 456-464.
62.
Lovenberg TW, Baron 13M, de Lecea L, Miller JD, et al. Neuron 1993; 11(3): 449-458.
63.
Prosser RA, Miller JD, Heller HC. Brain Res 1990; 534: 336-339.
64.
Kawahara F, Saito H, Katsuki H. J Physiol 1994; 478(1): 67-73.
65.
Hunter AJ, Leslie RA, Gloger IS, Lawrence M. 1995; TINS 18: 329-331.
66.
Miquel M-C, Sari Y, Sibella C, Kia HK, et al. 1995; Soc Neurosci Abstr 21" 1851.
67.
Sandou F, Djamel AA, LeMeur M, Ramboz S, et al. Science 1994; 265" 18751878.
68.
Papolos DF, Yu Y, Rosenbaum E, Gardener EL, et al. Neuroscience Abstract 1993.
69.
Toth M, Robinson T, Benjamin D, Shenk T. Soc Neurosci Abstract 1994.
70.
Tecott LH, Sun LM, Akana SF Strack AM, et al. Nature 1995; 374: 542-546.
71.
Bourson A, Borroni E, Austin RH, Monsma FJ Jr, et al. JPET 1995; 274: 173-180.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardin/L, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
99
Design and Synthesis of Agonists and Antagonists of the Serotonin 5-HT4 Receptor Subtype Daniel P. Beckerw Bella Goidstin (t), Gary W. Gullikson(t), Richard Loeffler (t), Alan Moormannw Chaffiq Moummi(t ), Roger Nosalw Dale Spanglerw Clara I. Villamilw Dai-C. Yang (t), Daniel L Zabrowskiw and Daniel L. Flynnw Departments of Medicinal Chemistryw and Gastrointestinal Diseases Research(t ), Searle Research and Development, Skokie, IL USA 60077
Introduction Serotonin is unsurpassed among monoamine neurotransmitters in the number of receptor subtypes reported to-date. Fourteen subtypes of seven major serotonin receptor classes have been identified in the central nervous system, the peripheral nervous system, myeloid/immune cell types, and smooth musculature of mammals [1]. While discreet, endogenous release of serotonin is able to manifest a variety of responses and behaviours, exogenous administration of serotonin (and nonselective mimetics) suffers from indiscriminate actions at all populations of accessible receptors. Hence, the discovery of subtype-specific agonists and/or antagonists to the various serotonin receptors have been sought as specific therapeutic agents. For some time, it has been known that serotonin exerts profound effects in the enteric nervous system (ENS). Gaddum and Picarelli originally characterized the serotonin-induced contractions of guinea pig ileal smooth muscle as being direct (D-receptor) and indirect (M-receptor) [2]. While the former receptor was later clearly defined as being the 5HT2 site, the ENS-mediated indirect contractions of the GP ileum were less clearly defined. In 1989, Dumuis et al reported that a series of prokinetic benzamides were found to a c t as agonists at the newly defined CNS 5-HT4 receptor subtype [3], which had been identified in mouse CNS colliculi neurons [4-5]. Subsequent studies by independent investigators confirmed the 5-HT4 agonist properties of benzamides, and demonstrated that the peristaltic reflex induced in the GP ileum by these benzamides was attributed to 5-HT4 agonism within the myenteric plexus [6-7]. While many benzamides have been disclosed over the last two decades, these compounds were designed either empirically or by rational
lO0
design to block other monoamine receptors, including the serotonin 5-HT3 receptor and the dopamine D-2 receptor. Therefore, while many of these compounds, including metoclopramide and cisapride, do interact with 5HT4 receptors, they also interact at other serotonergic, adrenergic, and dopaminergic sites. Our research goal was to design potent gastrointestinal prokinetic agents based upon a 5-HT4 receptor agonist mechanism. A parallel goal was to demonstrate that a class of 5-HT4 agonists could be selective as GI prokinetic agents, lacking untoward effects in other CNS [8] and cardiac tissues [9] where 5-HT4 receptors have been functionally described. Finally, an optimal goal was to identify a class of chemistry which would afford therapeutic dual 5-HT4 agonism/5-HT3 antagonism at such a dose which would be free of actions at other monoamine receptors, most notably antagonism at dopamine D-2 receptors. Such a dual 5-HT4 agonist/5-HT3 antagonist was considered ideal for the treatment of a variety of GI dysmotilities, including irritable bowel syndrome, gastroparesis, nonulcer dyspepsia, gastroesophageal reflux disease, emesis, and other abnormal states characterized by altered ENS sensory reflex mechanisms.
Molecular
Approach
Several indole analogs of serotonin were evaluated for 5-HT4 agonism in the recently described rat esophageal tunica muscularis mucosae (RatTMM) preparation [10]. Serotonin acts at 5-HT4 receptors in this preparation to relax the esophageal smooth muscle" pre-contracted with carbachol. We, and others [10-12] demonstrated that 5-HT [(1), EC50 = 10 nM] and 5-methoxytryptamine [(2), EC50 = 148 nM] are potent full agonists at the 5-HT4 receptor. However, tryptamine [(3), EC5o - 8000 nM] and 2-methyl-5HT [(4), EC50 - 20,000 nM] are much less active. See table 1. The activity of the phenolic-containing (1) and the methoxyl-containing (2) --- but dramatic decrease in activity of the 5-unsubstituted analog (3) --- suggested that a hydrogen-bond acceptor interaction is required at the R s-position of 5-HT4 agonists. The very weak agonist properties of 2-methyl-5HT (4) suggested an unfavorable A1,3 strain interaction between the 2-methyl group and the ethylamine side chain in the conformation(s) required for 5-HT4 agonism. Such A1,3 strain would predominate in conformers wherein Tau 1 = 180 ~ Figure 1 illustrates this set of conformers, wherein the side chain amino group may be positioned at rotamer sites A, B, and C [13].
101
Table 1 Agonist activities
of various
indoles NH2
R5
Proposed
Figure 1 5-HT4 Bioactive
D = 8.0 A for B, C rotamers; 7.6 A for A rotamer. .A.
R2
R2
R5
D N->L.P., A
A
L.P.
H Compd
Conformers
I
5-HT4 EC50, l.tM
1
H
OH
10
2
H
OMe
148
3
H
4
Me
H
-8,000
OH
~ 20,000
HIO ~ ~ ~ _ _ NR H Illustration of Tau 1 = 180 ~ conformers of 5-HT (R = H) and 2-Me 5HT (R = Me). The asterisks indicate the bonds defining the Tau 1 torsional angle. The amino group is oriented at rotameric sites A, B, or C.
Figure 2 illustrates a Ramachadran plot of energetically accessible conformations (< 2 Kcal/mole above global minimum) available to both The two extended/gauche serotonin (R = H) and 2-Me5HT (R = OH3)[14].
102
conformations and one fully extended conformation illustrated in Figure 1 are available to serotonin but not available to 2-Me5HT (compare local minima for Tau 1 = 180 o). When mapped as distances between the amino group and the proximal phenolic hydroxyl lone electron pair, D[N ~L.P.], the three exclusive conformations of 5-HT have distances out to 8 A for the two mirror image extended/gauche conformers (rotamers B and C), and out to 7.6 A for the fully-extended conformer (rotamer A). These distances are noted in figure 1.
Original
Aza-Adamantane
Series
We then modeled several of the previously reported benzamides shown to exhibit 5-HT4 agonism. Most benzamides, typified by the modest agonists metaclopramide and zacopride, exhibited D[N ~L.P.] of --- 7.1 A-- somewhat shorter than the distance of 7.6-8.0 A modeled with serotonin in the conformations proposed in figure 1. An initial series of azaadamantylcontaining benzamides of varying D[N-~L.P.] distances were produced which both confirmed our conformational analysis and provided a foundation upon which to design further analogs. Figure 3 illustrates the proposed functional group distance relationships for these benzamides, wherein the distal carbonyl lone electron pair serves as a surrogate for the serotonin
103
5-OH lone pair. Among the four syn- and anti-substituted adamantyl derivatives, the syn-aminomethyl compound (SC-54750) was predicted to be the most active analog due to the optimal DIN ~L.P.] of 8.1 ,&,. Evaluation of these four analogs confirmed our prediction. The synaminomethyl analog SC-54750 exhibits an EC50 = 74 nM in the ratTMM assay. See table 2. A second notable relationship emerged. While the syn-aminomethylene (n = 1) analog is the most potent analog, the next most potent member is the epimeric anti-normethylene (n = 0) analog SC51718. This suggested that a similar spatial orientation of the basic bridgehead 3 ~ amine is achievable for this set of syn/anti-epimeric analogs.
Table 2 A comparison of various aza-adamantyl ratTMM 5-HT4 agonist assay
containing
n ~
0
benzamides
in the
Anti ,n
n2rq Compound SC-51718 SC-51717 SC-54750 SC-54815
IMe Type Anti Syn Syn Anti
n 0 0 1 1
Distance, A 7.0 7.3 8.1 8.8
5-HT4 Agonism* 262 (107) 538 (36) 74 (12.8) >1000
~'ECso (SEM), nM. Rat TMM functional assay. Scaffold
Optimization
With the aza-adamantyl prototype in hand, we undertook a systematic effort to probe for subtle variations in 1) scaffold bulk, and 2) spatial orientation of the bridgehead nitrogen with respect to the benzamide attachment site. Additionally, the syn/anti-epimeric analog relationship at the attachment site was further developed. Figure 4 illustrates the target aza-noradamantane scaffolds 1-5. Each of these scaffolds is the
104
result of an enantiotopic removal of a bridging methano group from the parent aza-adamantane scaffold (I). The target number represents that compound produced by removal of the corresponding-numbered methano group from the orignal aza-adamantane scaffold.
R
benzamide acyl moiety
//
fl\
1 Aza-adamantane
1
n
-
pT
RHN
3_ _2 RHN
n
n
N
RHN
" /n /
RHN
Figure
Target Aza-noradamantane
4
\
/
4
scaffolds
selected
for
evaluation
Removal of the enantiotopic methano bridges 1 and 2 produced the corresponding aza-noradamantanes as shown in table 3. Compound SC52491 of Type 1 is very potent in the ratTMM assay, EC50 = 51 nM [15]. However, the enantiomer SC-52490 of Type 2 is 70-fold less active. This indicated that the combination of removal of the C-1 methano bridge and retention of the C-2 methano bridge leads to an aza-noradamantane with a
105 5-fold improvement in potency relative to the parent aza-adamantane SC51718 (cf. table 2).
Table 3 Aza-noradamantanes
of T y p e s
1 and
2:
SC-52491
Scaffold Q~
o .,,~,. A
Typel NH-R
H2N
R
I
~
....
Me
Compound
R,,
Type2
Type
Distance, ,i.
5-HT4 Agonism*
SC-52491
1
7.0
SC-52490
2
7.0
3,870 (1061)
7.0
262 (107)
SC-51718 (table 2)
51 (6.6)
* EC50 (SEM), nM. Rat TMM functional assay.
Removal of the C-3 methano bridge of the parent aza-adamantane afforded an opportunity not only to probe the importance of this methylene, but also to probe a different spatial orientation of the 3 ~ bridgehead amine lone pair (with respect to the benzamide attachment site). See figure 4, target (3). Moreover, this new aza-noradamantane (3) retains the desirable plane of symmetry of the parent aza-adamantane (I), obviating the requirement for asymmetric synthesis or resolution of racemic intermediates. Evaluation of both nor-methylene (n = O) and methylene (n = 1 ) s y n - / a n t i - pairs of this symmetric azanoradamantane again revealed that the epimeric analogs syn-(3C)/anti-(3A) exhibit the more potent 5-HT4 agonism. See table 4. However, these two compounds exhibit potencies similar to each other even though the 7.1 A distance of (3A) is considerably shorter, than the more ideal 7.9 A distance of (3C). Moreover, the potency of (3C), EC50 = 421 nM, is reduced compared to the analogous syn-aminomethyl aza-adamantane SC-54750, EC50 = 74 nM (cf.
106
table 2). The distorted spatial orientation of the 3 ~ amine lone pair compared to the aza-adamantyl system may explain this discrepency. In the event, however, this meso-azanoradamantyl system (3) offered no advantage over the original aza-adamantane series. Table
4
Meso-Azanoradamantyl
scaffold
of Type
o
3 Anti
n H
Syn
I Me
Type
n
3A
Anti
0
7.1
382 (24)
3B
Syn
0
7.3
712 (84)
3C
Syn
1
7.9
421 (87)
3D
Anti
1
8.6
660 (126)
Compound
Distance, ~.
5-HT4 Agonism*
* EC5o (SEM), nM. Rat TMM functional assay. As illustrated in figure 4, removal of the enantiotopic C-4 and C-5 methano bridges from the azadamantane series (I) would afford the 03, 7bridged pyrrolizidines of Types (4) and (5). Again, these derivatives afforded an opportunity not only to probe the role of these scaffold methylenes, but also to probe another unique spatial orientation of the 3 ~ bridgehead amine lone pair. Unfortunately, resolution efforts were unsuccessful in providing enantiopure derivatives (4) and (5) separately, requiring that comparisons be made using racemates. 5-HT4 agonist activites of this series is shown in table 5. The epimeric analogs exo (4/5-C)/endo (4/5-A) exhibit the more potent 5-HT4 agonism as anticipated. Additionally, the racemic exo-aminomethyl analog (4/5-C) having the closest distance D[N ~L.P.] (7.8 A) to that modeled for serotonin (8.0 A) exhibits the greatest 5-HT4 agonist potency, EC50 = 176 nM. Assuming that one of the (4/5-C) antipodes is responsible for activity,
107
this C3,7-bridged pyrrolizidine offers potency similar to the parent azaadamantane SC-54750, EC50 = 74 nM (cf. table 2). Table
5
C3,7-bridged
pyrrolizidines
0
/
CI H2N
'l
of
Types
(4)
and
Type 4
n
(5) Scaffolds
Type 5
exo
N H' ' ~ scaffold
exo
endo Me
Type
n
endo/exo
Distance, ,~
5-HT4 Agonism*
4/5 A
0
endo
7.0
542 (151)
4/5 B
0
exo
7.2
1518 (167)
4/5 C
1
exo
7.8
176 (58)
4/5 D
1
endo
8.4
415 (70)
* EC50 (SEM), nM. Rat TMM functional assay. Transition to Pyrrolizidine Scaffold There were two considerations which led us to probe this pyrrolizidinecontaining series more fully. First, we sought a pyrrolizidine system wherein enantiomerically pure isomers could be evaluated. Second, given the observation that the aza-noradamantane series of Type 1 (lacking the C-1 bridging methylene; see table 3), afforded the more potent compound SC-52491 (EC50 = 51 nM), we were interested if a similar improvement in potency would be obtained by removal of the analogous "C-1 like" bridging methano group from series (4/5).
Results from the enantiomeric pyrrolizidine types (6) and (7) are shown in table 6. With one exception ( e n d o 6/7-D), each of the exo- and e n d o- analogs was prepared by asymmetric synthesis as previously reported [16]. The type 6 antipodal series afforded the more potent 5-
108
HT4 agonists. Again, the epimeric analog pair (exo-6C), n = 1, and (endo6A), n = 0, are more potent than other members. Compound (exo-6C), SC53116, exhibits potency (EC50 = 17 nM) approaching that observed for serotonin (EC5o = 10 nM) in this rat TMM model. The key distance D[N--->L.P.]= 8.0 .A, of SC-53116 matches that proposed for serotonin (see figure 1, rotamers B and C). Table Potent
6 5-HT4 agonsim
by pyrrolizidine series:
SC-53116 Scaffold
Type 6 O
n NH
H2N
Me
COMPOUND TYPE SC-55967
SC-53116
Endo/Exo
Type 7
en0o -- - 7 exo
n
DISTANCE, A 0
7.0
endo
5-HT4 AGONISM*
6A
Endo
167 (27)
6B
Exo
0
7.1
6C
Exo
1
8.0
17 (4)
6/7-D
Endo
1
8.4
280 (38)
2369 (560)
7A
Endo
0
7.0
>3,000
7B
Exo
0
7.1
>3,000
7C
Exo
1
8.0
323 (46)
* EC5o (SEM), nM. Rat TMM functional assay. Selective ( $ 0 - 5 3 1 1 6 ) and Dual 5-HT3 blocking (SC-52491) Serotonin 5-HT4 Agonists Taken together, the rigorous analysis of the parent aza-adamantyl scaffold (I) led to the identification of azanoradamantane Type 1, SC52491 (EC5o = 51 nM) and pyrrolizidine of Type 6C, SC-53116 (EC5o = 17
109
nM) as potent 5-HT4 agonists. Receptor binding profiles for these two agents are shown in Table 7. Not surprisingly, both SC-52491 and SC53116 exhibit potent Ki constants, 29 nM and 5.2 nM, respectively) in a radioligand 5-HT4 receptor binding assay [17]. Neither compound exhibits affinity for 5-HTl-like, 5-HT2, dopaminergic, or adrenergic receptors up to 10 ~M [18]. SC-52491 exhibits potent affinity for the serotonin 5-HT3 receptor (Ki = 1.2 nM) [19]. However, SC-53116 is much less potent for 5HT3 receptors (Ki = 152 nM), affording a selectivity ratio of 30 in favor of 5-HT4 affinity. The affinity of both compounds for 5-HT3 receptors functionally results in pure antagonism by analysis in the von BezoldJarisch reflex model [20]. Thus, SC-52491 is regarded as a dual 5-HT4 agonist/5-HT3 antagonist, while SC-53116 is a more selective 5-HT4 agonist.
Table
7
Receptor
Profiles
C' o H2N
of
SC-52491
o
and
SC-53116
c,._
Me
SC-52491
Me
,yJ. o
COMPOUND
5HT-4*
5HT-4
5HT-3
SC-52491
51 (6.6)
29 (3)
1.2 (0.3)
SC-53116
17 (4)
5HT-1, 5HT-2, D-l, D-2, r >10K
5.2 (0.5) 152.0(1.0)
* EC50 (SEM), nM in rat TMM assay. appropriate binding assays.
SC-53116
>IOK
All other values are Ki (SEM), nM in
Development of 5-HT4 Antagonists The high 5-HT4 potency of the pyrrolizidine system prompted us to consider other applications of this scaffold. The N-methyl quaternary salt of SC-53116 was prepared and evaluated in both 5-HT4 ligand binding and functional assays. As shown in table 8, the quaternary salt SC-53785 is eight-fold less potent than the parent SC-53116 in the ratTMM functional assay (compare EC50s of 129 and 17 nM). However, radioligand binding studies revealed a divergent trend for 5-HT4 receptor affinity.
110
The quaternary salt is very potent, Ki = 350 pM, being more than an order of magnitude higher affinity than the parent SC-53116. This result suggested that the pyrrolizidine ring system could possibly be manipulated to afford 5-HT4 antagonists. The pyrrolizidine ester SC-55822 confirmed this hypothesis. While this compound did exhibit very high potency in the functional ratTMM assay, ECs0 = 1.5 nM, its maximal efficacy was only 57% that of serotonin. Binding studies revealed very potent affinity, Ki = 183 pM, for this compound.
Table 8 5-HT4 functional activity and affinity of pyrrolizidine analogs of SC-53116 ..... 5.HT4 ..... ECs0 (SEM), nM RatTMM Ki (SEM), n M
COMPOUND SC-53116
O
H 17.0 (3)
5.2 (0.5)
129 (10)
0.35 (0.05)
1.5 (0.2) 57 % E of 5-HT
0.183 (0.033)
Me SC.53785
o
CI~~o, H2N
NH Me
SC.55822 _
Me
le
H
Me
Encouraged by these results, we sought other aromatic acid ring systems for coupling to our pyrrolizidine scaffold. A series of indole-
lll
type pyrrolizidine esters was prepared and evaluated both for 5-HT4 agonist and/or antagonist activity using a modification of the ratTMM assay [12]. Among all compounds investigated, the N-methyl indazole (8) (pA2 = 8.56), 5-fluoroindole (9) (pA2 = 8.50), and the N-methylindole (10) (pA2 = 9.20) are particularly noteworthy. These compounds are illustrated in table 9. None of these antagonists exhibit agonist properties up to a ceiling test concentration of 10 ~M in this assay. Compound (10), SC56184, is comparable in potency to the recently described Glaxo antagonist GR-113808 (pA2 = 9.39) [21].
Table 9 Indole-like
pyrrolizidine
esters
as
Compound
(8)
O
5-HT4
antagonists 5-HT4 Antagonism
pA2, Rat TMM
H 8.56
Me
(9)
O
H 8.50
(10) O SC-56184 ~.~
H 9.20
Me
One short-coming of most previously reported 5-HT4 antagonists is that the aminergic-containing side chain is connected to the aromatic nucleus by an ester bond (e.g. GR-113808, SC-56184 and others). While this linkage is of little consequence for in vitro use, we considered that
112
5-HT4 antagonists wherein this ester linkage is replaced with the more metabolically stable amide bond might find broader utility for those studies requiring the in vivo administration of a 5-HT4 antagonist. After screening several chemical series, we were gratified to find that a series of imidazopyridine amides afforded moderately potent antagonists. It has been previously appreciated that 5-HT4 agonism requires a planar orientation of the amide bond and the aromatic ring system. As illustrated in figure 5, this feature contributes to the potent agonist properties of the ortho-alkoxy benzamides i, wherein intramolecular hydrogen-bonding maintains such a planar conformation. Esters ii have much less tendency to exist in this planar conformation due to the replacement of the H-bond donor amide with an ester oxygen. Although the imidazopyridine series iii has retained the amide NH group, it has less tendency toward intramolecular H-bonding due to the relatively weak Hbond acceptor properties of the imidazopyridine 2-N lone pair and also due to its distorted orientation by virtue of the strain invoked by the fused 5membered imidazole ring. o
o s, ec a,n
I
Alkyl
o side chain
s,Oec a,n
I
Alkyl
~N~
Figure 5 i, Benzamides
ii, Benzoates
iii, Imidazopyridines
intramolecular H-bond. 5-HT4 agonism
No intramolecular H-bond. 5-HT4 antagonism
No intramolecular H-bond. 5-HT4 antagonism
Table 10 illustrates the structures of two of these antagonists. Notably, the amide (11), SC-53606, is ten-fold more potent in the ratTMM assay than the corresponding ester (12); compare pA2 values of 8.13 and 7.0, respectively. We have recently reported that SC-53606 exhibits similar pA2 values in this preparation regardless of the 5-HT4 agonist used in the Schild analysis [12]. Thus, values of 8.13, 7.67, 7.63, and 7.68 have been observed for SC-53606 vs. 5-HT, 5-methoxytryptamine, renzapride, and SC-53116, respectively.
113
Table 10 5-HT4 antagonist
properties
(11) SC-53606 O
H NH
(12)
O
of selected
SC-53606
imidazopyridines
5-HT4 AGONISM ECso, RAT TMM
5-HT4 ANTAGONISM pA2 (SEM)
>10,000 nM
8.13 (0.06)
>10,000 nM
7.0 (0.05)
H
Profiles of 5-HT4 a n t a g o n i s t s Table 11 provides receptor profiling data on the partial agonist SC-55822, and the full 5-HT4 antagonists SC-56184 (indole ester) and SC-53606 (imidazopyridine amide). For comparison, data for the reference antagonist GR-113808 are also shown. The partial agonist SC-55822 exhibits potent affinity (Ki = 183 pM) for the 5-HT4 receptor in the ligand-binding assay, with a 280-fold selectivity vs. the serotonin 5-HT3 receptor (Ki = 51 nM). The full antagonist SC-53606 exhibits good potency in the 5-HT4 binding assay (Ki = 1.4 nM), with a 185-fold selectivity vs. the 5-HT3 receptor (Ki = 259 nM). The indole ester SC-56184 is a more potent 5-HT4 antagonist (Ki = 233 pM), but less selective (21-fold) vs. the 5-HT3 receptor. By comparison, GR-113808 exhibits a Ki = 57 pM with a 24,000-fold selectivity vs. the 5-HT3 receptor when evaluated under identical conditions. None of the compounds exhibit appreciable affinity for 5-HTl-like, 5-HT2, dopamineric, or adrenergic receptors. In vivo properties of the amide-containing antagonist SC-53606 will be reported separately.
114
Table
11
Receptor profiles of selected 5-HT4 antagonists O
H
Me O
Cl
SC-55822 H
H
NHH ~ , , . ~ ~ N
SC-53606
SC-56184 Me
COMPOUND
5HT-4
5HT-3
5HT-2
CZ-2
SC-55822
0.183 (0.033)
51.6 (5)
1,700 (100)
7,400 (800)
>10K
SC-53606
1.40 (0.5) pA2 8.13 (0.06)
259.0 (28)
> 10 K
>10K
>10K
SC-56184
0.233 (0.033) pA2 9.2
5.0 (1.0)
> 10 K
>10K
>IOK
1,400 (200)
> 10 K
>10K
>10K
GR-113808 0.057 (0.003) pA2 9.39 (0.10)
5HT-1, ~-1, [3-1/2, D-l/D-2
Ki (SEM) nM, except otherwise noted for pA2 values. Design
of
a
Scalable
Synthesis
of
SC-52491
Given the desired 5-HT4/5-HT3 receptor profile and pharmacological efficacy of SC-52491, a scalable synthesis was developed. The azanoradamantane ring system found in this agent possesses four contiguous chiral centers within its tricyclic framework. An asymmetric route was deemed to be superior to resolution of racemates, as previous efforts along the latter pathway resulted in difficult separations of fairly endstage intermediates [22]. A route based on an initial asymmetric Diels-Alder reaction is summarized in Scheme 1. Cycloaddition of a chiral fumarate ester (A)
115
with cyclopentadiene would afford the adduct (B). Regioselective Hofmann rearrangement of a derived exo amide would give the exo-protected amine (C), wherein all relative and absolute stereochemistry required for the elaboration of the substituted azanoradamantane is intact. Ozonolysis of olefin (C), with reductive work-up and subsequent primary amide formation, would afford the highly-substituted diol (D). Reduction of the amide, followed by a double-cyclization protocol, would afford the desired aza-noradmantane (F). Scheme 1 Asymmetric Synthetic
Design
to
Prepare
Azanoradamantane
F NHTos
~
'
A
B
OH
OH
,,,,,,,,,,~
H,N....rF,,,," ---~ (~
NHTos
D
CO2H
CO2R*
C
OH
,E-),,,,,,,,,,I,
....
OH
,
..
BOCHN
,
NHTos
E
F
Following the key finding of Helmchen [23], the bis-(S)-ethyl lactate ester of fumaric acid was prepared from fumaryl chloride, as illustrated in Scheme 2. Cycloaddition of this dienophile with cyciopentadiene to give (3) required some experimental modification in order to eliminate halogenated solvents. A survey of solvents and reaction conditions revealed that triethylamine as solvent allows both a reasonable reaction rate and high diastereomeric excess (d.e. = 93%) when the reaction is conveniently conducted at room temperature.
116
Scheme 2 Asymmetric
Diels-Alder
reaction
CH3
CIOC j
90%
R,O2C.j
1
TEA 2
3
CO2R*
d.e. = 9 3 %
The exo ester function was regioselectively manipulated as illustrated in Scheme 3. Basic hydrolysis of (3), followed by iodo-lactonization of the endo-acid onto the olefin, afforded the highly crystalline (4). Recrystallization of (4) afforded material of > 99% ee in 75% over-all yield from (2). The exo-primary amide (5) was prepared to set the stage for the subsequent rearrangement. Koser's method [24], utilizing hydroxytosyloxy-iodobenzene (HTIB), was employed for the modified Hofmann rearrangement. Tosylation of the exo-amine gave the tosylamide (6) in 72% overall yield. Reductive elimination of the iodolactone afforded norbornene (7)in high yield. Scheme 3 Synthesis of key norbornene (7) CO2R.
1) LiOH/H20 =
I
CO2H
1) S O C I
96%
CO2R* 75% from D.A.
5
4
I.
~
1) HTIB 76%
2) TsCI/py 95%
ONH 2
=
2) NH3
2) KI/12/H20 3
2
O ~)~ ~ " 0 6
NHTos
Zn/HOAc
~
95%
O
NHTos
C02H 7
ll7
Finally, the substituted norbornene (7) was transformed into the desired aza-noradamantane (11) as shown in Scheme 4. Ozonolysis of (7), with sodium borohydride work-up, gave the bicyclic lactone (8) quantitatively. Ammoniolysis of (8) afforded the primary amide (9), which was then reduced with borane/THF to afford (10) after BOC protection. Double cyclization was performed based on a modification of our earlier reported procedure [22]. Bis-tosylation, followed by TFA mediated removal of the BOC group, afforded the cyclization precursor in situ. Double-cyclization was effected by employing di-isopropylethylamine in acetonitrile at RT. Chromatography afforded azanoradamantane (11) in 78% yield. This material was conveniently stored for future use. After calcium metal reductive removal of the tosylamide group, the free amine (11, Tos = H) was coupled to the commercially available 5-chloro-2-methoxy-4-aminobenzoic acid to give SC-52491 as previously reported [22b]. This scalable synthesis of the azanoradamantane (11) is asymmetric, takes place with only one chromatography, and does not utilize halogenated solvents. Scheme 4 Completion of synthesis.
~
NHTos
OH
/,,,....... r~
O 3 0
CO2H
Aza-noradamantane (11)
//"" ~
NaBH4 100%
O
........../
NHTos
OH
/,,,,,,,,,,~ OH
NH3/MeOH 100~
H2N_...rr,,,,'" - - ~ / / ~ O NHTos 9
OH /
OH
1) TosCl/py
..........
2) BOC20 70%
I BOCHN
NHTos 10
H__~ ,
T o . . .
3) Hunig's/MeCN 4) chrom 78%
11
118
Conclusion A series of aza-adamantyl containg benzamides were initially made based upon a computationaily-derived model of 5-HT4 agonism. Further refinement of this series led to the very potent Type 1 aza-noradamantane SC-52491 (5-HT4 agonist EC50 = 51 nM; Ki = 29 nM). Other azanoradamantane types were not as active as those derived from Type 1. Further scaffold reduction led to a series pyrrolizidine 5-HT4 agonists, highlighted by SC-53116 (5-HT4 agonist EC50 = 17 nM, Ki = 5.2 nM). Pyrrolizidine containing 5-HT4 antagonists were also identified. SC56184 (Ki = 233 pM; pA2 = 9.2) and SC-53606 (Ki = 1.4 nM; pA2 = 8.14) were shown to exhibit no agonist activity up to 10pM concentration. Both compounds also are 5-HT3 antagonists. Otherwise the compounds are quite selective vs. other serotonergic, adrenergic, and dopaminergic receptors. Finally, a scalable synthesis of SC-52491 was developed to facilitate its preclinical development. This synthesis is highlighted by an asymmetric Diels-Alder reaction and a novel double-cyclization step to produce the aza-noradamantane ring system. No halogenated solvents are employed and only one chromatography is incurred late in the synthesis. The overall yield for this process currently stands at 25%.
References
1
Hoyer D, Clarke DE, Fozard JR, Hartig PR, et al. Pharmacological Reviews 1994; 46: 157-203.
2.
Gaddum JH, Picarelli ZP. Br. J Pharmacol Chemother 1957; 12: 323328.
3.
Dumuis A, Sebben M, Bockaert J. Naunyn-Schmied Arch Pharmacol 1989; 340: 403-410.
4.
Dumuis A, Bouhelal R, Seben M, Cory R, et al. 34: 880-887.
5.
Bockaert J, Fozard JR, Dumuis A, Clarke DE. Trends in Pharmcol Sciences 1992; 13: 141-145.
6.
Buchheit K-H, Buhl T. Eur J Pharmacol 1991; 205: 203-208.
Mol Pharmacol 1988;
119
7.
Taniyama K, Nakayama S, Takeda K, Matsuyama S, et ai. J Pharmacol Exp Therap 1991; 258: 1098-1104.
8.
Fagni L, Dumuis A, Sebben M, Bockaert J. Br J Pharmacol 1992; 105: 973-979.
9.
a) Villalon C, der Boer MO, Heiligers J, Saxena PR. Br J Pharmacol 1990; 100: 665-667. b) Ouadid H, Seguin J, Dumuis A, Bockaert J, et al. Mol Pharmacol 1992; 41: 346-351. c) Sanders L, Kaumann A. Naunyn-Schmied Arch Pharmacol 1992; 345: 382-386.
10.
Baxter GS, Craig DA, Clarke DE. Naunyn-Schmied Arch Pharmacol 1992; 343: 439-446.
11.
Ohia SE, Cheung Y-D, Bieger D, Triggle CR. Gen Pharmacol 649-658.
12.
Yang D-C, Goldstin B, Moormann A, Flynn D, Gullikson G. J Pharmacol Exp Therap 1993; 266:1339-1347.
13.
Spangler DP, Flynn DL. Presentation at 204th National American Chemical Society Meeting, Division of Medicinal Chemistry Symposium: Molecular Design Strategies in New Drug Discovery, Washington, D.C., Aug 23-26, 1992.
14.
Conformational analysis was performed for serotonin and 2-methyl serontonin varying tau 1 and tau 2 at 15 degree increments. For each conformation all other coordinates were optimized using MacroModel/BatchMin Version 3.1 with a modified MM2 force field (C. Still, Columbia University).
15.
Flynn DL, Becker DP, Nosal R, Gullikson GW, et al. Bioorg Med Chem Lett 1992; 2: 1613-1618.
16.
Flynn DL, Becker DP, Nosal R, Villamil CI, et al. J Med Chem 1992; 35: 1486-1489.
17.
Grossman CJ, Kilpatrick GJ, Bunce KT. 624.
1992; 23:
Br J Pharmacol 1993; 109: 618-
120 1 8.
Radioligands used for receptor profiling studies: [3H] 5HT for 5-HT1like receptors; [3H] ketanserin for 5-HT2 receptors; [3H] SCH23390 for D-1 receptors; [3H] spiperone for D-2 receptors; [3HI prazosin for alpha-1 adrenergic receptors; [3H] rauwolscine for alpha-2 adrenergic receptors; [3HI dihydroalprenolol for beta receptors.
19.
[3H] zacopride was used as the radioligand. Pinkus LM, Sarbin NS, Barefoot DS, Gordon JC. Eur J Pharmacol 1989; 168: 355-362.
20.
Saxena PR, Lawang A. Arch Int Pharmacodyn 1985; 277: 235-252.
2].
Flynn DL, Becker DP, Spangler DP, Nosal R, et al. Presentation at 206th National American Chemical Society Meeting, Division of Medicinal Chemistry Symposium: Antagonists to the Serotonin 5-HT4 Receptor Which are Based on a Pyrrolizidine Scaffold. Chicago, Illinois Aug 22-27,1993
22.
a) Flynn DL, Zabrowski DL, Nosal R. Tet Lett 1992; 33: 7281-7282. b) Flynn DL, Becker DP, Nosal R, Zabrowski DL. Tet kett 1992; 33: 7283-7286.
23.
Helmchen G, Goeke A, Lauer G, Urmann M, et al. Angew Chem Int Ed Engl 1990; 29: 1024-1025.
24.
Lazbin IM, Koser GF. J Org Chem 1986; 51: 2669-2671.
Perspective in Receptor Research
D. Giardin~, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
121
t~l-Adrenergic R e c e p t o r Subtypes and Signal T r a n s d u c t i o n T.L. Theroux, T.A. Esbenshade, and K.P.Minneman Department of Pharmacology, Emory University, Atlanta, GA 30322, USA
ABSTRACT Two pharmacologically distinct ~l-adrenergic receptors (ttXA and alB) have been described in animal and human tissues using selective antagonists and site-directed alkylating agents, whereas three different al-adrenergic receptor subtypes have been cloned (ttlb. Gilt, and ttl~/d), all of which are members of the G protein linked receptor superfamily. The relationship between native and cloned subtypes was a source of much confusion. However, recently a consensus was reached that the ale clone encodes the pharmacological a ~g subtype, and that both the axb and ttl~/d clones encode receptors with ~tlB-like pharmacology. It is now agreed that the cloned and native receptors should be called ~a/alg, aXb/~B, and ttld/~m, and the tz~cdesignation has been dropped. Initial studies in isolated smooth muscle tissue suggested that the pharmacologically defined {Xlg subtype preferentially activates voltage-gated Ca 2+ influx while the ttlB subtype stimulates formation of inositol (1,4,5) trisphosphate (IP3*) and mobilizes intracellular Ca 2+ stores. Although these two signalling pathways are generally associated with different receptor subtypes, further work with both native and recombinant subtypes has provided exceptions to this rule. For example, tt~g-adrenergic receptors are linked to inositol phosphate (InsP) formation and mobilization of intracellular Ca 2+ in human SK-N-MC neuroblastoma cells, and tz~b- and a ld-adrenergic receptors are linked to voltage-gated Ca 2+ influx in rat medullary thyroid carcinoma (rMTC 6-23) cells. The exact mechanisms by which tZladrenergic receptor subtypes initiate these signals in native cells and tissues are still unclear. When expressed in mammalian cells, all three cloned subtypes can stimulate InsP formation and release intracellular Ca 2+, however the efficiency with which each subtype couples to this signalling system appears to vary (ttla>ttlb>ald). No model system is currently suitable for similar studies examining tt~-adrenergic receptor modulation of voltagegated Ca 2+ influx in response to transfection of the cloned subtypes. These subtypes also activate other second messenger systems following transfection, including phospholipase A2 (PLA2) and adenylate cyclase. Use of inducible vectors to titrate the density of each cloned subtype in transfected cell lines allows direct determination of the efficacy of each subtype in activating second messenger responses.
*Abbreviations used are as follows: AR, adrenergic receptor; [Ca2+]i, intracellularcalcium; CEC, chloroethylclonidine: DAG, diacylglycerol;EPI, epinephrine; CNS, central nervous system; HEK 293, human embryonic kidney 293 cells; IPTG, isoproply-13-D-thiogalactoside;InsP, inositol phosphate; IP3, inositol (1,4,5) trisphosphate; NE, norepinephrine; PI, phosphatidylinositol; PLA2,phospholipaseA2; PLC, phospholipaseC; PLD, phospholipase D; PKC, protein kinase C; rMTC 6-23, rat medullarythyroid carcinoma cells; VDCC, voltage-dependent Ca2§channel.
122 INTRODUCTION Norepinephrine (NE) and epinephrine (EPI) act as neurotransmitters and hormones in both the peripheral and central nervous systems (CNS). NE is released from neurons throughout the CNS and periphery to participate in a variety of physiological functions, while both NE and EPI are released from the adrenal medulla in response to stress. NE and EPI modulate fluid homeostasis, cardiac function, energy metabolism, and may play a role in depression. At the cellular level, these actions are mediated by multiple adrenergic receptor (AR) subtypes and second messenger systems. Adrenergic receptors are part of a large family of G protein coupled receptors. There are three well defined classes of ARs: 13-ARs, al-ARs, and ~2-ARs. The 13- and iz2-ARs are positively and negatively coupled to adenylyl cyclase through Gs and Gi, respectively. The al-ARs trigger an increase in intracellular Ca2+ concentration ([Ca2+].), through two pathways. a l-ARs couple to the Gq class of G proteins to activate PLC, generating I P 3 and causing a release of intracellular Ca 2+ stores, a l-ARs can also couple to voltage-dependent Ca 2+ channels (VDCC) in the plasma membrane allowing influx of extracellular Ca 2+. Members of the a x-AR family have been characterized based on antagonist selectivity, tissue distribution, coupling mechanisms, and molecular cloning. Selective antagonists distinguish between ~IA and a lB-AR subtypes [1] in various tissues and cell lines (uppercase letters designate pharmacologically defined subtypes while lowercase letters indicate cloned subtypes), a IA-ARs are expressed in a wide variety of tissues and generally couple to VDCCs [2,3], although this subtype can also couple to InsP formation and the release of intracellular Ca 2+ stores in certain tissues [4] and cell lines [5]. ~IB-ARs generally couple to InsP production and release of intraceUular Ca 2+ stores, although this subtype also couples to VDCCs in certain in vitro systems [6]. Molecular cloning has revealed three distinct subtypes: ala, ~lb, and a~d [7,8,9,10]. In expression systems, the three subtypes demonstrate distinct pharmacological profiles. The a la shows a lA-like antagonist pharmacology, while both the a lb and kid clones demonstrate pharmacological characteristics more similar to the pharmacologically def'lned tt IB-AR. New compounds have recently been developed which distinguish between the a lb and a ld subtypes [11]. All three clones couple to phospholipase C (PLC) and InsP generation in a variety of expression systems. The ability of each clone to couple to VDCCs has not yet been examined, since there is as yet no good expression system for examining this coupling. Other a~-AR subtypes have also been proposed. In particular, Muramatsu et al. [12] described an afAR subtype exhibiting a low affinity for prazosin. This subtype has recently been proposed to mediate the contraction of prostatic smooth muscle [18] and therefore could play a key role in the development of more effective drug therapies to treat benign prostatic hypertrophy. Unfortunately, it has been difficult to identify this subtype with radioligand binding approaches, and its relationship to the cloned subtypes is currently uidmown. More selective compounds, further functional studies, and the cloning of this receptor subtype will help to clarify this issue. The existence of multiple a l-AR subtypes coupled to multiple second messenger systems and co-expressed in a variety of tissues complicates studies aimed at defining the role of each receptor subtype. Expression of the cloned subtypes and the development of subtype selective compounds will help to evaluate the function of this class of receptors.
123 P H A R M A C O L O G I C A L CHARACTERIZATION OF CZlA-AND cxm-AR SUBTYPES Morrow and Creese [ 1] demonstrated multiple ,t~-AR subtypes by differences in their affinities for phentolamine or WB 4101 (both rigA-selective compounds) and prazosin (nonselective) in various rat tissues. The high and low affinity binding sites for these selective compounds are designated a lh- and *tlB-ARs, respectively. Other antagonists, such as (+)niguldipine and 5-methylurapidil, also have a relatively high affinity for the ,tlA-AR subtype as compared to the tt 1B-AR subtype [ 13, 14]. Chloroethylclonidine (CEC) is currently the only known antagonist exhibiting ,t 1Bselectivity. CEC binds to both receptor subtypes, but alkylates only the *tlB and not the a~A subtype [15]. Using these pharmacological tools, tissues were characterized as expressing heterogeneous or homogeneous populations of these two a l-AR subtypes and distinct biochemical and physiological responses were attributed to each subtype. Functional studies revealed tZlA-ARs are commonly expressed in the vasculature mediating the contractile responses to NE and EPI. Other tissues expressing primarily ,t 1A-ARs, as assessed by binding assays, include regions of the brain, vas deferens, the submaxillary gland, and prostate, a mARs were found to be the major subtype expressed in spleen and liver (rat), also assessed through binding studies. Functional studies revealed that the ,tin subtype consistently couples to InsP generation and release of intracellular Ca 2§ stores. Recently, more selective drugs have been identified or synthesized, allowing a more precise pharmacological analysis and reevaluation of the functional differences between these subtypes (Table 1).
Table 1.
Highly selective antagonists for a t-AR subtypes. Antagonist (+)niguldipine 5-methylurapidil BMY 7378 RS 17053 SNAP 5089 chloroethylclonidine
Selectivity a 1g a 1A axd tXla
axa tt lb and a 10
Reference [16] [ 17] [11] [18] [19] [20]
CLONING OF al~-, a lb- AND IXld-Ag SUBTYPES The first al-AR subtype to be cloned was from a hamster DDT1 MF-2 cell line [7]. When expressed in heterologous systems, this clone demonstrates classical ~m-AR pharmacology, with a low affinity for tt 1A-selective antagonists and a high sensitivity to CEC inactivation. Northern blot analysis also demonstrated that this clone shows an tim-like tissue distribution, mRNA expression and the pharmacological profile of this clone led to its identification as the a lb-AR. The a la-AR was first cloned from bovine brain [8], but was originally thought to
124 encode a novel subtype called a lc. When expressed in COS-7 cells the agonist and antagonist pharmacology of this subtype was characteristic of the defined GtIA-AR, with the exception of CEC. The expressed receptor showed a marked sensitivity to CEC inactivation, unlike the native UIA subtype. In addition, Northern blot analysis and in situ hybridization of various bovine, rat, rabbit, and human tissues revealed expression only in human dentate gyms and rabbit liver. In contrast, the pharmacologically defined ~xA-AR is known to be widely expressed in many tissues based on both antagonist binding and functional assays. Such observations led to the classification of this clone as a novel subtype (Ulc)- However, when the human [21] and rat [22] homologs were cloned, rnRNA levels were detected in the tissues previously characterized as expressing a~A-ARs (vas deferens and submaxillary gland, among others), contradicting the previous findings for the bovine clone. This a~-AR sequence was renamed a~a. The a ld-AR was cloned from rat [9, 10] and, although there was some initial confusion concerning the identity of this receptor based on initial mRNA distribution studies [9], this clone was found to have an ~B-like pharmacology. Like the a lb clone, expression of this clone resulted in a receptor with a low affinity for many a~A-selective antagonists (phentolamine, WB4101, (+)niguldipine, and 5-methylurapidil) and a high sensitivity to alkylation by CEC. Recently, BMY 7378 has been found to be selective for the a xd-AR subtype, giving us a tool to better characterize this receptor. Cloning of these subtypes and their human homologs allows for a more definitive pharmacological characterization, and facilitates investigation of both their physiological functions and biochemical coupling mechanisms.
IS THERE A SUBTYPE WITH A LOW AFFINITY FOR PRAZOSIN? An al-AR subtype with a relatively low affinity for prazosin was proposed by Flavahan and Vanhoutte [23] in 1984. The pharmacology of this subtype was further described in 1990 by Muramatsu et al. [12] based on contractile studies of various blood vessels from four different species: dog, rabbit, guinea-pig, and rat. The a I-AR subtypes expressed in vascular smooth muscle from these sources were classified into three groups based on the difference between the pA2 values for prazosin and for either WB4101 or HV723. One of these groups, classified as the a lL-AR subtype, shows a low affinity for all three compounds. Evidence that this subtype is also expressed in human prostate was found by Ford et al. [18]. A new ala selective antagonist, RS 17053, demonstrates an approximately 100-fold selectivity in binding at the a la-AR subtype over the a lb and ttxa subtypes (based on studies with both cloned and native subtypes). This compound potently inhibits NE-stimulated contraction of isolated perfused kidney (expressing the tt 1A-AR subtype) and weakly inhibits the NE stimulated contraction of aortic rings (expressing the a lB or alD-AR subtype), verifying its predictable activity in functional studies. In contrast, RS 17053 only weakly inhibits the contraction of prostatic smooth muscle. This is surprising, since previous correlation studies had concluded that the a lA-AR is the predominant subtype expressed in prostate and is thought to mediate contraction of prostatic smooth muscle [24]. To resolve these apparently contradictory studies, RS 17053 has been proposed to distinguish a subset of the prostatic a ~A-AR subtype population, namely the a 1L-
125 AR subtype. This compound may serve as a useful pharmacological tool in establishing the existence of an additional subtype and defining its pharmacological profile.
PARALLEL COMPARISON OF ANTAGONIST PHARMACOLOGY OF THE CLONED r SUBTYPES Currently, the most valuable approach for directly comparing the al-AR subtypes is to express the three cloned receptors individually in heterologous expression systems. Many of the compounds previously described as being =lA-selective have nearly identical low aff'mities for both the alb and =ld subtypes. We investigated the binding selectivity of a variety of drugs by expressing the bovine r hamster r ~b, and rat a ld in human embryonic kidney 293 (HEK 293) cells [20]. We used the radioligand [3H]tamsulosin to label the cloned subtypes and examined a variety of compounds for their ability to compete with this ligand. Tamsulosin, in its unlabeled form, is being developed for the treatment of benign prostatic hypertrophy and demonstrates some selectivity for the alk subtype. Like its unlabeled counterpart, [3H]tamsulosin labels all three cloned receptor subtypes, although with slightly different affinity constants. Association and dissociation curves of [3H]tamsulosin binding to membranes from HEK 293 cells expressing the human a xa-AR subtype is shown in Fig. 1. This compound has an association rate constant (k0 of 1.3 x 109 1/mole-min and a dissociation rate constant (k.~) of 0.0185 min-~; with a calculated KD of 14 pM (k.x/kl) (compared to a similar value of 46 pM from saturation binding curves) at the cloned r la-AR subtype. [3H]Tamsulosin demonstrates lower affinities at the =lb and =~d binding sites, with
100. E
8o.~
D'I
"
9
Association
I;
70~
.__. ~c "Q o ~ o 9
30-
~
0
1004
Dissociation
90-
-"
70-
~"
605030-
I~
20.: 100
3'o
6'o time (min)
9'0
~o
20100
0
6'0
1:20
180
240
time (min)
Figure I. Association and dissociation curves for [3H]tamsulosin in HEK 293 cells expressing the cloned human ala receptor. Binding assays were performed at 30~ [3H]Tamsulosin binding at 40 min is used for the maximal binding in association studies. In dissociation studies, the membranes were incubated for 40 rain in the presence of [3H]tamsulosinto allow equilibrium to be reached prior to measuring the dissociation of the ligand. Points represent the mean +_S.E.M. of two experiments performed in duplicate.
126 KDS of 534 pM and 209 pM, respectively. In screening a number of =IA selective antagonists, such as 5-methylurapidil and ( + )niguldipine, for inhibition of [3H]tamsulosin binding, we found that the r selective compounds had high affinities at the cloned r and much lower affinities at both the r and r subtypes. CEC, a compound previously established as an =m-selective alkylating agent, was closely examined to determine its selectivity for inactivating the cloned r Inhibition of [3H]tamsulosin binding by pretreating membranes with CEC showed that all three cloned subtypes were partially sensitive to alkylation, although to varying degrees (Fig.2). At low concentrations of CEC (I~M), 40% of the eta sites were blocked, and 70% of each the r and aid sites were blocked. Therefore the traditional pharmacological tools previously used to distinguish the r and =IB subtypes are insufficient to distinguish the cloned r from the cloned r BMY 7378, initially described as a serotonin receptor agonist with selectivity for the 5-HT~A subtype [25 26] has recently ' ' been suggested to be selective for the r subtype as well [11]. We tested this compound for its selectivity at the expressed human e x-AR clones. In [3H]tamsulosin binding inhibition curves, BMY 7378 potently inhibited binding at the =ld subtype with a ten-fold selectivity over the =~b-AR subtype and one hundred-fold selectivity over the r subtype (see Fig. 3). Other newly developed compounds, such as SNAP 5089 [19], are also being screened for their selectivity in binding to each subtype. Characterization of these compounds and the development of other selective compounds will be valuable in future studies of the function of each receptor subtype in intracellular signalling and physiological function.
10080~
,-
9~
-V-
60-
0
Til I
40-
c
o
~
20-
tt
O~la
O~lb
~ld
Figure 2. CEC inhibition of [ 3 H ] t a m s u l o s i n bindingin HEK 293 cells expressing a~-AR subtypes. Membranes from transfected cells were incubated with 1~tM CEC for 10 min at 37~ Membranes were washed and binding of a 90% saturating concentration of [3H]tamsulosin determined. Values are expressed as a % of specific binding in membranes pretreated under identical conditions in the absence of CEC, and are the mean + S.E.M. of 5 experiments performed in duplicate.
T 11o.1 1 0
90 80 70 8o so 40 3o 2o lO 0
10-~o
T
1
0
T
'1"
~
..
:~.._ 10-9
10-8
10-7
l~.S
-s
[BMY 7378] (M)
F i g u r e 3. Inhibition of [3H]tamsulosin binding by BMY 7378 at cloned human c~-AR subtypes. Values are the mean + S.E.M. of 3 experiments each performed in duplicate. The concentration of [3H]tamsulosin was adjusted for each subclone to give 90% saturating conditions: for cx~a:0.52nM, for a~b:5.2 nM, and for CX~d:1.9 nM.
127
=I-AR SIGNALLING VIA M I ~ T I P L E SECOND MESSENGER PATHWAYS The r are known to couple to the Gq class of G proteins to activate phospholipase C (PLC). PLC activation catalyzes the cleavage of phosphatidylinositol (4,5) bisphosphate to form inositol (1,4,5) trisphosphate (IP3) and diacylglycerol (DAG). IP3 mediates the release of intracellular Ca 2§ stores and DAG potently activates protein kinase C (PKC) which leads to activation of a number of downstream effectors. r can also activate other members of the G protein superfamily. EPI stimulates GTP[a32p] labeling of Gh, a high molecular weight G protein [27]. Gh couples el-ARs purified from rat liver to PLC in a reconstituted system [28]. In addition to PLC coupling, r also couple to second messenger systems apparently through both PLC dependent and independent pathways. Multiple G proteins may contribute to the diversity seen in r second messenger signalling (Fig. 4). Evidence of =~-AR coupling to multiple signalling pathways have come from both in vivo and in situ studies. r couple to Ca2§ influx through VDCCs in the vasculature [3] and in rat medullary thyroid carcinoma cells (rMTC 6-23) [6]. r can couple to phospholipase A2 (PLA2) through two parallel, PLC-independent and -dependent pathways. In the MDCK [29] and FRTL-5 cell lines [30], and in transfected COS-1 cells [31], r mediated PLA2 stimulation can occur independent of PLC activation. For example, in transfected COS-1 cells, PLA2 activation is dependent upon Ca2§ influx and is blocked by the VDCC antagonist, nifedipine. In contrast, studies in rat pineal gland [32] and MDCK cells [33] demonstrate that PKC is able to ~,-AR mimic the activation of PLA2 seen with r tAR I stimulation. This suggests that PLC is able to I '1 G~ G, mediate the coupling to PLA2 through PKC activation and/or release of intracellular Ca 2§ I I stores. PLC VDCC Similar to PLA2 coupling, I i = phospholipase D (PLD) coupling is stimulated DAG IP~ through two parallel but independent pathways by r [34]. Evidence for this PKC [C +1, includes f'mdings from experiments in rat ....i>-::; cerebral cortical slice preparations. The effects of NE and phorbol 12-myristate 13PLA, PLD acetate, a potent activator of PKC, are additive in stimulating PLD, and inhibitors of Figure 4. Schematic diagram of ctl-AR PKC do not inhibit PLD activation by NE. intracellular signalling. Solid lines represent the Similar results were found in MDCK cells established signalling pathways while dotted lines show pathways supported by some experimental [35] supporting the idea that r couple to evidence. Abbreviations used are as follows: multiple second messenger systems through PLC - phospholipase C; VDCC - voltage divergent but interacting pathways. dependent Ca 2§ channel; DAG - diacylglycerol; In addition to these second messenger IP3 - inositol (1,4,5) trisphosphate; PKC - protein systems, a l-ARs activate other downstream kinase C; PLA2 - phospholipase A2, PLD phospholipase D. effectors through mechanisms that are not well understood, a l-ARs potentiate isoproterenol
128 (a 13-AR agonist) stimulated adenylate cyclase [2], increase N-acetyltransferase [36] activity, induce c-fos expression [37, 38], and trigger release of adenine nucleotides [39]. Defining the role of each aI-AR subtype in catecholamine signalling is complicated not only by the multiplicity of second messenger pathways coupled to r but also by the limited availability of subtype selective compounds. Molecular biological techniques and the development of subtype selective compounds are now allowing us to clarify subtype pharmacology, tissue expression, and the individual roles of each subtype in cellular signalling.
NATIVELY EXPRESSED D I F F E R E N T CA 2+ P O O L S
al-AR
SUBTYPES
COUPLE
SELECTIVELY
TO
al-ARs are known to couple to increases in intracellular Ca 2+ through both Ca 2+ influx and the release of intracellular Ca 2+ stores. Each subtype was evaluated for its ability to interact with each of these pools with the use of antagonists selective for the NIF 15o pharmacologically def'med a lA and a lB-AR subtypes. In various rat blood vessels, there is a direct correlation between expression of the e~A-AR subtype and the importance of extracellular Ca 2§ in muscle contraction [40]. In rabbit aorta, similar results were NE found for the alA-AR-extracellular Ca 2§ 0 dependence [41]. In the same study, a o 2'0 4'o o'o 8'0 16o ~o ~.o parallel correlation was observed for e~Btime (sec) ARs coupling to PLC and mobilization of intracellular Ca 2§ stores. These observations led to the hypotheses that r Figure 5. NE stimulation of Ca2§through VDCC in ARs couple to Ca 2§ influx while r rMTC 6-23 cells. Fura-2 loaded cells were exposed couple to phosphatidyl inositol (4,5) to 30~tM NE followed by 1~tM nifedipine (NIF). bisphosphate (PI) hydrolysis and release of [Ca2§ determinations were made by measuring the intracellular Ca 2§ stores to trigger blood emission wavelength at 510nm when the cells were exposed to excitation wavelengths of 340 and vessel contraction. 380nm. Data are from a single experiment, Exceptions to this generalization representative of three experiments. Fura-2 loaded have since been observed in both SK-N-MC cells were prepared as follows: Cell suspensions and rMTC 6-23 cell lines. SK-N-MC ceils, were incubated in DMEM with 5 ~tM fura-2/AM for a human neuronal cell line, express a lA30 min at 37~ Cells were washed twice and resuspended in BSS (130mM NaC1, 5mM KC1, lmM ARs which couple to PI hydrolysis and the MgCI2, 1.5mM CaCI2, 20mM HEPES, 10mM mobilization of intracellular Ca z§ stores glucose, 0.1mM glucose, 0.1% BSA). Fura-2 loaded [5]. rMTC 6-23 cells demonstrate both PI cells were then transferred to a cuvette for hydrolysis and Ca 2§ influx through VDCCs spectrofluorometric analysis. (see Figs. 5 and 6). However, Northern blot analysis, binding inhibition curves,
129
10.0
, . .
7.5
o ~9
5.0
2.5
0.0 basal NE
Figure 6. Analysisof [3H]inositolphosphate formation in rMTC 6-23 cells in response to 30~tM NE. Cells were prelabeled with myo-[3H]inositoland production of [3H]InsPswas determined as follows: cells were washed with Krebs buffer containing 10raM LiCl and incubated with or without 30~tMNE for lh. The medium was removed and the reaction stopped with methanol. [3H]InsPswere isolatedby extraction and anion exchange chromatography.
and sensitivity to CEC show that rMTC 623 cells do not express ~A-ARs [6]. In addition, these two pathways appear to be independently activated by NE. With an 80% inhibition of PI hydrolysis by U-73122, a potent PLC inhibitor, there is no decrease in NE stimulated Ca 2+ influx through VDCC. In addition, potassium chloride, which depolarizes rMTC 6-23 cells and triggers Ca 2§ influx through VDCC, does not stimulate PI hydrolysis. These exceptions have required a re-evaluation of the previous theories on how a~-ARs couple to signal transduction machinery. Recent studies suggest that the subtypes, although able to couple to multiple signalling pathways, do so with different efficiencies. To conduct such investigations, a thorough understanding of agonist pharmacology is necessary.
COMPARISON OF AGONIST PHARMACOLOGY AT CLONED
II1-A.Rs
Agonist profiles are another useful means to classify receptors and provide additional evidence for differential coupling efficiencies of the a l-AR subtypes to various second messenger systems. We evaluated a number of agonists for affinities and intrinsic efficacies at each of the cloned receptors expressed in HEK 293 clonal cell lines [42]. In radioligand binding assays, the endogenous catecholamines, NE and EPI, showed high affinities for the ald-AR subtype, intermediate affinities for the a~b-AR subtype, and low aff'mities for the ~laAR subtype with nearly a 100-fold difference between the a~a and a ld subtypes. Surprisingly, this binding selectivity was not reflected in functional assays (InsP accumulation). NE and EPI were equipotent in stimulating PI hydrolysis through each of the three cloned a l-AR subtypes. Many widely used synthetic agonists appear to be highly selective for the tt~a-AR subtype. Methoxamine and phenylephrine are partial agonists at the ~b- and ~t~d-ARsubtypes, but act as full agonists at the a la-AR subtype. Cirazoline and SKF 89784 bind to all three subtypes, but only activate InsP generation in ~tla-AR expressing cells, causing low levels of activation only at extremely high agonist concentrations in a~b- and ~ld-AR expressing cells. One possible explanation of these findings focuses on the ability of an agonist to activate a second messenger response through the different receptor subtypes. Although agonists may be able to fully activate each receptor subtype, each activated subtype may not be able to equally activate G proteins resulting in differences in the magnitude of the second messenger response. Conversely, the agonists may indeed show selectivity in their ability to
130 activate the different receptor subtypes. Other explanations for the discrepancies between agonist affinity and efficacy in eliciting a response were investigated. To test the possibility that differential receptor reserves influenced the results, we used the non-selective alkylating agent phenoxybenzamine to decrease receptor density and knock out any receptor reserve. Treatment with phenoxybenzamine caused little or no rightward shift in the dose-response curves to NE indicating very little or no receptor reserves in any of the transfected cell lines. In a system where there is no receptor reserve, measuring the maximum response to agonist is a fairly direct measurement of the relative efficacy of the agonist at a particular receptor subtype. We also investigated possible effects of the conditions under which radioligand binding measurements were performed to see if the affinity of agonists for the receptors was changed in any way. Receptors can exhibit multiple affinity states when bound by an agonist. These multiple affinity states reflect the dynamic properties of agonists. When an agonist binds to a receptor, the conformation of the receptor is changed and the affinity for the agonist can be affected. In addition, the local ionic and osmotic environment can play an important role in determining agonist-receptor interactions. However, in this system the presence of Mg 2§ (to modulate the ionic environment of the receptor) or GTP (to shift receptor equilibrium to a low affinity conformation), NE and EPI still demonstrate selectivity for the a~d-AR subtype. Neither receptor reserve nor multiple affinity states of the receptors can account for these findings. Our evidence suggests that all three cloned a~-AR subtypes do, in fact, couple to Gq and the PLC pathway, although possibly with different efficiencies.
D ~ ' F E R E N T EFFICIENCIES OF COUPLING OF u l-ARs TO PLC
PLC
More direct evidence for differential ! I I I I coupling of ~I-AR subtypes to PLC is seen in a study by Schwinn et al. [43] where the ttla-AR Gq G,1 G,4 Gls subtype appears to couple to PLC with a greater efficiency than the alb-AR, similar to our findings in HEK 293 cells. When a la- and a lb-ARs were (~la (~la ~la l~lb independently, stably expressed in HeLa cells at 700 (~,lb (~lb (~,lb fmol/mg and 1500 fmol/mg, respectively, NE (~,ld (~,ld stimulated PI hydrolysis in both cell lines. Surprisingly, the a~a transfected cell line produced an InsP response roughly twice the magnitude of the Figure 7. G protein subtypes mediating a lb cell line even though it expressed less than half InsP response to NE in Cos-7 cells the level of receptors. Further investigation into transfected with different subtypes as determined by Wu and colleagues [44]. this question will require use of inducible expression vectors to directly examine how the modulation of receptor density affects the intracellular signalling. Wu et al. also demonstrated selectivity in the coupling of ~I-AR subtypes to different G protein subtypes [44]. a l-ARs couple to PLC through the Gq class of G proteins which include Gq, Ga~, G14, and G16 (as well as the recently described G15 not used in this study).
I
I
i
I
131 Wu and colleagues cotransfected each of the three a x-AR subtypes with each of the four cloned Ga subunits into Cos-7 cells. They found that the a subunits of both Gq and Gll couple all three a l-AR subtypes to PI hydrolysis upon NE stimulation. In contrast, the a subunits of G~4 and G16 exhibit some selectivity in coupling to each of the subtypes. NE stimulates PI hydrolysis in a lb-AR transfected cells when either G14 or G16 is cotransfected into the system. In ald-AR transfected cells, neither G~4 nor G16 is able to mediate NE stimulated PI hydrolysis. NE stimulates PI hydrolysis through a la-AR when cotransfected with G~4 but not G~6 (fig. 7). These findings suggest an additional mechanism by which signal transduction pathways can be selectively activated by the a~-AR subtypes.
COMPARING RECEPTOR DENSITY TO SECOND MESSENGER RESPONSE Esbenshade et al. investigated the InsP response to NE in hamster DDT1 MF-2 smooth muscle cells [45]. These cells natively express the a~B-AR subtype and are the original source for the cloning of this subtype [7]. When transfected with an IPTG inducible vector containing the hamster a Ib-AR clone, a range of expression levels was achieved, from 860 fmol/mg protein in the absence of IPTG to 2300 fmol/mg protein in the presence of IPTG [45]. NE was able to stimulate PI hydrolysis at all levels of receptor expression with an equal potency, indicating there was no receptor reserve even at high levels of receptor expression. Instead, there was an increase in the maximal response to NE that paralleled the increase in receptor density. Modulation of a IB-AR density by the cell would provide a mechanism to directly modulate the magnitude of the ImP response. With other a l-AR subtypes there may not be a direct relationship between receptor density and the InsP response. A receptor that is very efficiently coupled to a second messenger response would rapidly develop a receptor reserve as the receptor density is increased. In this case there would be a heightened sensitivity to NE and EPI. Comparisons similar tO this a lb-AR-InsP study will help to evaluate the ability of each subtype to stimulate second messenger responses.
CONCLUSIONS Our understanding of a 1-AR subtypes has dramatically changed in recent years with the cloning of three receptor subtypes. Molecular based pharmacological studies have required a re-evaluation of the traditional pharmacology based on the natively expressed subtypes. Inconsistencies between the pharmacological profiles of the subtypes based on these two approaches raises the possibility that there may be other subtypes that have yet to be cloned. The very existence of multiple subtypes, well conserved among many species also raises the question of why so many subtypes should be expressed if they have very similar functions. Current research now focuses on distinguishing the subtypes and determining what unique role they play in receptor signalling at the cellular level and in mediating a physiological responses. Experimental evidence suggests that the answer may lie in subtype-G protein coupling. Although the receptor subtypes apparently couple to the same multiple signalling pathways,
132 subtle differences in coupling efficiencies may cause a range of physiological responses to NE and EPI depending upon which subtype, and which G protein subunits, are expressed. The use of molecular biological techniques and the development of more selective compounds is now allowing us to critically analyze these questions.
ACKNOWLEDGEMENTS Supported by Grant NS 32706 from the National Institutes of Health.
REFERENCES 1. 2. 3. 4. 5. 6. 7. 8. .
10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. 22.
Morrow AL, Creese I. Mol Pharm 1986; 29: 321-330. Minneman KP. Pharm Rev 1988; 40:87-119. Nelson MT, Standen NB, Brayden JE, Worley III JF. Nature 1988; 336: 382-385. Chiu AT, Bozarth JM, Timmermans PBMWM. J Pharm Exp Ther 1987; 240: 123-127. Esbenshade TA, Han C, Murphy TJ, Minneman KP. Mol Pharm 1993; 44: 76-86. Esbenshade TA, Theroux TL, Minneman KP. Mol Pharm 1994; 45: 591-598. Cotecchia S, Schwinn DA, Randall RR, Letkowitz ILl, Caron MG, Kobilka BK. Proc Natl Acad Sci USA 1988; 85: 7159-7163. Schwinn DA, Lomasney JW, Lorenz W, Szldut PJ, Fremeau Jr. RT, Yang-Feng TL, Caron MG, Lefkowitz RJ, Cotecchia S. J Biol Chem 1990; 265:8183-8189. Lomasney JW, Cotecchia S, Lorenz W, Leung WY, Schwinn DA, Yang-Feng TL, Brownstein M, Lefkowitz RJ, Caron MG. J Biol Chem 1991; 266" 6365-6369. Perez DM, Piascik MT, Graham RM. Mol Pharm 1991; 40: 876-883. Goetz AS, King HK, Ward SDC, True TA, Rimele TJ, Saussy DL. Eur J Pharm 1995; 272: R5-R6. Muramatsu I, OhmuraT, Kigoshi S, Hashimoto S, Oshita M. Br J Pharm 1990; 99:197201. BoerR, GrasseggerA, SchudtCH, GlossmanH. EurJPharm 1989; 172: 131-145. Minneman KP, Atkinson B. Mol Pharm 1991; 40: 523-530. Minneman KP, Han C, Abel PW. Mol Pharm 1988; 33: 509-514. Schwinn DA, Lomasney JW. Eur J Pharm 1992; 227: 433-436. Gross G, Hanft G, Rugevics C. Eur J Pharm 1988; 151: 333-335. Ford APDW, Arredondo NF, Blue DR, Bonhaus DW, Kava MS, Williams TJ, Vimont RL, Zhu QM, Pfister JR, Clarke DE. BrJ Pharm 1995; ll4(supp): 24P. Wetzel JM, Miao SW, Forray C, Borden LA, Branchek TA, Gluchowski C. J Med Chem 1995; 38: 1579-1581. Hart C, Hollinger S, Theroux TL, Esbenshade TA, Minneman KP. Pharm Comm 1995; 5:117-126. Hirasawa A, Horie K, Tanaka T, Takagaki K, Murai M, Yano J, Tsujimoto G. Biochem Biomed Res Comm 1993; 195: 902-909. Laz TM, Forray C, Smith KE, Bard JA, Vaysse JJ, Branchek TA, Weinshank RL. Mol Pharm 1994; 46: 414-422.
133 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45.
Flavahan NA, Vanhoutte PM. Trends Pharm Sci 1986; 7: 347-349. Forray C, Bard JA, Wetzel JM, Chiu G, Shapiro E, Tang R, Lepor H, Hartig PR, Weinshank RL, Branchek TA, Gluchowski C. Mol Pharm 1994; 45: 703-708. Stubbs CM, Trezise D, Connor HE, Feniuk W. J Auton Pharm 1991; 11: 237-245. Higgins GA, Elliot PJ. Eur J Pharm 1991; 193: 351-356. Baek KJ, Das T, Gray C, Antar S, Murugesan G, Im MJ. J Biol Chem 1993; 268: 2739027397. Das T, Baek KJ, Gray G, Im MJ. J Biol Chem 1993; 268: 27398-27405. Slivka SR, Insel PA. J Biol Chem 1987; 262: 4200-4207. Burch RM, Luini A, Axelrod J. Proc Natl Acad Sci USA 1986; 83: 7201-7205. Perez DM, DeYoung MB, Graham RM. Mol Pharm 1993; 44: 784-795. Ho AK, Klein DC. J Biol Chem 1987; 262:11764-11770. Weiss BA, Insel PA. J Biol Chem 1991; 266: 2126-2133. Llahi S, Fain JN. J Biol Chem 1992; 267: 3679-3685. Slivka SR, Meier KE, Insel PA. J Biol Chem 1988; 263: 12242-12246. Klein DC, Sugden D, Weller JL. Proc Natl Acad Sci USA 1983; 80: 599-603. Thonberg HS, Zhang SJ, Tvrdik P, Jacobsson A, Nedergaard J. J Biol Chem 1994; 269: 33179-33186. Okazaki MZ, HuZW, FujinagaM, HoffmanBB. J Clinlnv 1994; 94: 210-218. Shinozuka KM, Hashimoto M, Masumura S, Bjur RA, Westfall DP, Hattori K. Br J Pharm 1994; 113: 1203-1208. Hart C, Li J, Minneman K_P. Eur J Pharm 1990; 190: 97-104. Suzuki E, Tsujimoto G, Tamura K, Hashimoto K. Mol Pharm 1990; 38: 725-736. Minneman KP, Theroux TL, Hollinger S, Han C, Esbenshade TA. Mol Pharm 1994; 46: 929-936. Schwinn DA, Page SO, Middleton JP, Lorenz W, Liggett SB, Yamamoto K, Lapetina EG, Caron MG, Lefkowitz ILl, Cottecchia S. Mol Pharm 1991; 40:619-626. Wu D, Katz A, Lee CH, Simon MI. J Biol Chem 1992; 267: 25798-25802. Esbenshade TA, Wang X, Williams NG, Minneman KP. Eur J Pharrn 1995; 289: 305-310.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardin/~, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
135
tXl-ADRENOCEPTORS: SUBTYPE- AND ORGAN-SELECTIVITY OF DIFFERENT AGENTS A. Leonardi a, 1LTesta a, G. Motta a, P.G. De Benedettib, P. Hieble c, D. Cfiardin~d a
b
R & D Division R,ecordati S.p.A., Via M.Civitali 1, 20148 Milano, Italy Dip. Chimica, Un. Modena, Via Campi, 183 - 41100 Modena, Italy Div. PharmacoL Sciences, SmithKline Beecham Pharmaceuticals, King of Prussia, Pennsylvania, 19406, U.S.A. Dip. Scienze Chimiche, Un. Ca,merino, Via S. Agostino, 1 - 62032 Camerino, Italy.
INTRODUCTION The subclassitication of the a-adrenergic receptor (a-AR) into the al" and a2-subtypes [ 1, 2] made possible the development of effective antihypertensive drugs that either selectively block the ctl-AR or activate the ct2-subtype. About ten years after this subclassification, the first suggestions for fimher subdivision were made based on functional and radioligand binding data [3-5]. Currently, the presence of at least three distinct al-ARs has been established through molecular biology, radioligand binding and fimctional studies [6]. The functional evidence of further distinct subtype(s) showing low affinity for prazosin is still awaiting confirmation from molecular biology [3, 7, 8]. The presence of these different subtypes of the ctl-AR in blood vessels and other smooth muscles offers the oppommity for selective drug action. Selective antagonists are perhaps the most useful tools for receptor characterization and classification. The aim of this short review is to update the knowledge on tx~-AR subtypes selectivity of known and novel a~-AR antagonists, based on radioreceptor binding study results. By this technique, the affinity of most of the antagonists for several other G-protein-coupled receptors was also investigated. Functional correlates to the observed selectivity patterns were investigated by studying the effects of selected antagonists on blood vessels and lower urinary tract organs on in vitro and in vivo models. Antagonists endowed with high selectivity for the lower urinary tract are presented.
r
SUBTYPES
The initial subdix~'on of the a~-AR into subtypes was based primarily on the selectivity of WB-4101 for the c~subtype [4] and the selective alkylation of the era-subtype shown by chloroethylclonidine (CEC) [5]. Other antagonists selective for the ct~-AR have been identified, such as 5-methylurapidil [9] and (S)-(+)-niguldipine [10].
136 Molecular biology techniques allowed to identify cDNAs encoding three afARs, namely the rat ala/d, the hamster alb and the bovine a~c [ 11-15]. Originally, the pharmacological characteristics of the cloned subtypes appeared to be inconsistent with the pharmacological properties of the native subtypes, the existence of four subtypes being also suggested [15]. It is now clear that the three recombinant a~-ARs which have been cloned to date correlate closely with the three a f A R subtypes, that have been identified in native tissues and which mediate their functional responses [16-18]. These receptors are now designated as a m (ala), alB (alb) and aiD (a~d), with lower case subscripts being used to designate the recombinant receptors, and upper case subscripts to denote the native receptors [6]. The recombinant receptor previously designated as the a~c-AR has now been shown to correspond to the native atA-AR [16, 19-21], and as such, the terms a~c- or alc-ARs are no longer used [22]. Furthermore, the atD-AR has now been characterized in detail, and antagonists capable of discriminating between this receptor and the a~A-AR are now available, confirming that these two receptors represent distinct a f A R subtypes [23, 24]. As such, the previously used designation of am/o-AR is now inappropriate. In addition to these am, alB, and atD-AR subtypes, which share a high affinity for prazosin, the existence of additional al-ARs has been proposed. These are called a~L-ARs and are characterized by a low functional affinity for prazosin, intermediate affinity for WB 4101 and 5-methylurapidil and resistance to CEC alkylation [3, 7, 8]. No related recombinant ax-AR has yet been identified and the radioreceptor binding characterization of these sites is still difficult, perhaps due to the lack of a suitable radioligand. Nevertheless, there is clear evidence from functional studies for the existence of these ax-ARs in blood vessels and other smooth muscles [8], and these prazosin-low affinity al-ARs may represent an additional therapeutic target for selective drug action. CLASSIFICATION OF al-ADRENOCEPTORS ANTAGONISTS The considered a f A R antagonists were classified according to their affinity for the different receptors and cloned subtypes, determined by radioreceptor binding studies. Tritiated prazosin was utilized to assess the affinity for the native a f A R (rat cerebral cortex [4]), the UtA (rat hippocan~us pretreated with CEC [25]) and the a~B (rat liver [25]) adrenergic subtypes, as well as for the recombinant bovine Ula (formerly a~c [12]), hamster alb [11], and rat a~d (formerly a~a/d [13]) transiently expressed in COS-7 cells. Transfected COS-7 cells were provided by Dr. S. Cotecchia (University of Lausanne, Switzerland). The same ligand was utilized to investigate the affinity of a few compounds on the human recombinant afsubtypes expressed in CHO cells [20]. Where not specifically stated, the affinity for the cloned subtypes in Tables 1 and 2 was determined in the animal clones. The rat cerebral cortex data are reported here in order to maintain a link with the data in the older literature and the native subtypes data for comparison with the results obtained on the cloned subtypes. The other G-protein coupled receptors investigated by us were the ae-AR ([3H]rauwolscine, rat cerebral cortex [26]), the 5-HT~-serotonergic ([3H]-8-OHDPAT, rat hippocan~us [27]),
137 the 5-HTzA-serotonergic ([3H]ketanserine, rat cerebral cortex [28]), the D2-dopaminergic ([3H]spiperone, rat striatum [29]). For the 1,4-dihydropyridines, also the binding to the 1,4-DHP binding site was investigated ([3H]isradipine, rat brain [30]). For most of the data, at least 2 determinations (in triplicate) were performed. Regression analysis of the affinity obtained for the native atA vs the cloned C~a and for the native a~B VS the cloned a~b-subtypes gave the following equations (n=45): pKi~a = 0.91 pKitA + 0.34 (R e = 0.89) pKi~b = 0.90 pKi~B + 0.94 (R2 = 0.93) Only for 9 compounds a difference in pKi higher than 0.5 was experienced, this difference being in the range 0.5-0.7, thus confirming the good agreement between the two data-sets. Representative examples from the known classes of cxl-AR antagonists, as reported in several reviews [31-36], have been evaluated by radioreceptor binding screening. In addition, compounds from more recently introduced classes were also included. The results are shown in Table 1. The structure of considered compounds, grouped by class, is shown in Figure 1.
Poorly subtype-selective al-antagonists (Table 1) Yohimbanes : corynanthine The most constrained and less potent among the known a~-AR antagonists confirmed its low potency and showed little subtype selectivity. Although corynanthine is reported to be a selective al-adrenoceptor antagonist, affinities for a~- and oq-ARs were nearly equal.
Ergot alkaloids: dihydroergocryptine Another very rigid molecule which unexpectedly showed the same high affinity for the a~b, and the other receptors tested.
ala ,
Ouinazolines This is one of the most investigated classes in this field, due to the high selectivity for the al-AR vs the ~ - and the other receptors, with prazosin being the prototype drug. Subtype selectivity is very low for the "classical" quinazolines and in some cases also al/a 2 selectivity proved low. The main interaction with the receptor has been proposed [37] and later confirmed [38] to occur through a charge-reinforced H-bond between an anionic site in the receptor (aspartate ion on the 3rd transmembrane a-helix) and the protonated quinazoline N 1. The presence of a substituted amino group at position 2 contn'butes to protonability and stabilization of the charge on the N 1 whereas N 3 is not mandatory for activity [39], as supported in Table 1 by the data for abanoquil, a very potent but non subtype-selective c~l-blocker, in comparison to its quinazoline analogue I [37]. The modelling and QSAR for a series of quinazoline derivatives has recently been published [40]. Structural modification of the amino group at position 2 afforded novel derivatives endowed with some selectivity for the a~B-AR subtype, discussed below.
N-A rylpiperazines This is another class of compounds extensively investigated mainly for their activity at a1-[34 ]
138
and 5-HT~-receptors [41]. The older representatives, urapidil and A R C 239, listed in Table 1, s h o w no relevant subtype selectivity and no separation o f the affinity for the t w o receptors. F u r t h e r w o r k on this class led to m o r e selective c o m p o u n d s , discussed below.
Table 1. Binding affinity o f
RECEPTOR
ct~-AR antagonists (Ki, nM).
~1
0[~IA 0{~IB 0['la 0~lb
0{~ld 0['2
5-HTtA 5-HT2A D2
Ca++
Poorly selective Corynanthine Dihydroergocriptine Prazosin Terazosin
367 3.7 0.7 3.4 0.1 0.2
424 5.8 0.9 5.8 0.1 0.2
671 9.9 0.4 3.5 0.7 0.1
128 4.8 0.7 26 0.2 0.1
1.0a
2.0a
0.4 a
855
272
429
290
1302
Phentolamine SNAP 5036
31
16
89
3.2 4.4 c
Tamsulosin WB 4101 Benoxathian (+)-SPAL (-)-SPAL Indoramm RS 17053
3 8.7 5.3 121 10 29
0.2 4 1.3 26 2.0 57 44 619 1.2 60 28 43 0.7 e,f
Compd 2
10 28 4.0 10 2.8 13 4.3 2.1 28
Compd 1 Abanoquil ARC 239 Urapidil
1070 10 0.5 26 0.2 0.1
287 33 1.4 35 0.2 0.1
2167 2.4 2357 > 2325 2489
868 13 > > 7800 717
1818 3.5 > > 2474 2207
1658
437 1.7 147 174 11 43 10a,b 2343
213
8445
3181
89 72c
67 155 c
40 126c,d
1263
432
>
0.2 0.6 0.7 4.1 0.4 5.0 0.3 f
6.7 57 55 159 21 24 14f
1.8 6.3 21 140 6.8 274 17f
237 15 109 1504 2035 1634
4.4 7.3 5.3 890 510 74
> 716 182 890 577 243
220 144 281 822 97 315
0.6 2 3.5 0.3 1.3 0.4 0.4 0.2 0.4 1.8c
16 775 12 52 5.1 118 44 18 25 85c
4.1 175 39 24 43 14 5.3 7.6 82 191 c
23 432 12 92 193 151 19 110 1611 372 c,g
125 1.2 93 11 162 22 7.4 54 >
158 > 364 1257 1882 915 197 952 712
69 1051 369 100 153 101 37 100 295
0.4 h
230h
427 h
562 h'g
0.5 1.3h
76 269 h
468 9415 1047 h 1318h'g
0.6 h
324h
CXla-AR selective
5-Me-urapidil
Compd 3 Compd 4 Compd 5 Compd 6 Compd 7 Compd 8 (S)(+)-Niguldipine
1.9 4.7 1.4 1.3 1.7 0.9 0.5 0.3 6.4
15 220 3.2 11 13 62 20 10 380
Compd 9 SNAP 5089
28
2.6
229
Compd 10 Compd 11
0.8 h 2754 h
31 h >
2385
899
(+)-Cyclazosm AH l l l l 0 A Spiperone Risperidone
652 794 h
204 h'g
>h
4467 h 1072d,h
>h
631 h
CZlb-AR selective Compd 12 Compd 13
0.3 9.1 c
1.4 173 5.8
2.2 503 25
1.1 23 0.2
7.5 424 33 161 2511 i
0.5 101 0.7 40 76 i
10 622 27 382 2574 i
1561 4804 613
4498 > >
1364 > > >
8005 3557 3533 >
7.0 9.3
38 4.7
1.6 2.2
13 2.8
7.1 7.7
22 25
136 14
25 371
2.3 0.5
0.6 6.6
139
Table 1-Follows RECEPTOR
r
~1
I~IA 0~IB (~la O~lb O~ld 0['2 5"HTxA 5-HT2A D2 Ca++
selective
BMY 7378 Compd 14 Compd 15 SKF 104856 SKF 106686 (-)-Discretamine
282 422 314 0.7 0.8
273 1.2
495 426 43 1038 1.2 0.2 36a 29a 58a 15a 6171 3631
5.7 474 0.1 1.6a 1.2a 251
657 > 3.4a'b 15a'b
0.4 287 >
493 >
53 > 29
Notes to Table 1. > means > 10000 nM. a) human clones; b) Ot2b-Subtype, see ref. [24] for method; c) human clones, see ref. [43]; d) c~b-subtype; e) rat submaxillary gland; f) see ref [46]; g) a2csubtype; h) human clones, see ref. [52]; i) see ref. [57]; 1) functional data, see ref. [64].
a~a-Adrenoceptor selective antagonists (Table 1) lmidazolines : phentolamine Based on our results, this well known compound can be considered endowed with a slight selectivity for the C~a over the C~b- and eqd-AR, in agreement with the affinity for the cloned oq-subtypes recently reported for it [42]. Phenylalkylamines Although BE 2254 is devoid of subtype and ~ / % selectivity, slight modification of its structure afforded SNAP 5036 [43], showing moderate selectivity for the a~a-AR. A relevant decrease in affinity for the %-AR must also be noticed. Of the other phenylalkylamines, tamsulosin was confirmed to be very potent at the C~a-AR and more selective over the C~b- than the Oqd-Subtype. A significant affinity for the serotonin 5-HT~A receptor is also present. Benzodioxanes In our hands, WB 4101 confirmed its high potency at the Oqa-AR with a moderate selectivity respect to the C~ad-and a pharmacolo~cally relevant difference with regard to the Oqb-Subtype. Significant affinity for the ~ - A R and the 5-HT~A receptor was also found. Its thio analog, benoxathian, gave approximately the same profile, but with a lower affinity for the ~-AR. Introduction of a 4-tolyl group at position 3 of the benzodioxane nucleus of WB 4101 afforded a very interesting compound whose trans isomer showed a unique selectivity for c~over ~ - A R [44]. This compound was resolved [45] and the two enantiomers, (+)- and (-)-SPAL proved stereoselective at the c~-subtypes and D2-dopaminergic receptors, maintaining a remarkable C~a-subtype selectivity and practically losing the affinity for the ~ - A R and the serotonergic 5-HT~A-receptor present in WB 4101.
140 Figure 1 - Structure o f the investigated CZl-amagonists, grouped by class (the letter close to the structure shows the c~I-AR subtype selectivity, if any) Yohimbanes:
H
~
o~,~o~
Ergot alkaloids: H,c CH, OH
~N
O ":'H
I~-C~~ \"o
,--/ "H
H..r
Dibenzoquinolizines:
OH
HO
CH~
OCH,
Dihydroergocrypfine
Corynanthine Quinazolines:
R= --N
R=
~N
~
N X__J
O
Terazosin
~>.~
H3CO1 v ~
,.~o,es:
Abanoquil
,~o~~~o~ H,NO~S
H
H,C
SNAP5036(a) Benzodioxanes:
"N"
(+)-Cyclazosin (b)
[~~ OCH3
H
OC o Compd 12~o)
H3CO~
Compd 13 (b)
~~~.~~
Prazosin
Compd I
OCHs
Phenylalk~lamines:
o
(-)-Discretamine (d)
~- ~ ~ - ~ o ~
"~OCH3
R=/N ~
o~
N k..._./
R=
_~
OCH,
OvClA
Tamsulosin(a)
lmidazolines: N/"7
~
-NxH
, c ~Phentolamine ~ - ~ (a)~
OCH3
~H Indoramin (a)
X= O R= H WB 4101 (a) X= S R= H Benoxathian (a) X= O R= 4-tolyl (trans) SPAL (a)
;~ ~ o _ ~
H,C C%
"H
RS 17053 (a)
o~.~~ c,,
H.CO
Compd 2 (a)
141 Figure 1- Follows
H3C
N-Arylpiperazines:
x
ARC 239
R o
~
o H~C CH3 ci~ n I J O~~N~
Urapidil
OCH3 Compd 3 Comlxl 4 (a) OCH3
o
N~
N
5-Me-Urapidil (~c .~N~~[~ C~ o BMY 7378 (a)
OCH3
o
~N---
Ph
Compd 6 (a)
t-Bu
o ~
Various structures:
F
~
2-OCH3
Compd $ (a) Cy-Hex
2-OCH3;5-C1
1,4-D~ydropyridines:
H
~N ~.O
~'~N'~ H
%c- -~--c% n
CI
(S)(+)-Niguldipine (a) Compd 9 (a) SNAP 5089 (a)
NO 2
H
OH NH
Spiperone %c ...2-/~
Risperidone
R 3-NO2 3-NO 2 4-NO 2
",,..,,,~Ph Ph X O NH NH
o@o
O
H
C
2-OCH3
Compd 7 (a) Cy-Hex
OCH3 I
O
o-N I 'k_/~
2-OCH3;5-C1
OCH3
~N--
Compd 15
Ph Ph
Compd 5 (a)
o o Compd 14
R1 H 2-OCH3
R
%c
O_
o
Eompd 10 (a) Compd 11 (a)
Ph
R2 R6 R4 c2r% Crt2OCH2Crt2~ 2 t,h CH3 CH 3 COOCH3
s~N-c%
Thienobenzazepines: Cl
R
R
SKF 104856 (d) CH2=CH2 SKF 106686 (d) C2H5
142 Indoles The first example in this class, indoramin~ exhibits a substantial selectivity for the {~la-Al{ over the a~d-subtype, lndoramin has also significant affinity for the serotonin 5-HT~A and histamine H~ receptors. Another indole bearing a basic alkyl chain at position 3 is KS 17053 [46] which proved more than 40-fold more potent at a~a- than at a~b- and a~d-AR. A different substitution pattern is present in compound 2 [47], showing a moderate a~a- over a~b-AR selectivity but an interesting lower affinity for the serotonergic 5-HTtA-receptor, in spite of the presence of the N-(2-methoxyphenyl)piperazinyl group in the ether chain. N-A rylp ipe razi nes 5-Methylurapidil is the best known of this class and was confirmed to be higly selective both for the aL~- and a~a-AR. Very high affinity for the 5-HTtA-serotonergic receptor was also confirmed. The effect of different substituents on the phenyl ring of the arylpiperazine moiety was investigated in a small series of 8-flavonecarbamylalkyl derivatives, compounds 3-5 [48, 20] where the nature and length of the spacer had previously been optimized. With regard to subtype selectivity, the 2-methoxy group gave the best results. The introduced substituents gave lower affinity for the ~ - A R with regard to the unsubstituted derivative 3. Interestingly, the introduction of a chlorine atom para to the methoxy group gave a satisthctory selectivity for the ala-AR with regard to the 5-HTm-serotonergic (compound 5, more than 100-fold difference). This trend is confirmed also by the results obtained with compounds 7 and 8, differing from the above series for the presence a cyclohexyl instead of a phenyl group at position 2 of the chromone ring. The best Ctla/alb selectivity in this series was obtained with the introduction of a tert-butyl group at position 2 of the chromone ring (compound 6). Based on our results and literature findings [49] it can be stated that careful modulation, series by series, of the substitution pattern of the aryl and alkyl substituents on the two piperazine nitrogen atoms can result in selective ax-antagonists. 1, 4-Dihydropyridines (S)-(+)-Niguldipine was the first 1,4-dihydropyridine to combine tx~-AR and Ca~-chatmel blockade. Studies on its action in native tissues showed selectivity for the auc vs the CqB-A1L Due to its very lipophilic nature both its absolute potency and subtype selectivity may vary significantly between laboratories [50, 17, 20, 22, 51]. Formal transformation of the aminoalkyl ester of niguldipine into the corresponding N-aminoalkyl carboxamide afforded compound 9 [52], endowed with striking selectivity for the human axa-AR with regard to the a~b- and a~d-subtype , but still active on the Ca~-channel. ala-Selectivity was maintained and affinity for the Ca~-channel was greatly reduced in compound SNAP 5089 [52, 53], which is the 4-nitrophenyl isomer of 9. Extensive structural modification was performed on the 4-(4-nitrophenyl)- 1,4-dihydropyridine structure [52] which led to compounds which can be considered specific for the human axa-AR, based on the reported results (see for example compounds 10-11 in Table 1).
143
a~b-Adrenoceptor selective antagonists (Table 1) In contrast with the ala- and axd-ARs , highly selective axb-AR antagonists have not yet been identified. A very promising agent, (+)-cyclazosin, is introduced here, whereas the selectivity of compounds initially reported to be a~B-selective was not confirmed in our study.
Novel quinazolines Screening a series of novel 4-amino-6,7-dimethoxy-2-substituted quinazolines [54], compounds 12 and 13 were identified, showing a moderate albeit inconsistent selectivity. The acylpiperazine derivative 12 showed a moderate selectivity for the recombinant Crib-subtype , whereas it proved non-selective when native ARs where used. On the contrary, the tetrahydropapaverine derivative 13 exhibited moderate selectivity on native but not on recombinant subtypes, in analogy with spiperone (see below). (+)-Cyclazosin was obtained by one of the authors (D.G.) by resolution of the racemic compound [55], which showed moderate selectivity for both rat liver CqB- and ham~er c~xb-AR [56]. This (+)-enantiomer can be considered the first al~-AR selective antagonist, being on the order of 100-fold selective vs the native c ~ and about 40-fold vs the recombinant a~a and Cqd-AK Research to assess its absolute stereochemistry is in progress. Various structures The novel amidine AH l l l l 0 A has been recently reported to show more than 30-fold selectivity for the hamster Ct~b-AR, compared to the ~a- or Ctld-subtypes [57]. In our hands, this compound showed a much lower selectivity on the same clones. Analogous results were obtained for spiperone, which proved about 20-fold selective on the native subtypes and non-selective on the clones, in agreement with the results from other laboratories [58, 22]. Also another dopamine receptor antagonist, risperidone, was reported to show some selectivity for the eqB-AR [59], which was not confirmed in our studies. These problems of reproducibility might be ascribed to the marginal selectivity of the currently investigated compounds. a~d-Adrenoceptor selective antagonists (Table 1) N-A rylpiperazines BMY 7378 is by far the most selective C~ld-AR antagonist reported [23]. Its high selectivity was confirmed also in our studies, together with its relevant affinity for the serotonergic 5-HT~-receptor. On replacing the spirocyclic imidoyl group of BMY 7378 with a phthalimidoyl group (compound 14 [60]) both selectivity and affinity went lost, suggesting a specific interaction of the spirocyclic group with the AK Compound 15 [49] shows a very high affinity for the C~ld-AR but also a very low subtype selectivity. Structural modification of 15 afforded compounds endowed with very high selectivity for both ~ld- and ~a" versus ~lb-AR [61]. Hetero-fused 3-benzazepines This is a novel class of compounds showing moderate selectivity for the human recombinant C~ld-Clone. The thienobenzazepines SKF 104856 and 106686 show a high potency and a
144 moderate selectivity, more marked vs the ala" than the O~lb-and ~b-AR [62]. Di be nzoqumolizines : (-) -discre tamme This alkaloid was extracted from the plant Fissisti~a glaucescens [63] and its select'wiry for the a~D-AR was assessed through functional studies where antagonism to NA-induced contraction of rat aorta (a~D), spleen (alB) and vas deferens (au0 was determined [64]. The results available so far strongly suggest further evaluation of this (class of) compound(s).
LIGAND-RECEPTOR INTERACTION MODEI.J.JNG AND QSARs The heuristic-direct QSAR approach introduced by one of the authors [65] was applied to the study of the interaction of selected ligands with the three ~I-AR subtypes. This approach, applicable when the atomic resolved structure of the target is unknown but predictable by means of the available experimental information, is able to handle the heterogeneous experimental information on the ligands and their targets, and to synthesize and translate them into QSAK models. The first step of this procedure consisted of the computer-aided three dimensional (3-D) model building of the seven transmembrane (7-TM) helix bundles of the three aI-AR subtypes. The seven helices, selected from the human primary sequences [66, 51, 67], were initially packed according to the seven-helix wheel projection models proposed by J. Baldwin [68]. The packing preferences of the 7-TMs were then investigated by comparative Molecular Dynamics (MD) simulations (QUANTA-CHARMm) [69]; the structures averaged over the last 100 picosecond time period of the MD simulation and minimized are reported in Figure 2. The 7-TM models of the three al-ARs show different dynamic behaviours and different topographies of the binding sites, which are however mainly constituted by conserved residues. The structure/dynamic analysis of the three models shows that the r site is more flexible and structurally different with respect to that of the other two subtypes. The second step in our approach involved docking simulations with selected ligands maximizing the complementarity between ligand and receptor. The following ligands in their protonated forms were considered: (-)-noradrenaline (Kiocaa--4530.2; Kia~b = 9264.7; Kia~d = 770.4), WAY 100635 (Kia~a = 143.4; Kialb = 183.7; KiOt,d = 63.1), prazosin, spiperone, urapidil, compound 12, BMY 7378, phentolamine, tam_~osin, WB 4101, compound 4, (S)-(+)-niguldipine and 5-methylurapidil. These ligands were docked into the three a~-AR subtype binding sites. For each complex several rninimirations were performed in order to optimize several fundamental interactions i.e. the charge reinforced H-bonds between the protonated nitrogen atom of the ligands and the carboxylate of the Asp307 present in the TM-3 (the numbering is arbitrary: the first digit indicates the helix and the next two digits indicate the position of the residue in the helix). In the final step, a detailed extensive correlation analysis between the computed binding energies (BE) and the experimental binding aff-mities determined on the cloned bovine a~a-, hamster a~b" and rat ~qd-AR subtypes were carried out in order to evaluate the consistency of the QSAR models proposed [70].The binding energies (BE) were computed according to the following formula: BE = IE (receptor-ligand) + E (receptor) + E (ligand), where IE (receptor-ligand) is the total interaction energy of the ligand-receptor complex, and E (receptor) and E (ligand) are the distortion energies of the receptor and of the ligand,
145
Figure 2. Minimi7ed average structures of the three All, subtypes. The helix bundles are seen from the intracellular side in a direction parallel to the main helix axes.
/
.t
X,
Figure 3. Details of the interaction between the antagonist 5-methylurapidil and the conserved amino acids of the three AK subtypes, postulated to make the major contribution to antagonist-receptor interaction. The green color labels for favourable interactions while the red color labels for unfavourable interactions.
146 respectively, calculated as differences between the energies of the bound and of the flee optimized molecular forms. Very satisfactory correlations were obtained: pKodla = 5.109 (+ 0.346) - 0.094 (+ 0.010) BEaaa, n=13, R=0.94 pKicqb = 3.692 (+ 0.381) - 0.136 (+ 0.014) BEa~b, n=13, R=0.95 pKkxld = 4.645 (+ 0.390) - 0.084 (+ 0.010) BECZxd, n=13, R=0.91 Very similar correlations have also been obtained by plotting the binding affinities of nine ligands ((-)-noradrenaline, prazosin, spiperone, urapidil, phentolamine, tamsaflosin, WB-4101, compound 4 and 5-methylurapidil) measured on human cloned al subtypes versus the computed BE (pKia~a vs BEaxa: n=9, R=0.96; pKi(Xlb vs BEalb: n=9, R=0.95; pKiald vs BEard: n=9, R=0.78). By analyzing the ligand-receptor complexes we found that compounds endowed with high affinity and no selectivity, like prazosin, interact mainly with the same conserved residues of the three subtype binding sites without inducing relevant distortions. Potent and selective ala-AR antagonists like 5-methylurapidil interacts without inducing significant distortions only with the (Xta-AR binding site, as indicated by the green color which labels the binding site residues of the (Zxa-AR subtype in Figure 3. In contrast, 5-methylurapidil induces significant unfavourable distortions into the a~b" and the a~d-AR binding sites, as indicated by the red color. On these bases, the molecular determinants for affinity and selectivity can be postulated. VITRO AND I N VIVO SELECTIVITY
~-Antagonistic activity on different organs was investigated for selected compounds, with the final aim of verifying how the subtype selectivity observed in the radioreceptor binding studies translates into functional results. Rat aorta, rabbit aorta and urethra, and human prostate tissues were utilized. Prototypic tissues for the functional investigation of the different al-AR subtypes are under continuous investigation, and the correspondence of rat aorta with the Cqd-, rat spleen with the alb" and rat kidney with the (Z~a-subtype seems currently the most accepted [19,71,64,72]. For other tissues, in particular those of the lower urinary tract, the situation is, in our opinion, less clear. From one side there is convincing evidence of the functional relevance for human prostate of the atA-AR [17,18,73], in addition to analysis ofmRNA expression [74,75] and to radioreceptor binding studies [20,76]. On the other hand, resistance to CEC and low affinity for prazosin suggest that the functional affinity to this tissue could be due to the (ZlL-AR [8,77,46]. Listed in Table 2 are the functional and receptor binding results of some selected antagonists, in an attempt to clarify this issue. A good correlation is present between affinity for the aid-subtype and potency in inh~iting the contraction of rat aorta (R2 = 0.696), confirming our previous findings [72]. With regard to human prostate, the very poor functional results obtained by other authors with KS 17053 and abanoquil, despite their high potency in inh~iting NA-induced contractions in rat isolated kidney and vas deferens, respectively, and their high affinity for the cloned and native ala-ARs [46,78], destroy the relatively good correlation present with the other antagonists (Figure 4A), and suggest the relevance of other receptors, possibly the alL-AK
147 In our screening procedure in a program aiming to identify selective al-AR antagonists for use in BPH, we used the rabbit urethra preparation for screening in vitro functional activity of the compounds. Table 2
~ty
of S d ~ e d I~I-ARanta~oni~Lsfor the recombinant animal ~x-AR subtypes (pKi= -loga0Ki, nM) and functional affinity for the %-ARs of vascular and lower urinary tract tissues (pKb = -lOgl0 Kb, nM, for inhibition of NA-mduced contraction). R.A.= rat aorta; RB.A.= rabbit aorta; RB.U. = rabbit urethra; H.P.=human prostate. The in vivo effects after i.v. administration in the dog model are also listed. Data represent the doses ( ~ t ~ ) active in inhibiting by 50% the urethral contractions reduced by NA (UP), the doses active in lowering diastolic blood pressure (DBP) by 25%, and the ratio between them. The ratios between the in vitro potency (Kb) on rabbit aorta and urethra are also reported (1/2).
COMPD
O~la
Otlb
r
S.A.
pig
pKi
pig
pKb
RB. A. pKb 1
RB. U.
pKb 2
H.P. UP
DBP DBP/
1/2
UP
pKb ED~ ED~
Prazosm
9.14
9.34
8.86
9.90
9.00
8.11
8.25
3.6
6.6
1.83
0.13
Terazosm
7.58
7.52
7.46
8.58
7.92
6.95
7.38
21
61
2.9
0.11
Tamsulosm
9.82
8.17
8.75
9.35
9.54
9.31
9.20
1.1
11
10
0.59
5-Methylur.
8.69
6.10
6.80
7.65
7.52
8.04
7.93
1.4
10.8
7.7
3.31
SNAP 5089 9.33
7.12
6.33
6.65
3000 >10
>3.40
a) N-[3-[4-(2-hydroxyphenyl)- 1-piperazinyl]propyl]-3-methyl-4-oxo-2-phenyl-4H1-benzopyran-8-carboxamide; b) extrapolated from ref. [78]; c) from ref. [46]. This tissue has initially been characterized as am-subtype dependent [25]. However the results obtained by us with compound 12 and SNAP 5089, both showing much lower functional affinity than expected based on their binding affinity for the ala-AR (see Figure 4B), and the low pKb of prazosin, suggest that also in this case the presence of ctxL-AR must be accounted for. In our view, this tissue could be considered as (XlL-AR dependent and SNAP 5089 and compound 12 may be very poorly active on this subtype. We investigated organ selectivity of our compounds by utili~ng, in addition to rabbit urethra, rabbit aorta which is reported to be dependent on alL" and (XlB-AR population [8]. The classification of this tissue seems difficult with the data presented here. In any case, we can observe that among the tested compounds three groups can be considered, based on the results on rabbit urethra and aorta. The quinazoline derivatives prazosin, terazosin and compound 12 show higher affinity for the vascular tissue than for the urethra; tamsulosin and
148 10,5 -
10.5 -
~'
9.5
A
-
D,.
s 9162149 ~l
o.
== 8 . 5
4-- 8 . 5 W
Rra'zosin
r
o. 7.5 -
(D t._
abanoquil
r a
s
E
RS
17053
r
-
L_
9 9
O L_
,.r=~
/is/
B
9.5 -
9
=
9
5.5
!
I
I
I
6-5
7.5
8.5
9.5
,
00
9P r a z o s i n
/ 9
-9 6 . 5 -
5.5
9
is/S 9
7.5
==
6.5 -
9
9
.."
."
/
compd
/
5.5
10.5
,
5.5
12 9 , SNAP
6.5
7.5
cloned o l = - A R (pKi)
I
5089
==
'~1
8.5
I
9.5
10.5
cloned o T , - A R (pKi]
Figure 4. Correlation between the binding affinity (pKi) for the cloned ala-AR and the functional affinity (pKb) for the cxl-adrenoceptors in human prostate (A) and rabbit urethra (B). Dashed line represents the line of identity.
100
-
o
80-
/
/
Z
0
.,.._
z
50
-
40
-
/
-
60r
40-
/
0
/
30
-
I
0.1
'
'
' t'"'l
'
'
' t'"'l
1
,
n
u inJ=n !
10 DOSE
[pglkg
100
=
,
n lUnnl !
,
m Z
O I--
20
o D C3
LU I:Z:
"
s
/
20-
-
/
o.. 1:3
-
I--
IZ]
-
-
10
-
0
1000
i.v.)
Figure 5. Effects of cumulative i_v. injection of increasing doses of compound 4 (squares) and prazosin (circles) on NA-induced increase of urethral pressure (filled symbols), and decrease in DBP (open symbols). Each point represents the mean + s.e. of the responses obtained in 9 and 6 dogs, respectively.
149 5-methylurapidil show roughly the same potency on the two tissues, and the tlavone derivatives 4, 7 and 16 exhibit higher potency on urethra than on aorta, suggesting functional selectivity for the lower urinary tract. The in vivo effects of selected a~-AR antagonists were studied, after intravenous admmi~ration, in a dog model [79] where the active doses inh~iting by 50% the urethral contractions induced by k a. NA injection, and the doses active in lowering diastolic blood pressure by 25% were simultaneously evaluated in the same dog. The results obtained are listed in Table 2 together with m vitro and in vivo selectivity indices. A typical dose-response curves for prazosin and compound 4 are shown in Figure 5. In this model, prazosin, terazosin and compound 12 proved devoid of selectivity, whereas tam~axlosin and 5-methylurapidil exhl"oited limited selectivity. SNAP 5089 showed some selectivit3, but its potency against the urethral contractile response was unexpectedly very low. On the contrary, the novel N-arylpiperazine derivatives (in particular compounds 4 and 7) were endowed with very high potency and selectivity for the lower urinary tract. Interestingly, a good correlation between the in vitro potency on rabbit urethra and the in vivo potency in inhibiting the urethral contractions induced by i.a. injection of the agonist in the dog was found (R2 = 0.925). In summary, compounds endowed with high potency in inh~iting contractions of isolated rabbit urethra (a ~tL-AR dependent tissue) showed high potency in inh~iting also the NA induced contraction of the dog urethra in vivo, and in vitro selectivity could be correlated to m vivo selectivity. CONCLUSIONS The growing interest in the field of r subtypes and their antagonists has yielded a number of novel compounds, whose pharmacological evaluation allowed an update of the classification of the tz~-antagonists based on radioreceptor binding studies. A novel class of 4-(4-nitrophenyl)-l,4-dihydropyridines, originated from niguldipine's structure modification, represents the most selective ala-AR antagonists available, highly selective for this subtype versus both other a~-ARs and the other neurotransmitters receptors studied. The N-arylpiperazine derivatives represent the more flexa'ble class which, upon "calibrated" modifications, afforded very selective ~la" and cirri-antagonists. Modification in the benzodioxane group also proved whorthwile, with the identification of t~la-Subtype selective derivatives with a cleaner profile with regard to other receptors. The quinazoline derivatives were confirmed to be ctl-AR selective and unexpectedly afforded a compound si,~nificantly selective for the tZ~b-subtype. The combined use of comparative structure/dynamics simulations and QSAR analysis applied to a set of complexes between structurally heterogenous ligands and the tz~-AR subtypes allowed to obtain energy-minimized models of the ligand-AR interaction. The single residues involved in this interaction have been identified and their effects quantitated, allowing for a more rational design of novel ligands. Correlation of the radioreceptor binding affinity for the tX~d-AR and rat aorta was straightforward, whereas selectivity for other tissues was not so clear. In particular, activity on the tissues of the lower urinary tract of different species, formerly thought to be tzL~-dependent was not confirmed, mainly by using selective Ct~a-AR antagonists. The functional relevance of
150 the atL-AR subtype both in vitro and in vivo could not be ruled out, suggesting the involvement o f this subtype in the pathopysiological response o f the prostate in BPI-I. Although a~L-AR selective antagonists are not yet known, their involvement in the effects on the lower urinary tract in vivo can be anticipated. The screening procedure adopted in this project, based on radioreceptor binding and in vitro and in vivo functional assays, allowed us to select compound 4, which is n o w undergoing Phase II clinical trials for the therapy o f B P H with the codes g e e 15/2739 or SB 216469. Receptor subtyping and development o f selective agents are the means to novel and improved drugs.
REFERENCES
9 10 11 12
14 15 16 17 18 19 20 21 22 23 24 25 26 27
Langer SZ. Brit J Pharmacol 1974; 60: 481-497. Starke K, Montel H, Gayk W, Marker tL Naunyn-Schmiedeberg's Arch Pharmacol 1974; 285: 133-150. Flavahan NA, Vanhoutte PM. Trends Pharmaeol Sci 1986; 7: 347-349. Morrow AL, Creese I. Mol Pharmacol 1986; 29: 321-330. Hart C, Abel PW, Minneman KP. Nature 1987; 329: 333-335. Hieble JP, Bylund DB, Clarke AE, Eikenberg DC, Langer SZ, Lefkowitz ILl, Minneman KP, Ruffolo RR Jr. Pharmaeol Rev (in press). Muramatsu I, Ohmura T, Kigoshi S, I-IashimotoS, Oshita M. Brit J Pharmacol 1990; 99:197-201. Muramatsu I, Ohmura T, Hashimoto S, Oshita M. Pharmacol Comm 1995; 6: 23-28. Hanft G, Gross G. Brit J Pharmacol 1989; 97: 691-700. Boer R, Grassegger A, Schudt CH, Glossman H. Eur J Pharmaeol 1989; 172: 131-138. Cotecchia S, Sehwinn DA, Randall LL, Letkowitz ILl, Caron MG, Kobilka BK. Proc Natl Acad Sci USA 1988; 85: 7159-7163. Schwinn DA, Lomasney JW, Lorenz W, Szklut PJ, Fremeau RT, Yang-Feng TL, Caron MG, Lefkowitz ILl, Cotecchia S. J Biol Chem 1990; 265: 8183-8189. Lomasney JW, Coteechia S, Lorenz S, Leung W, Sehwinn DA, Yang-Feng TL, Brownstein M, Lefkowitz ILl, Caron MG. J Biol Chem 1991; 266: 6365-6369. Perez DM, Piaseik MT, Graham RM. Mol Pharmacol 1991; 40: 876-883. Schwirm DA, Lomasney JW. Eur J Pharmaeol 1992; 227: 433-436. Laz TM, Forray C, Smith KE, Bard JA, Vaysse PJJ, Branchek TA, Weinshank RL. Mol Pharmacol 1994; 46: 414-422. Forray C, Bard JA, Wetzel JM, Chiu G, Shapiro E, Tang R, Lepor H. Hartig PR, Weinshank RL, Branchek TA, Gluchowski C. Mol Pharmacol 1994; 45: 703-708. Forray C, Bard JA, Laz TM, Smith KE, Vaysse PJJ, Weinshank ILL, Gluehowski C, Branchek TA. Faseb J 1994; 8: A353. Ford APDW, Williams TJ, Blue DR, Clarke DE. Trends Pharmacol Sci 1994; 15: 167-170. Testa R, Ta_,:lcl_eiC, Poggesi E, Destefani C, Cotecchia S, Hieble JP, Sulpizio AC, Naselsky DP, Bergsma DJ, Ellis C, Swift A, Ganguly S, Ruffolo RR Jr, Leonardi A. Pharmacol Comm 1995; 6: 79-86. Pimoule C, Langer S, Graham D. Eur J Pharmacol-Mol Pharmacol 1995; 290: 49-53. Schwinn DA, Johnston GI, Page SO, Mosley MJ, Wilson KH, Worman NP, Campbell S, Fidock MD, Fumess LM, Parry-Smith DJ, Peter B, Bailey DS. J Pharmacol Exp Ther 1995; 272: 134-142. Goetz AS, King HK, Ward SDC, True TA, Rimele TJ Saussy DL Jr. Eur J Pharmacol 1995; 272: RS-R6. Hieble JP, Rutfolo RR Jr, Sulpizio AC, Naselsky DP, Conway TM, Ellis C, Swift AM, Ganguly S, Bergsma DJ. Pharmaeol Comm 1995; 6: 91-97. Testa R, Guarneri L, Ibba M, Strada G, Poggesi E, Taddei C, Simonazzi I, Leonardi A. Eur J Pharmacol 1993; 249: 307-315. Cheung YD, Barnett DB, Nahorski SR. Eur J Pharmacol 1982; 84: 79-85. Hoyer D, Hengel G, Kalkman HO. Eur J Pharmacol 1985; 118: 13-23.
151 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60 61 62 63 64 65 66 67
Creese I. Snyder SH. Eur J Pharmacol 1978; 49: 201-202. Meltzer HY, Matsubara S, Lee JC. J Pharmacol Exp Ther 1989; 251: 238-246. Lee HR, Roeske WR, Yamamura HI. Life Sci 1984; 35: 721-732. Ruffolo RR Jr, Stadel JM, Hieble JP. Med Res Rev 1994; 14: 229-270. Ruffolo RR Jr. Tetrahedron 1991; 48: 9953-9980. De Marinis RM, Wise M, Hieble JP, Ruffolo RR Jr. In: Ruffolo RR Jr, ed. The al-adrenergic receptor. Clifton, N.J.: Humana Press, 1987; 211-265. Vizi ES. Med Res Rev 1986; 6:431-449. Timmermans PBMWM, De Jonge A, Thoolen MJMC, Wilffert B, Batink H, van Zwieten PA. J IVied Chem 1984; 27: 495-503. Melchiorre C. Farmaco Ed Sci 1980; 35: 535-550. CamlYoell SF. In: AS Horn and KF De Ranter eels. X-Ray Crystallography and Drug Action. Clarendon: Oxford, 1984; 347-366. De Benedetti laG, Menziani MC, Rastelli G, Cocchi M. J Mol Struct (Theochem) 1991; 233: 343-351. Campbell SF, Hardstone TD, Palmer MJ. JMed Chem 1988; 31: 1031-1035. Rastelh G, FaneHi F, Menziani MC, Cocchi M, De Benedetti 1:)(3. J Mol Struct (Theochem) 1991; 251: 307-318. Glennon RA. In: JG Cannon ed. Advances in CNS drug-receptor interactions. London: JAI Press Inc, 1991; 131-178. Clarke DE, Ford APDW, Williams TJ, Bonhaus D, Vimont R, Blue DR Jr. Pharmacol Comm 1995; 6: 9-14. Chiu G, Gluchowsky C. WO 95/07075 Quaglia W, Pigini M, Tayehati SK, Piergentili A, Giannella M, Marucci G, Melchiorre C. J Med Chem 1993; 36: 1520-1528. Pigini Met al.: personal commumcation. Ford APDW, Arredondo NF, Blue DR, Bonhaus DW, Kava MS, Williams TJ, Vimont RL, Zhu QM, Primer JR, Clarke DE. Brit J Pharmacol 1995; 114: Proc Suppl, 24P. Leonardi A, Riva C, De Toma C, Boi C, Pennini R, Sironi G. Eur J IVied Chem 1994; 29: 551-559. Leonardi A, Motta G, Riva C, Testa R. EP 558245. Russo F, Romeo G, Guccione S, De Blasi A. J Med Chem 1991; 34: 1850-1854. Michel MC, Buscher R, Kerker J, Kraneis H, Erdbrugger W, Brodde O-E. Naunyn-Schmiedeberg Arch Pharmacol 1993; 348: 385-395. Weinberg DH, Trivedi P, Tan CP, Mitra S, Perkins-Barrow A, Borkowski D, Strader CD, Bayne M. Biochem Biophys Res Comm 1994; 201: 1296-1304. Gluchowski C, Wetzel JM, Chiu G, Marzabadi MR, Wong WC, Nagarathnam D. WO 94/22829. Wetzel JM, Miao SW, Forray C, Borden LA, Branchek TA, Gluchowski C. J Med Chem 1995; 38: 1579-1581. Leonardi A, Motta G, Boi C, Testa 1L IT MI 94A000506 Giardina D, Guhni U, Massi M, Piloni MG, Pompei P, Rgmani G, Melchiorre C. J Med Chem 1993; 36: 690-698. Giardina D, Grucianelli M, Melchiorre C, Taddei C, Testa 1L Eur J Pharmacol (in press). King; HK, Goetz AS, Ward SDC, Saussy PL Jr. Neuroscience Abstracts 1994; 20: 526. Michel AD, Loury DN, Whiting RL. Brit J Pharmacol 1989; 98: 883-889. Sleight AJ, Kock W, Bigg DCH. Eur J Pharmacol 1993; 238: 407-410. E1-Bermway M, Raghupathi R, Ingher SP, Teitler M, Maayani S, Glennon RA. Med Chem Res 1992; 2: 88-95. Romeo G, Russo F, Guccione S, Barberulo D, De Blasi A. Farmaco Ed Sci 1995; 50: 471-477. Ruffolo RR Jr, Bondinell W, Ku T, Naselsky DP, Hieble JP. Proc West Phannacol Soc 1995; 38: 121-126. Lu ST, Wu YC, Led SP. Phytochemistry 1985; 24: 1829-1834. Ko FN, Guh I ~ Yu SM, Hou YS, Wu YC, Teng CM. Brit J Pharmacol 1994; 112: 1174-1180. De Benedetti laG, Menziani MC, Fanelh F, Cocchi M. J Mol Struct (Theochem) 1993; 285: 147-153. Ramarao CS, Kinkada Denker JM, Perez DM, Geivin ILl, Riek RP, Graham RM. J Biol Chem 1992; 267: 21936-21945. I-Iirasawa A, Morie K, Tanaka T, Takagaki K, Mural M, Yano J, Tsujimoto G. Biochem Biophys Res Comm 1993; 195: 902-909.
152
68 69 70 71 72
73 74 75 76 77 78 79
Baldwin JM. E M B O J 1993; 12: 1693-1703. Fanelli F, Menzian/MC, Cocchi M, De Benedett/PG. J Mol Stmct (Theochem) 1995; 333: 49-69. Fanelli F, Menzian/MC, Cocchi M, Leonardi A, De Benedetti PG. J Mol Struct (Theochem) 1994; 314: 265-276. Graham RM, Perez DM, Piascik MT, Riek RP, Hwa J. Pharmacol C o m m 1995; 6: 15-22. Testa R, Destefan/C, Guarner/L, Poggesi E, Simonazzi I, Taddei C, Leonarcli A. Life Sci 1995; PL 159-PL 163. Forray C, Chiu G, Wet,zel ~ Bard JA, Weinshank RL, Branchek TA, Hartig P, Gluchowski C. J Urol 1994; 151: 267A. Price D, Schwinn DA, Lomasney JW, Allen LF, Caron MG, Lefkowitz RJ. J Urol 1993; 150: 546-551. Faure C, Pimoule C, Vallancien G, Langer SZ, Graham D. Life Sci 1994; 54: 1595-1605. Goetz AS, Lutz M W , R/mele TJ, Saussy D L Jr. J Pharmacol Exp Ther 1994; 271: 1228-1233. Hieble JP, Caine M, Zalaznik E. Eur J Pharmacol 1985, 107: 111-117. Marshall I, Burr RP, Anderson PO, Chapple CR, Greengrass PM, Johnson GI, Wyllie MG. Br J Pharmacol 1992; 107: 327P. Imagawa J, Akima M, Saka/K. J Phannacol Meth 1989; 22:103- II I.
Perspective in Receptor Research D. Giardinh, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All fights reserved.
153
M o l e c u l a r R e c o g n i t i o n in A d e n o s i n e R e c e p t o r s K.A. Jacobson,* A.M. van Rhee, S.M. Siddiqi, X.-d. Ji, Q. Jiang, J. Kim, and H.O. Kim Molecular Recognition Section, Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, MD 20892 USA. INTRODUCTION Extracellular adenosine acts as a modulator within many physiological systems [1], in general, to compensate for excessive activity of a given organ and to protect it against the detrimental effects of stress. This is a negative feedback loop, by which locally produced adenosine (originating from degradation of either intracellular or extracellular ATP) both reduces the energy demand and increases the oxygen supply. Adenosine is particularly critical m maintaining the homeostasis of essential organs such as the brain, heart, and kidneys, but is also very important for the immune system. In the brain, exogenously administered adenosine agonists have proven exceptionally efficient in neuroprotection [2] (in part, by counteracting the damage caused by excessive glutamate release), and adenosine has been shown to be revolved in pain, cognition, movement, and sleep [3]. Thus, there is a tremendous impetus for the development of therapeutic agents based on selective interactions with adenosine receptor subtypes. Four subtypes of adenosine receptors have been cloned: A 1, A2A, A2B, and A3 [4]. Adenosine agonists, which are almost exclusively derivatives of adenosine, have been sought as potential anti-arrhythmic, anti-lipolytic' (thus anti-diabetic), and cerebroprotective agents (A1), and hypotensive and anti-psychotic agents (A2A). Adenosine antagonists, of which xanthines and a number of fused heterocyclic compounds are representative [1], have been under development as antiasthmatic, anti-depressant, anti-arrhythmic, renal protective, anti-Parldnson's, and cognition enhancing (h~gs. The adenosine receptors are members of the G protein-coupled receptor family, having seven transmembrane helical regions [5]. Site-directed mutagenesis of the A 1 and A2A receptors [6,7] has yielded much insight into structure function relationships. The emerging receptor models [7-9] are approaching a degree of resolution that promises to assist in the design of improved ligands. The discovery of a novel and distinct adenosine receptor subtype, the A 3 receptor [10], has opened new therapeutic vistas m the purmoceptor field. This receptor subtype has a unique SAR (structure activity relationship) profile, tissue distribution, and effector coupling. Reports on A 3 selective agonists have recently appeared [11-13], but the physiological role of A 3 receptors remains to be clarified.
154 In spite of the massive effort to develop selective ligands, a number of agents that initially looked promising did not survive clinical trials [1]. Nevertheless, the interest in adenosine-based therapy has not waned. On the contrary, as our knowledge of the biological effects of adenosine advances, promising, newly envisioned therapeutic applications have become evident. Recently, the use of adenosine agonists in treating stroke has come into focus [2], since, m acute clinical use the interference by some of the previously documented side effects may be tolerable. RECENTLY-DEVELOPED SELECTIVE LIGANDS Highly selective ligands for adenosine A 1 and A2A receptors [1] have been designed both by classical medicinal chemical approaches and by a functionalized congener approach [14]. By the latter approach, a chemically functionalized chain is incorporated at a specific site on a pharmacophore leading to increased flexibility of substitution and enhancement of potency/selectivity via distal interactions at the receptor. In general, for adenosine agonists (Table 1), modification of the N6-position with hydrophobic moieties (such as CHA, N6-cyclohexyladenosine, 9, and CPA, N 6cyclopentyladenosine, 1 0 ) h a s provided selectivity for A1 receptors (and thus cerebroprotective properties). Substitution at the C2-position (such as occurs in CGS 21680, 2-[4- [(2-cm'boxyethyl)phenyl] ethylamino]-5'-N-ethylcarboxamidoadenosine, 11, and its ethylenediamine conjugate, APEC) has resulted in A2A selectivity. A thio- substitution of the 4'-oxygen of 2-chloroadenosine was found to enhance affinity selectively at A2A receptors [23]. Selective antagonists for A 1 receptors (Figure 2) include many 8-aryl and 8-cycloalkyl xanthine derivatives, such as the amine congener XAC, 12, and CPX, 13, which is -500-fold selective for A1 receptors. A thio group may be substituted at the 2-position, but not 6-position, carbonyl group of the xanthine with retention of high affinity. Selectivity for A2A receptors in xanthines has been more difficult to achieve. However, 8-styrylxanthines, such as CSC (8-(3chlorostyryl)caffeine), 14, are A2A selective. The non-xanthine ZM241385, 15, is the most selective A2.,~ antagonist (6800-fold) yet reported [15]. The most recent challenge in the medicinal chemistry of adenosine receptors has been to produce A3 selective agents. A 3 selective antagonists have not yet been reported. One principle of achieving A3 selectivity among adenosine agonists is the combination of optimal substitutions at the N 6- and 5'-positions of adenosine [9,11]. Specifically, among alkyl, cycloalkyl, and aralkyl N 6substituents, a benzyl group is favored, due to its diminished potency at A1 and A2A receptors (Table 1). The A3-selectivity enhancing effects of N6-benzyl
155 Table 1. Affinities (~tM) of adenosine derivatives at rat brain A 1, A2A, and A 3
NLN
receptors, arranged in order of decreasing affinity at rat A3 receptors.a NHR 3
R2
,>
N
!
HO OH
Compound 1. b 2. c 3. d 4. 5. 6. e 7. f 8. g 9. h 10. i 11. J
R1
R2
R3
CH3NHCO C1 3-I-Bz CH3NHCO H 3-I-Bz CH3NHCO H 3-I-4-NH2-Bz CH3NHCO CH3S 3-I-Bz CH3NHCO CH3NH 3-I-Bz C2HsNHCO H Bz CH3NHCO H H C2HsNHCO H H HOCH 2 H cyclohexyl HOCH 2 H cyclopentyl C2HsNHCO NH(CH2)2-pH Ph(CH2)2-
Ki(A1)
Ki(A2A)
0.82 0.054 0.018 2.14 4.89 0.087 0.0836 0.0063 0.0013 0.00059 2.6
0.47 0.056 0.197 3.21 4.12 0.095 0.0668 0.0103 0.514 0.462 0.015
Ki(A3) 0.00033 0.0011 0.0013 0.0023 0.00312 0.0068 k 0.072 k 0.113 k 0.167 k 0.24 k 0.584 k
a. Ki+ S.E.M. determined in radioligand binding assays expressed in ~M, using the following radioligands: A1, [3H]N6-R-phenylisopropyladenosine in rat cortical membranes; A2A, [3H]CGS 21680, 11, binding in rat striatal membranes; A 3, [i25I]AB-MECA, 3, binding, unless noted, in membranes of CHO cells stably transfected with the rat A3-cDNA; b. CI-IB-MECA; c. IBMECA; d. I-AB-MECA; e. Bz-NECA; f. MECA; g. NECA; h. CHA; i. CPA; j. CGS 21680; k. A 3 affinity determined versus [125I]APNEA (N 6aminophenylethyladenosine) binding.
156 modification are additive with the A3-affinity enhancing effects of the 5'uronamido function, as in NECA (adenosine-5'-N-ethyluronamide), 8. The first such hybrid molecule to show A3 selectivity [9] was NG-benzyl-NECA, 6. In a comparison of various 5'-uronamido groups in mono-substituted adenosine derivatives, the 5'-N-methyluronamide, 7 [11], had particularly favorable A3 receptor vs. A1/A2A affinity. Empirical and QSAR studies [11,20] of substituent effects on the N6-benzyl group have shown that substitution at the 3-position with sterically bulky groups, such as the iodo group, is optimal, leading to the development of the highly potent A3 agonist N6-(3-iodobenzyl)-adenosine-5'-Nmethyluronamide (IB-MECA), 2, which is 50-fold selective for A3 vs. either A1 or A2 receptors in vitro [11] and appears to be highly A 3 selective in vivo [21]. [125I]I-AB-MECA, 3, was introduced as a high affinity radioligand for A 3 receptors [21]. 2-Substitution in combination with modifications at N 6 and 5'-positions was found to further enhance A 3 selectivity [12]. Adenosine derivatives bearing an N6-(3-iodobenzyl) group, reported to enhance the affinity of adenosine-5'uronamide analogues as agonists at A3 adenosine receptors, were synthesized starting from 1-O-methyl ~-D-ribofuranoside in 10 steps [12]. 2-Chloro-N6-(3 iodobenzyl)-adenosine-5'-N-methyluronamide, 1, which displayed a Ki value of 0.33 nM, was selective for A3 vs. A1 and A2A receptors by 3600- and 2000-fold, respectively. Compound 1 was 66,000-fold selective for A3 receptors vs. the Na +independent adenosine transporter, as indicated in its weak displacement of [3H]S-(4-nitrobenzyl)thioinosine binding at this transporter in rat brain membranes. In a functional assay of rat A3 receptors expressed in CHO cells, it inhibited adenylyl cyclase with an IC50 of 67 nM. 2-Methylthio-N6-(3 iodobenzyl)adenosine-5'-N-methyluronamide, 4, and 2-methylamino-N6-(3 iodobenzyl)-adenosine-5'-N-methyluronamide, 5, were less potent, but nearly as selective for A3 receptors.
DO XANTHINES BIND TO A3 RECEPTORS? The A3 adenosine receptor cloned from rat [10] was shown to be unique among the subtypes in that agonist action is not antagonized by xanthines, such as theophylline. The affinity of certain xanthines, such as BWA522 (Figure 1), 16, is greater in the human and sheep homologues of the A 3 receptor [16,17] than in the rat. Typical Ki values at rat A 3 receptors of roughly 10 .4 M have been obtained for many xanthines that have nearly nanomolar potency at the A1 or A2A subtypes. X A C , 12, for example at 1 pM, has been used in
157
Pr\
N
O .LL
H I~I I
O LI
H ~
O,-~N /
--~N
Pr\
/
oc
I
I
Pr
~
-
Pr
12, XAC
13, CPX
O OH3 CH3.N.,~ _~1~1
C!
.o
~
CH3
NH.,J,,.-N,,,J~N/~--' I
Bu
o~~~> I
H 18
19
]'1'
HO OH
20
~-~ HO OH
Ki (~M) at Rat A 3 Receptors:
143
6.03
0.229
A1 Receptors:
0.50
4.19
37.3
Figure 2. Ribose moiety at the 7-position anchors xanthine in the A 3 binding site.
158 pharmacological experiments in vitro and in rodents [2] for distinguishing A3 receptors from A1 and A2A receptors. In an effort to synthesize A3 antagonists, we have attempted to maximize the affinity of xanthine derivatives at the binding site [18]. One such xanthine is compound 17, which had a K i value at rat A3 receptors of 9.4 ~M. The presence of an anionic group on the xanthine tended to diminish the affinity at A1 and A2A receptors. Thus, compound 17 is 7-fold selective for rat A3 vs. A2A receptors. Molecular modeling of adenosine receptors (supported by mutagenesis) suggests that two histidine residues in the sixth and seventh transmembrane domains (TM6 and TM7) are important for ligand recognition [5-8]. We have proposed [9] that the ribose moiety of adenosine, that is relatively more important for high affinity binding to A3 receptors than at other subtypes, is coordinated to a conserved histidine residue in TM7 (His278 of hA2A, see below). Consequently, we tested the hypothesis that a means of anchoring xanthines in the A 3 binding site is by adding a sugar moiety at the 7-position to form xanthine-7-ribosides (Figure 2). Several members of this class of compounds, the 1,3-dialkylxanthine-7-ribosides, have been synthesized and were previously found by IJzerman and colleagues to bind weakly to A1 receptors [19]. At rat brain A3 receptors, 1,3-dibutylxanthine-7-riboside (DBXR), 19, was found to bind with a Ki value of 6.03 ~M [9],. whereas the parent xanthine, 1,3dibutylxanthine, 18, displayed a Ki value of 143 p~M. Thus, the presence of the ribose moiety enhances affinity of xanthines at rat A3 receptors, while at A 1 receptors the xanthine-7-riboside derivatives are, as a rule, less potent than the parent xanthines. Functionally, 1,3-d~butylxanthine-7-riboside, 19, as a structural hybrid of classical A1/A2A agonist and antagonist molecules, appeared to act as a partial agonist at rat A 3 receptors [9], providing hope that this was a means of designing antagonists. However, upon structural modification that increased the potency and selectivity of the xanthine ribosides at A 3 receptors, full agonism was achieved. Specifically, the structural parallel between adenosine derivatives and the xanthine-7-ribosides was maintained with respect to A 3 receptor affinity. This parallel lead to the design of 1,3-dibutylxanthine-7riboside-5'-N-methylcarboxamide (DBXRM, Figure 2), 20, having a Ki value of 229 nM at A3 receptors with 1G0-fold and >400-fold selectivity for A3 vs. A1 and A2A receptors, respectively. The selectivity of this compound is a result of incorporation of the 5'-methyluronamide group, found to enhance A 3 selectivity in IB-MECA, 2, and optimization of the alkyl chain length at positions 1 and 3. Unlike 1,3-dibutylxanthine-7-riboside, DBXRM acted as a full agonist in the rat A3 receptor-mediated inhibition of adenylyl cyclase. Thus, there was a tendency
159 towards the increase of efficacy as the affinity increased within the same series of compounds.
SITE-DIRECTED MUTAGENESIS AND MOLECULAR MODELING OF ADENOSINE R E C E P T O R S In addition to chemical probing of the binding sites of adenosine receptors, we have also used molecular biological approaches to determine structure function relationships for human A2A adenosine receptors [7,22]. Amino acid residues were mutated and the constructs expressed in COS-7 cells. Numerous residues in transmembrane domains 3, 5, 6 and 7, individually replaced with alanine and other amino acids, were identified as essential for ligand recognition. An immunological method [8] was used to determine whether the pharmacologically inactive mutant receptors were properly oriented at the cell surface and not simply retained in an intracellular compartment. Thus, an epitope tag was attached near the N-terminus of the receptor to allow detection with an antibody. This extra l 1-amino acid sequence did not interfere with ligand binding or adenylyl cyclase activation by the human A2A receptor. The pharmacological properties of mutant receptors were determined both m radioligand binding experiments and through stimulation of adenylyl cyclase. Specific binding of [3H]CGS 21680, 11, and [3H]XAC, 12, an A2A agonist and antagonist, respectively, was measured. Although A 1 selective in the rat, XAC has greatly enhanced affinity at human A2A receptors, and it is useful as a nonselective radiotracer in this species. High affinity binding of either agonist or antagonist was absent m the following Ala mutants: F182A, H250A, N253A, I274A, H278A and $281A, although they were well expressed in the plasma membrane. The hych'oxy group of $277 is required for high affinity binding of agonists, but not antagonists, since the $277A mutant bound [3H]XAC but not [3H]CGS 21680 similar to the ~dld type receptor. /h~ N181S mutant lost affinity for adenosine agonists substituted at the N 6 or C-2, but not at the C-5' position. The mutant receptors I274A, $277A, H278A showed full stimulation of adenylyl cyclase at high concentrations of CGS 21680. The functional agonist potencies at mutant receptors that lacked radioligand binding were >30-fold less than those at the wild type receptor. A molecular model based on the structure of rhodopsm was developed concurrently to mutagenesis studies in order to visualize the environment of the ligand binding site. We have found that the low resolution structure of rhodopsin serves as a more versatile template for G protein-coupled receptors (GPCRs) than the coordinates of bacteriorhodopsin [7]. The A2A receptor model was composed in steps including: construction and energ~ minimization of each helix individually, composition of the helical bundle based on consideration of
160
161 Figure 3 [upper left]. Vicinity of the bound ligand NECA in the rhodopsin-based molecular model of the human A2A receptor. Docking of the ligand was based on mutagenesis results, e.g. single amino acid substitution of the receptor, followed by energy minimization [7]. A putative pocket consisting of four aromatic residues shown m space f'filing form surrounds the adenine moiety of adenosine. Figure 4 [lower left]. Vicinity of the ribose moiety of NECA to hydrophilic residues of TM7 m the rhodopsin-based molecular model of the human A2A receptor [7]. Mutation to Ala of each of the residues shown in space filling form prevents the high affinity binding of [3H]CGS 21680.
Figure 5. Putative overlay of the agonist NECA, 8, and the antagonist XAC, 12, h~ the human A2A receptor model according to the 'N6/C8' hypothesis. Transmembrane helical regions are represented as ribbon structures. NECA was docked and minimized as above [7]. The amino group of XAC rests near the exofacial side of the receptor.
162 homology among GPCRs and rotation of amphipathic helices such that predominantly hydrophobic faces are pointed towards the lipid bilayer. Only the final step in construction of the model, i.e. docking of a ligand, NECA, took into account the results of mutagenesis. Thus, the receptor portion of the model, based only on computational methods and previous pharmacological structural insights into adenosine receptors [8,9] and other GPCRs, was highly predictive of which residues were potentially facing m the direction of the binding site. Most of the positions at which alanine mutation was not tolerated, were calculated to have site chains pointing into the binding cleft. This helped to define the placement of the ligand in the binding site. The current model takes into consideration only the contribution of the transmembrane helical domains, however a mutagenesis study by Olah et al. [24] has revealed that a portion of the second extracellular loop is also involved m antagonist recognition by A 1 receptors. Some of the residues targeted in this study may be involved in the direct interaction with ligands in the human A2A adenosine receptor. Four aromatic side chains in TM6 and TM7 appear to form a cluster in the receptor model that constitutes a putative binding pocket for the adenine moiety (Figure 3). The residues are H250, Y257, and F182 (all found to be essential by mutagenesis) and Y183 (not mutated in our study) [7]. H250 in TM6 appears to be a required component of a hydrophobic pocket, and H-bonding to this residue is not essential since the H250F mutant receptor binds radioligand like wild type. On the other hand replacement of H278 in TM7 with other aromatic residues was not tolerated in ligand binding, since hydrophilic groups at this position are required. According to the molecular model N253 is in proximity to form an Hbond with the exocyclic amino group of adenosine, h~ fact, the N253A mutant receptor did not bind either radioligand with high affinity. The xibose moiety appears to span a region of hydrophilic residues located in TM3 and TM7. Figure 4 shows the proposed interaction of the ribose moiety of NECA with hydrophilic residues of TM7. h~ our A2A adenosine receptor model the 5'-NH in NECA is hych'ogen bonded to $277 and H278. We have found that the T88A (TM3) mutant receptor binds antagonists like wild type receptors, but fails to bind agonists with high affinity [22]. In the biogenic amine receptors, an aspartate residue conserved in TM3 is essential as a counterion to the charged ammonium group of the ligand. In the adenosine receptors a Val residue occurs at the position of this Asp, and the essential T88 occurs roughly one helical turn closer to the cytoplasmic side of the helix. In the model, the ri.bose region is in proximity to T88. Certain mutant A2-x receptors have revealed differences in affinity shifts between agonists and antagonists, for example: $281N (agonists become more potent and antagonists less potent) and mutations of T88 (agonists become much less potent). Therefore, a partially overlapping set of amino acid residues
163 in the receptor are proposed to be involved in agonist vs. antagonist binding. We attempted to contrast the agonist and antagonist binding domains in the A2A receptor model. Based on SAR insights, the nitrogens of xanthines and adenosine derivatives, although both purines, certainly do not occupy similar coordinates in the receptor-bound state. At least three models for mapping of agonists onto antagonists have been proposed [25]. Extending this analysis to the entire receptor model clearly favors one of the three hypotheses, i.e. the 'N6/C8 ' model. In this hypothesis, the C-8 substituent of xanthines overlays the N 6 substituent of adenosine analogues. Figure 5 shows the relative orientation when the antagonist XAC is docked into our NECA-occupied receptor model according to this hypothesis, whereas in applying the 'flipped' hypothesis the C8substituent side chain of XAC protrudes into the helical segment (not shown), which is energetically untenable.
REFERENCES 1 Jacobson KA, van Galen PJM, Williams M. J Med Chem 1992; 35: 407-422. 2 von Lubitz DKJE, Lin RC-S, Popik P, Carter MF, Jacobson KA. Eur J Pharmacol 1994; 263:59-67. 3 von Lubitz DKJE, Jacobson KA In: Bellardinelli, L., Pelleg, A., Eds. Adenosine and Adenine Nucleotides: From Molecular Biology to Integrative Physiology; Kluwer: Norwell, 1995; 489-498. 4 Jacobson MA. In: Bellardinelli, L., Pelleg, A., Eds. Adenosine and Adenine Nucleotides: From Molecular Biology to Integrative Physiology; Kluwer: Norwell, 1995; 5-13. 5 van Galen PJM, Stiles GL, Michaels G, Jacobson KA. Med Res Rev 1992" 12:423-471. 6 Olah ME, Ren HZ, Ostrowski J, Jacobson KA, Stiles GL. J Biol Chem 1992; 267"10764-10770. 7 Kim J, Wess J, van Rhee AM, Sch6neberg T, Jacobson K. J Biol Chem 1995 270:13987-13997. 8 IJzerman AP, van Galen PJM, Jacobson KA. Drug Des Discov 1992; 9:49-67. 9 van Galen PJM, van Bergen AH, Gallo-Rodriquez C, Melman N, Olah ME, IJzerman AP, Stiles GL, Jacobson KA. Mol Pharmacol 1994; 45: 1101-1111. 10 Zhou QY, Li CY, Olah ME, Johnson RA, Stiles GL, Civelli O. Proc Natl Acad Sci USA 1992" 89:7432-7436. 11 Gallo-Rodriguez C, Ji XD, Melman N, Siegman BD, Sanders LH, Orlina J, Fischer B, Pu QL, Olah ME, van Galen PJM, Stiles GL, Jacobson KA. J Med Chem 1994" 37:636-646. 12 Kim HO, Ji XD, Siddiqi SM, Olah ME, Stiles GL, Jacobson KA. J Med Chem 1994 373614-3621.
164
13 Kim HO, Ji XD, Melman N, Olah ME, Stiles GL, Jacobson KA. J. Med. Chem. 1994; 37:4020-4030. 14 Jacobson KA, Ukena D, Padgett W, Kirk KL, Daly JW. Biochem Pharmacol 1987; 36:1697-707. 15 Poucher SM, Keddie JR, Singh P, Stoggall SM, Caulkett PWR, Jones G, Collis MG. Br J Pharmacol 1995; 115:1096-1102. 16 Linden J, Taylor HE, Robeva AS, Tucker AL, Stehle JH, Rivkees SA, Fink JS, Reppert SM. Mol Pharmacol 1993; 44:524-532. 17 Salvatore CA, Jacobson MA, Taylor HE, Linden J, Johnson RG. Proc Natl Acad Sci U S A 1993; 90:10365-10369. 18 Kim HO, Ji XD, Melman N, Olah ME, Stiles GL, Jacobson KA. J Med Chem 1994; 37:3373-3382. 19 IJzerman AP, van der Wenden EM, von Frijtag Drabbe Kiinzel J, MathSt RAA, Danhof M, Borea PA, Varani K. Naunyn Schmiedebergs Arch Pharmacol 1994; 350:638-645. 20 Siddiqi SM, Pearlstein RA, Sanders LH, Jacobson KA. Bioorg Med Chem 1995, 3:1331-1343. 21 Jacobson KA, Nikodijevic O, Shi D, Gallo-Rodriguez C, Olah ME, Stiles GL, Daly JW. FEBS Lett 1993; 336:57-60. 22 Jiang Q, van Rhee AM, Kim J, Yehle S, Wess J, Jacobson IC J Biol Chem 1995, submitted. 23 Siddiqi SM, Jacobson KA, Esker JL, Melman N, Tiwari KN, Secrist JA, Schneller SW, Cristalli G, Johnson CA, IJzerman AP. J Med Chem, 1995; 38:1174-1188. 24 Olah ME, Jacobson KA, Stiles GL, J Biol Chem 1994; 269:24692-24698. 25 van der Wenden EM, Price SL, Apaya RP, IJzerman AP, Soudijn W. J Comp Aided Mol Design 1995; 9:44-54.
Perspective in Receptor Research D. Giardin~,, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
C H E M I C A L AND P H A R M A C O L O G I C A L ADENOSINE RECEPTOR AGONISTS
165 P R O F I L E OF S E L E C T I V E
G. Cristalli, a E. Camaioni,b E. Di Francesco, b S. Vittorib and R. Volpinib aDipartimento di Scienze Farmaceutiche, Universi~ di Modena, 41100 Modena bDiparfimento di Scienze Chimiche, Universit~ di Camerino, 62032 Camerino, Italy INTRODUCTION There is evidence that the purine nucleoside adenosine specifically modulates neurotransmission through the interaction with four cell surface receptors recently classified as A1, A2a, A2b, and A3 [1 ]. These receptor subtypes have been cloned and characterized as belonging to the supeffamily of receptors with seven transmembrane helices that couple to G proteins [2]. Over the last decade a large number of adenosine analogs with high affinity and selectivity for A1 and A2a and, more recently, A3 receptors have been synthesized [3]. However, the existence of at least four AR subtypes necessitates the generation of more discriminating compounds which may ultimately have therapeutic use. More in detail, selective A I receptor agonists might be of therapeutical interest as antiarrhythmic and cardioprotective agents since: a) the cardiodepressant effects of adenosine appear to be important in the pathogenesis of arrhythmias in ischemic heart desease [4,5]; b) adenosine exerts Al-mediated cardioprotective action in preconditioning, a phenomenon by which one or more brief periods of ischemia result in improved functional recovery after substained ischemia [6,7]. Selective A2a receptor agonists have a potential for the treatment of cardiovascular disorders such as hypertension, ischemic heart desease, and atherosclerosis since it is known that stimulation of adenosine A2a receptors leads to vasodilation, inhibition of platelet aggregation, and neutrophil adhesion to vascular endothelium, and reduction in generation of oxygen free radicals by activated neutrophils [8,9]. Selective A3 receptor antagonists have potential use in the treatment of asthmatic and other allergic conditions, and may afford cardioprotection from prolonged ischemia [10]. In this paper our contribution both to the improvement of structure-activity relationships and to the development of A1 and A2a potent and selective agonists will be reported.
166
~~
N
.~ HO OH
HO OH 1-DeazaADO
ADO
C, N; X0-HO OH
1-DeazaCCPA
N
HO-~ HO OH CCPA
H O = ~ i~ HO OH CPA
Figure 1 CCPA We have developed interest in adenosine receptors by investigating the role of the purine nitrogens on adenosine activity. Several deaza analogues of adenosine were therefore synthesized and tested for affinity at A1 and A2a receptors in radioligand binding studies [11 ], and for their activity as inhibitors of human platelet aggregation [12]. We found that only 1-deazaadenosine (1-deazaADO) displayed appreciable aff'mity for adenosine receptors with a 25 fold A 1-selectivity, and presented moderate activity as inhibitor of human platelet aggregation induced by ADP (IC50 = 65 ktM). In a search for more potent and selective A1 adenosine receptor agonists, and taking in mind that at A1 receptor the most active analogues were N 6 substituted adenosines, a series of N6-cycloalkyl-1-deazaadenosine derivatives were synthesized and evaluated in radioligand binding studies and adenylate cyclase assays [13]. N6Cyclopentyl-2-chloro-1-deazaadenosine (1-deazaCCPA) showed the highest affinity at A1 receptor with more than two fold higher A1 selectivity than N 6cyclopentyladenosine (CPA).
167 Table 1 Effects of 1-deazaADO, 1-deazaCCPA, CCPA, and CPA on radioligand binding, and adenylate cyclase assays. Adenylate cyclase c Binding assay b Compd. a
A1 (Ki, nM)d
A2a (Ki, nM)d
A1 (IC50, nM)d
A2a (ECs0, nM)d
1-DeazaADO
115 (91-144)
2 900 (2400-3500)
6 7 00 (4100-11000)
18000 (11000-28000)
1-DeazaCCPA
1.6 13200 360 28000 (1.1-2.5) (10900-15900) (220-660) (20000-39000) CCPA 0.4 3900 33 3500 (0.2-0.7) (2500-4700) (22-51) (3200-3800) CPA 0.8 2000 58 2200 (0.6-1.0) (140o-2900) (24-140) (1500-320o) aThe structure of compounds is reported in Figure 1. bKi values were calculated from competition for [3H]PIA binding to rat brain membranes (A1 receptor), and for [3H]NECA binding to rat striatal membranes in the presence of 50 nM CPA (A2a receptor), cECs0 and IC50 values were calculated from the inhibition of rat fat cell adenylate cyclase (A1 receptor), and the stimulation of human platelet adenylate cyclase (A2a receptor), dData are means from at least three separate experiments; 95% confidence limits are in parenthesis [13,14]. The finding that the contemporary presence of a chlorine in 2-position and a cyclopentyl substituent on the N-6 amino group increased both A1 affinity and selectivity prompted us to synthesize the 2-chloro derivative of CPA, namely CCPA [141. In Table 1, the radioligand binding and the adenylate cyclase data referred to 1deazaADO, 1-deazaCCPA, CCPA, and CPA are reported. CCPA proved to be both more active and A 1-selective than the reference compound, showing A1 affinity in the subnanomolar range and a selectivity three fold that of CPA. CCPA is now available in the market also in the tritiated form, obtained by catalytic reduction in a tritium atmosphere of the corresponding cyclopentenyl derivative [15]. A detailed pharmacological profile of CCPA in binding, functional, and in vivo models has been recently reported by Monopoli et al. [16], and some data are shown in Table 2.
168 (,t:x~,)~,coox
Nx,
Nx,
1CH212 -HN-"~N/ " N" CH3-1CH213-'C--EtHNC~ EtHN~,~CO
EtHN~ HO
.,.
OH
HO
NECA
OX
CGS 21680
NH2
cx3-(cx2N~c EIXN~..~ HO OH
THENECA
NH2
EIHN~ HO OH
(R)-PHPNECA
HO
OH
HENECA
NH2
l~'~J EtHN~ HO OH
(S)-PHPNECA
Figure 2 I-IENECA As far as A2a receptor, over the last few years there has been a considerable effort directed toward both discovery and characterization of potent and selective agonists and antagonists of this subtype [17]. A major advance in the search of A2a adenosine agonists has been made with the introduction of C-2 substituents in the adenosine structure [18]. Selectivity and potency of the C-2 substituted adenosine analogs are greatly improved by replacing the 5'-hydroxyl group with other substituents, in particular with the N-ethylcarboxamide group. For example, the prototypical A2a agonist 5'-N-ethylcarboxamidoadenosine (NECA) showed little or no A2a selectivity [19] whilst [2-[4-(carboxyethyl)phenethyl]amino]adenosine-5'-N-ethylcarboxamide (CGS21680) is a moderately selective A2a agonist which is widely used as a reference compound in biochemical and pharmacological studies [20]. On this basis, we have reported the synthesis of the 2-hexynyl derivative of NECA, identified as HENECA, which showed high affinity at A2a adenosine receptors, and good A2a vs A1 selectivity. HENECA, in addition to having good A2a selectivity similar to that of CGS21680, shows higher inhibitory activity on platelet aggregation [21 ].
Table 2 Activity of some adenosine agonists on in vitro assays. Binding assaya Adenylate cyclaseb Compd. (fi,Me (EC50, n W e Rat brain Rat striatum Human PC12 cell A1 A2a platelet 130 2.2 14 4. I HENECA ( 1 16-145)
CGS21680
569 ( 5 1 1-634)
NECA
10.4 (9.4-1 1.5)
CCPA
I .3 (1.1-1.4)
(1.9-2.6)
11.0 (9.4- 12.9)
7.8 (6.6-9.1)
650 (555-762)
(13-15)
83 (7 8- 8 8)
Functional activityc (EC50,
Rat atria A1
>lOpM
(3.7-4.6)
72
(244-1460)
> 10pM
(66-78)
99
54.8
(82-1 16)
(30- 150)
1200
1200
(1000-1400) ( 1 170-1230)
115 (53-251)
( 1 10-112)
110
Rat aorta A2a 596
8.2
3 94
Antiaggregatory activityd (Ic50, nMIe Rabbit Human plateletf plateletb 70 50 (30- 150)
2160 (1620-3620)
210
(4 1-60)
820 (640-1 100)
3 60
(209-742)
(160-280)
(350-380)
>l o w
>lOpM
>lOpM
(4.4-15.3)
aReceptor binding affinity at A1 and A2a receptors was determined using (3HICHA and [3H]CGS21680 as radioligands, respectively [ 16,261. bECso values were calculated from the stimulation of PC12 adenylate cyclase and human platelet adenylate cyclase (A2a receptors) [ 151. CNegative chronotropic activity was studied on rat atria induced by cumulative addition of an agonist, and was recorded by measuring the isometric contractions: vasodilating effects were calculated on rat aorta precontracted by PGF2a (3 pM) [26]. dPlatelet aggregation was induced by ADP. eData are means from at least three separate experiments; 95% confidence limits are in parenthesis. ‘Data are from Sandoli et al. [24].
170 A detailed characterization of HENECA in comparison with CGS21680, NECA, and CCPA is reported in Table 2. In this Table, in addition to the binding data, the activity on the stimulation of adenylate cyclase in platelet and PC12 cells is shown. The results of the cyclase assays are in good agreement with the higher affinity of HENECA for the A2a receptor in comparison with the other adenosine agonists [22]. In the same Table the negative chronotropic activity in rat atria and the vasodilating activity in rat aorta of the four compounds are reported [23]: the profile of selectivity accounts for the higher chronotropic effect of CCPA in rat atria. The adenosine analogues were also evaluated as inhibitors of rabbit [24] and human [22] platelet aggregation induced by ADP. The ICs0 of NECA is about 200 nM in the rabbit, and about 350 nM in the human platelets. CCPA is a very poor inhibitor, according to its A1 selectivity; CGS21680 is less potent than NECA, while HENECA resulted from 3 to 7 times more potent than the model compound. Moreover, Dionisotti and coworkers evaluated in vivo rabbit platelet accumulation in response to ADP, given either before or after drug infusion. NECA, and, to higher extent, HENECA clearly decreased platelet accumulation in the rabbit. This antiaggregatory activity is of short duration, and does not appear to be due to hemodynamic changes, but rather is a result of selective drug action on platelet adenosine receptors [25]. In the same experiment the selective A1 adenosine agonist CCPA resulted inactive. The predominant effects on the cardiovascular system elicited by the same compounds in spontaneously hypertensive rats have been investigated by Monopoli et al. [ 16,26]. All the compounds induced a dose-dependent decrease in sistolic blood pressure. As expected, the effect of HENECA is due to vasodilation, and is accompanied by slight tachycardia. According to its A1 profile, CCPA markedly reduced heart rate and did not show any relevant vasodilating properties. The non selective agonist NECA induced bradicardia only at high doses. All tested drugs appear to penetrate into the brain. This is demonstrated by the general protection of pentylenetetrazole-induced lethal seizures in rats. HENECA showed protective effects at doses 10-fold higher than those active on the cardiovascular system, but the sleep-waking cycle was not influenced by the compound over the dose range examined. Conversely NECA and, to a lower extent CCPA, altered the sleep pattern mostly decreasing the REM sleep stage at high doses [16,26].
171 ALKYNYL AND ALKENYL DERIVATIVES OF NECA The therapeutic potential of HENECA for the treatment of cardiovascular diseases, and the need for more selective A2a agonists prompted us to synthesize a number of new 2-alkynyl derivatives of NECA bearing hydroxyl, amino, chloro, cyano, and heterocyclic groups or substituted aromatic or heteroaromatic tings in the side chain [27,28]. For the synthesis of the 2-alkynyl derivatives, three different approaches have been used, owing to different reactivity of the side chains or separative problems during purification. Briefly, from the common intermediate, the 5'-carboxyl-2',3'isopropyliden derivative, the two carboxamido derivatives I and 5 were obtained as starting material for the synthetic routes a-d depicted in Scheme 1. In any case the introduction of the alkynyl chain was carried out by a modification of the classical cross-coupling reaction catalysed by palladium. Moreover, some alkenyl and alkyl derivatives of NECA were also synthesized as classical modification of the triple bond [29]. In the case of the alkenyl derivatives, it is possible to design two geometric isomers (cis or Z, and trans or E). They were obtained by two different approaches illustrated in Scheme 1. The synthesis of Z-alkenyl derivatives were carried out by partial reduction of triple bond with the Lindlar catalyst (route f). The fully saturated derivatives were obtained starting from the same alkynes by using palladium on charcoal as a catalyst (route g). The synthesis of the E-alkenyl derivatives was accomplished by reaction of the corresponding alkynes with catecholborane followed by cross-coupling reaction of this product with the protected 2-iodoNECA. Deprotection in acidic medium gave the desired isomers (route a and b). The interaction of the new 2-alkynyl and 2-alkenyl derivatives of NECA with the adenosine receptors was evaluated using both radioligand binding technique and functional assays. The same compounds were also tested as inhibitors of platelet aggregation induced by ADP. The results of the binding experiments and antiaggregatory tests in the alkynyl series are reported in Table 3. From the binding data it is possible to draw some structure-activity relationships: -The presence of an o~-hydroxyl group in the alkynyl chain of NECA derivatives accounts for the A2a agonist potency, leading to compounds endowed with subnanomolar affinity in binding studies. However, these analogues also possess good A1 receptor affinity resulting in low A2a selectivity.
172 Scheme 1
NH2
N~ ' ~
N.H2
N
EtNH~~I~
N.H2
N~
(a)
N
R EtNHCO
N~
(b)
I
N
H--C=C.~NI~N~J HO
l
OH
3
(c)
NH2
Nx2 N~
N
N"~'"
EtNH~II~
NH2
N~ ' ~
N
.L. II ij R-C--C N,'~ z
HO
N
OH 59
EtNX~ HO OH 7-58
T NHz
(e)
NX2 ,.L-N i ~ Y
HO
OH 5
(a)
x
NH2 "
HO
OH
.'-(c
6
HO
OH 60
(a) Alkenyl catacholeborane derivatives, CH3CN, tetrakis, and K3CO3 at 90~ (b) CF3COOH or 50% HCOOH. (c) Alkynes, CH3CN or DMF, (Ph3P)2PdC12, CuI, and Et3N. (d) 1) Trimethylsylylacetilene, CH3CN or DMF, (Ph3P)2PdC12, CuI, and Et3N; 2) KOH. (e) Aromatic halide (Br or I), DMF, (Ph3P)2PdC12, CuI, and Et3N. (f) H2 18 psi, Lindlar catalyst. (g) H2 40 psi, 10% Pd/C.
173 -Aromatic or heteroaromatic tings conjugated to the triple bond conferred in general weaker activity and lower selectivity than aliphatic chains -Introduction of methylene groups between the triple bond and the phenyl ring markedly increased the A2a binding affinity resulting in compound 47 endowed with a 180-fold A2a selectivity, when three methylenes are present in the side chain (Table 4). -Reduction of triple to double bond led to compounds whose activity is strictly related to cis-trans isomerism, the trans isomers being more active and selective than the cis ones. Fully saturation of the side chain markedly reduced adenosine receptor affinity. The results of radioligand binding experiments performed on cis and trans isomers of 2-hexenylNECA, and on 2-hexylNECA are reported in Table 5. In comparison with HENECA, trans 2-hexenylNECA (THENECA, 3) showed similar aff'mity at AEa receptor, and a 3-fold higher selectivity for this subtype. Hence THENECA proved to be about 160-fold A2a selective with a Ki of 1.6 nM at A2a receptors. The antiaggregatory effect of the new alkynyl derivatives of NECA on rabbit platelet aggregation induced by ADP is reported in Table 3 as potency ratio calculated versus NECA. Some compounds resulted more potent than HENECA itself as inhibitors of platelet aggregation. All of them bear a hydroxyl group or a heteroatom in the side chain, and it is evident that introduction of a polar group in the side chain increased both affinity for A1 receptors and activity as platelet aggregation inhibitors. It is interesting to notice that PHPNECA (16) is about 15-times more active than NECA. More in detail, the most active compounds in the series present an a-hydroxyl group (Table 3, compds 11-13, 15, and 16), or a 5-hydroxyl group (10) which, however, may be so flexible to mimic the a-position. The importance of the hydroxyl group is stressed by comparing the three benzyl derivatives shown in Table 6. Introduction of an hydroxyl group on the methyl ct to the triple bond greatly improved the antiaggregatory potency in comparison with the first compound in the Table (45), resulting in the ct-hydroxybenzyl derivative PHPNECA (16), the most potent inhibitor in the series with IC50 of 13 nM. However, the contemporary amethylation markedly reduced the antiplatelet activity without affecting the A2a binding affinity, as shown by compounds 17 and 14 in Table 6. From these data it is clear that the hydroxyl group did not influence the A2a binding affinity while it is crucial for the antiaggregatory activity.
174 Table 3 In vitro pharmacological activity of 2-alkynyl derivatives of NECA. Compd.
Binding assay a (Ki, nM) b
R
Antiaggregatory activity (potency
Rat brain
Rat striatum
ratio vs NECA) c
A1
AZa
Rabbit platelets
6
H
35.4 (21.9-57.2)
57.8 (48.9-68.3)
0.30
7
CH2OH
14.1 (7.8-256)
9.1 (6.0-13.7)
2.30
8
(CH2)2OH
47.3 (42.8-52.4)
10.8 (9.8-12.0)
1.10
9
(CH2)aOH
99.9 (89.6-111)
11.3 (10.1-12.5)
2.20
10
(CH2)4OH
42.1 (39.9-44.5)
6.8 (6.0-7.7)
4.80
11
CH(OH~H3
11.1 (10.1-12.2)
7.6 (6.6-8.7)
4.70
12
CH(OH)CH2CH3
20.4 (18.5-22.6)
12.4 (10.8-14.2)
14.10
13
CH2CH(OH)CH3
69.6 (64.7-74.9)
56.4 (52.3-60.8)
3.20
14
C(OH,CHa)-[But
3.0 (2.5-3.6)
0.5 (0.4-0.6)
0.54
15
1-Hydroxycyclopentyl
4.0 (3.5-4.5)
0.6 (0.5-0.7)
5.30
16
CH(OH)Ph
2.5 (2.2-2.9)
0.9 (0.7-1.3)
15.70
17
C(OH, CH3)Ph
32.7 (29.7-35.9)
1.7 (1.6-1.8)
0.26
18
(CH2)2OPyr d
267 (191-375)
5.0 (4.8-5.2)
0.30
19
CH(OCH2CH3) 2
656 (515-835)
20
CH2NH2
48.6 (31.8-74.4)
38.3 (25.4-57.7)
0.40
21
CH2N(CH3)2
27.9 (25.0-31.1)
2.3 (2.2-2.4)
2.30
22
1-Aminocyclohexyl
39.4 (25.2-61.7)
0.60
23
(CH2)3C1
1.0 (o.8-1.2)
2.30
24
(CH2)aCN
184 (167-2o4)
4.7 (4.1-5.5)
2.10
25
1-Methylvinyl
167 (111-252)
27.3 (22.1-33.7)
0.66
26
1-Cyclohexen
3295 (3068-3538)
61.3 (55.5-67.6)
0.03
27
Cyclohexyl
236 (203-274)
9.2 (7.4-11.4)
0.66
28
Ph
698 (611-798)
29
p-PhCH3
500 (421-594)
36.3 (30.7-42.8)
0.013
30
p-PhCH2CN
1603 (1522-1690)
54.8 (49.0-61.4)
0.032
31
p-PhOCH3
414 (404-425)
52.4 (34.2-80.4)
0.004
398 (363-436) 37.9 (34.1-42.o)
441 (336-578)
120 (112-128)
0.02
0.010
175 Table 3 (Continued) 32
p-PhOH
497 (412-599)
20.6 (18.6-22.7)
0.006
33
p-PhNH2
599 (522-687)
113 (lO-123)
0.007
34
p-PhCF3
2432 (2308-2563)
315 (173-575)
0.01
35
p-PhF
3312 (3078-3563)
36
p-PhCONHe
7498 (6752-8325)
37
p-PhCOCH3
38
o-PhCHO
39
81.4 (63.5-104)
0.009
1500 (1411-1596)
0.003
187 (165-213)
0.019
6864 (6489-7261)
1158 (883-1518)
0.006
m-PhCHO
1435 (1127-1826)
176 (139-221)
0.004
40
p-PhCHO
1000 (911-1097)
6.3 (5.8-6.9)
0.110
41
p-PhNOe
900 (815-993)
21.5 (21.2-29.7)
0.014
42
p-Ph(CH)2COOtBu
>10~M
743 (698-792)
0.009
43
p-Ph(CH2)2COOH
>1000
>1000
0.008
44
1-naphtyl
488 (448-530)
0.003
45
CH2Ph
46
(CHE)2Ph
47
>10 gM
4463 (4077-4885)
1.6 (1.5-1.7)
0.01
448 (373-537)
7.0 (4.4-11.1)
0.19
(CHz)aPh
209 (194-226)
1.2 (1.1-1.4)
0.35
48
2-pyridyl
114 (100-131)
89.8 (79.7-101)
0.01
49
3-pyridyl
139 (116-167)
50
4-pyridyl
428 (372-492)
51
2-furyl
310 (267-359)
52
2-thienyl
597 (536-665)
53
2-thiazolyl
54
CH2-N-imidazolyl
55
CH2-N-piperidyl
56 57
27.4 (25.8-29.1)
234 (175-311) 87.0 (67-113) 130 (118-144)
0.06 0.11 0.01
19.5 (17.5-21.7)
0.023
41.3 (37.2-45.8)
0.44
16.5 (9.1-29.8)
3.5
27.5 (22.2-34.1)
4.3 (3.2-5.8)
4.7
CH2-N-piperazyl-4-CH3
35.9 (32.3-40.0)
19.1 (15.4-23.7)
nd
CH2-N-morpholyl
90.5 (77.7-105)
27.4 (15.5-48.2)
2.6
85.4 (80.3-90.9) 178 (166-191)
58 CH2-N-thiomorpholyl 52.8 (44.1-63.3) 5.9 (4.4-7.9) 2.9 aReceptor binding affinity at A1 and A2a receptors was determined using [3H]CHA and [3H]CGS21680 as radioligands, respectively, bData are means from at least three separate experiments; 95% confidence limits in parenthesis, cplatelet aggregation was induced by ADP. The potency ratio was calculated using the concentration of the test compound close to the IC50 value. In our experimental conditions the IC50 value for NECA was 0.2 gM; data are means from three separate experiments, dpyr = 2tetrahydro(2H)pyrane [27,28].
176 Table 4 Structure-activity relationships in some alkynyl derivatives Binding assay (Ki, nM) A1 A2a
Compd.
Selectivity A1/Aza
NECA-~C-(CH2)3-CH 3
130
28
NECA-C~C~
700
45
NECA-~C-CH 2~
27
1.6
17
46
NECA-C~C-(CH2)2 ~
450
7.0
64
47
NECA-C-':-=C(CH2)3~
210
1.2
175
2.2
60
120
Table 5 Structure-activity relationships in some alkynyl and alkenyl derivatives. Binding assay (Ki, nM) A1 A2a
Compd.
59
NECA- C~C-- (CH2)3-CH3
130
NECA..____~(CH2)3-CH3
950
H...L--I.=,,H
NECA..
~H
ir'~C~ (CH213-CH3
60
NECA-(CH2)5-CH3
250
3580
2.2
72
1.6
140
Selectivity A1/A2a 60
13
160
26
177 Table 6 Structure-activity relationships in some benzyl derivatives Compd.
~CH
45
16
A2a Binding assayb (Ki, nM)
Antiaggreg. activity c (IC50, nM)
2-
1.6
1100
H-
0.9
13
1.7
810
0.5
390
Ra
17 3 ox
14
CH3-CH-CH2-~CH3
aThe whole structure of compounds is reported in Figure 2 and in Scheme 1. bReceptor binding affinity A2a receptors was determined using [3H]CGS21680 as radioligand [27]. cplatelet aggregation was induced by ADP; data are means from at least three separate experiments. The IC5o values are calculated from the potency ratio listed in Table 3, the IC50 value for NECA is 0.2 ~tM. In conclusion, these findings give further support to the hypothesis that the A2 receptor on the platelets is not a typical A2a site. From our data it is possible to draw some structure-activity relationships for the platelet receptor: -The presence of the 5'-ethylcarboxamido group improved the activity in comparison with the corresponding adenosines. -The importance of the alkynyl chain in C-2 is strictly related to the substituents on the carbon in ot to the triple bond. -The introduction of polar groups in this position markedly increased the potency. -The contemporary presence of a lipophilic side chain improved the activity. -The fully substitution of the carbon in ~ to the triple bond led to much weaker inhibitors.
178 PHPNECA As already shown, the c~-hydroxybenzyl derivative PHPNECA is the most active
agonist which inhibits platelet aggregation. Since this compound bears a chiral carbon in the side chain, the enantiomeric resolution was undertaken to assess the enantioselectivity of A2a adenosine receptors. Separation of the two diastereoisomers was analytically achieved by chiral HPLC. To obtain the two isomers on a preparative scale, the suitable R and S alkynes were alternatively coupled to iodoNECA (5) in the usual cross coupling conditions to give the desired diastereoisomers S and R PHPNECA, respectively. The enantiomeric purity was assessed by analytical chiral HPLC, and by 1H NMR techniques. The binding affinity and the functional activity of the enantiomers in comparison with those of the racemic mixture are shown in Table 7. The S-diastereoisomer proved to be more potent than the R-diastereoisomer, and than the mixture as well, in all model systems. However, the most important achievement is that the platelet receptor clearly discriminates between the two compounds since R-PHPNECA is equiactive with NECA, whereas the S diastereoisomer shows an antiaggregatory potency about 30-fold that of NECA, i.e. about 7 nM, resulting in the most potent inhibitor so far known in the adenosine agonist derivatives [30]. Table 7 Preliminary in vitro activity of diastereomeric PHPNECA compounds. Binding assays b Functional activityc Antiaggr. activity (1 Compd. a
(Ki, nM) Rat brain Rat striatum
ECs0 (nM) Rat atria Rat aorta
A1
A~
A1
R-PHPNECA
6
2.6
85
132
200
S-PHPNECA
4
0.6
52
30
7
2.5
0.9
110
123
13
(R,S)-PHPNECA
A2a
(IC50, nM) Rabbit platelets A2
aThe whole structure of compounds is reported in Figure 2. bReceptor binding affinity at A1 and A2a receptors was determined using [3H]CHA and [3H]CGS21680 as radioligands, respectively.CNegative chronotropic activity induced by cumulative addition of an agonist was studied on rat atria, and was recorded by measuring the isometric contractions; vasodilating effects were calculated on rat aorta precontracted by PGF2a (3 btM) [27] dPlatelet aggregation was induced by ADP. dData are means from at least three separate experiments.
179 Acknowledgement This work was supported by grants from MURST, CNR, and Schering-Plough S.p.A. We also thank M. Brandi, F. Lupidi, and G. Rafaiani for technical assistance. The autors are involved in the concerted action ADEURO (EU, Biomed I). REFERENCES 1 Fredholm B, Abbracchio MP, Burnstock G, Daly JW, Harden TK, Jacobson KA, LeffP, Williams M. Pharmacol Rev 1994; 46 (2): 143-213. 2 Jacobson MA. In: Belardinelli L, Pelleg A, eds. Adenosine and Adenine Nucleotides: from Molecular Biology to Integrative Physiology. Boston: Kluwer Academic Publishers, 1995; 5-13. 4 Lerman BB, Belardinelli L. Circulation 1991; 83: 1499-1509. 5 Pelleg A. Coronary Artery Dis 1993; 4: 109-115. 6 Liu GS, Thornton J, van Winkle DM, Stanley AW, Olson RA, Downey JM. Circulation 1991; 84: 350-356. 7 Shen W-K, Kurachi Y. Mayo Clin Proc 1995; 70: 274-291. 8 Hori M, Kitakaze M. Hypertension 1991; 18: 565-574. 9 Cronstein BN. J Applied Physiol 1994; 76: 5-13. 10 Linden J. Trends Pharmac Sci 1994; 15: 298-306. 11 Cristalli G, Grifantini M, Vittori S, Balduini W, Cattabeni F. Nucleosides & Nucleotides 1985; 4: 625-639. 12 Antonini I, Cristalli G, Franchetti P, Grifantini M, Martelli S, Petrelli F. J Pharm Sci 1984; 73: 366-369. 13 Cristalli G, Grifantini M, Vittori S, Klotz K-N, Lohse MJ. J Med Chem 1988; 31: 1179-1183. 14 Lohse MJ, Klotz K-N, Schwabe U, Cristalli G, Vittori S, Grifantini M. NaunynSchmiedeberg's Arch Pharmacol 1988; 337: 687-689. 15 Lohse MJ, Klotz K-N, Schwabe U, Cristalli G, Vittori S, Grifantini M. NaunynSchmiedeberg's Arch Pharmacol 1989; 340: 679-683. 16 Monopoli A, Conti A, Dionisotti S, Casati C, Camaioni E, Cristalli G, Ongini E. Arzneim-Forsch/Drug Res 1994; 44 (12): 1350-1312. 17 van Galen PJM, Stiles GL, Michaels G, Jacobson KA. Med Res Rev 1992; 12: 423-471. 18 Matsuda A, Ueda T. Nucleosides & Nucleotides 1987; 6: 15-94. 19 Bruns RF, Lu GH, Pugsley TA. Mol Pharmacol 1986; 29: 331-346.
180 20 21 22 23 24 25 26 27 28 29 30
Hutchison AJ, Webb RL, Oei HH, Ghai GR, Zimmerman MB, Williams M. J Pharmacol Exp Ther 1989; 251: 47-55. Cristalli G, Eleuteri A, Vittori S, Volpini R, Lohse MJ, Klotz K-N. J Med Chem 1992; 35: 2363-2368. Cristalli G, Vittori S, Thompson RD, Padgett WL, Shi D, Daly JW, Olsson RA. Naunyn-Schmiedeberg's Arch Pharmacol 1994; 349: 644-650. Conti A, Monopoli A, Gamba M, Borea PA, Ongini E. Naunyn-Schmiedeberg's Arch Pharmacol 1993; 348: 108-111. Dionisotti S, Zocchi C, Varani K, Borea PA, Ongini E. Naunyn-Schmiedeberg's Arch Pharmacol 1992; 346: 673-676. Sandoli D, Chiu PJ S, Chintala M, Dionisotti S, Ongini E. Eur J Pharmacol 1994; 259: 43-49. Monopoli A, Conti A, Zocchi C, Casati C, Volpini R, Cristalli G, Ongini E. Arzneim-Forsch/Drug Res 1994; 44 (12): 1296-1304. Cristalli G, Volpini R, Vittori S, Camaioni E, Monopoli A, Conti A, Dionisotti S, Zocchi C, Ongini E. J Med Chem 1994; 37: 1720-1726. Cristalli G, Camaioni E, Vittori S, Volpini R, Borea PA, Conti A, Dionisotti S, Ongini E, Monopoli A. J Med Chem 1995; 38: 1462-1472. CristaUi G, Camaioni E, Vittori S, Volpini, Dionisotti S, Ongini E, Monopoli A. J Med Chem (submitted). Cristalli G, Camaioni E, Vittori S, Volpini R, Dionisotti S, Ongini E, Monopoli A. Biorg Med Chem (submitted).
Perspective in Receptor Research
D. Giardinh, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
181
Partial agonists for adenosine receptors A.P. IJzerman, E.M van der Wenden, H.C.P.F. Roelen, R.A.A. Math& and J.K. von Frijtag Drabbe Ktinzel Division of Medicinal Chemistry, Leiden/Amsterdam Center for Drug Research, PO Box 9502, 2300RA Leiden, The Netherlands.
INTRODUCTION Adenosine is generally considered as a 'local hormone' with profound physiological activity. It is thought to mediate a large variety of effects, as diverse as vasodilation in the cardiovascular system, inhibition of lipolysis in fat cells and depression of neuronal activity in the CNS. Most of its effects are mediated by membrane-bound receptors, called Px-purinoceptors, of which currently three subclasses have been defined: A l, A: and A 3. All three classes have been cloned, and are coupled to the enzyme adenylate cyclase, A~ and A 3 adenosine receptors in an inhibitory, and A2 receptors, of which two further subtypes A2a and A2b exist, in a stimulatory fashion [1]. A great number of both A 1 and A2a selective ligands are available, both agonists and antagonists [2]. This is not yet the case for the A2b receptor. Very recently selective agonists for the A 3 receptor have been reported [3]. The ubiquity of adenosine receptors in mammalian tissues suggests a vast array of therapeutic options. For example, it has recently been found that stimulation of the Azb receptors on human blood cells leads to complete inhibition of TNF-~ release, a crucial feature in interfering with septic shock [4]. A drawback, however, to a possible therapeutic use of adenosine receptor agonists is the common hypotensive action of this class of compounds. 1000
14.0 120
T E E v G_
~ "0 Q
80 c 0
-~o
70
Q
>'6
60
-~
5O
"=
40
0
o o Q
I,U
30
,
-10
'
-g
-8
-7
-6
I
-5
log [agonist] (mol/I)
'
1
-4
J
t
-3
Figure 6. Inhibition of the electrically evoked [3H] noradrenaline release from mouse brain slices by (R)- (filled squares) and (S)-a-methylhistamine (open squares) and impentamine in the absence (open circles) or presence (closed circles) of 10 nM clobenpropit. Data were taken from Leurs et al. [39].
subtypes? Within a series of closely related histo_m~ne analogues we have thus identified several H 3 receptor ligands with different pharmacological characteristics. Small modifications of the alkylamine side chain have a great impact on the H 3 agonistic activity at the guinea-pig jejunum. I m p e n t a m i n e , with a pentyleneamine sidechain, is a highly potent H 3 antagonist at the guinea-pig jejunum. In contrast, the same compound appears to be a H 3 agonist in the rat H 3 receptor
202 and mouse brain. These data provide prelimnary evidence for the existence of p u t a t i v e H 3 receptor subtypes (Table 1). However, before a definite subclassification of the H 3 receptor can be accepted a careful investigation of the pharmacological profile of several impentamine analogues is needed. Moreover, the identification of distinct H 3 receptor genes would provide ultimate proof. Yet, sofar no molecular biological information of the H 3 receptor is available.
Table 1 S u m m a r y of the H 3 receptor activities of the H 3 agonist (R)-a-methylhistamine, the H 3 antagonist thioperamide and the new agonist/antagonist impentamine [39]. compound
guinea-pig [3H]-NA release intestine mouse brain
(R)-a-methylhistamine pD 2 = 7.8
pD 2 = 7.9
impentamine
pA2 = 8.4
pD 2 = 8.2 (a = 0.6)
thioperamide
pA2 = 8.9
pA2 = S.4
[125I]IPPbinding rat cortex PitH = 8.46 PilL = 5.92 pI~.H = 8.50 PilL = 6.60 =S.37
Acknowledgement The work of Dr Leurs has been made possible by a fellowship of the Royal Netherlands Academy of Arts and Sciences.
REFERENCES 1. 2. 3. 4. 5. 6. 7.
Schwartz JC, Arrang JM, Garbarg M, Pollard H, Ruat M. Physiol Rev 1991; 71: 1-51. Arrang JM, Garbarg M, Schwartz JC. Neurosci 1987; 23: 149-157. Van der Werf JF, Bast A, Bijloo GJ, Van der Vliet A, Timmerman H. Eur J Pharmacol 1987; 138: 199-206. Clapham J, Kflpatrick GJ. Br J Pharmacol 1992; 107: 919-923. Fink K, Schlicker E, Neise A, GSthert M. Arch Pharmacol 1990; 342:513519. Schlicker E, Fink K, Detzner M, Gothert M. J Neural Transm 1993; 93: 1-10. Schlicker E, Fink K, Hinterthaner M, GSthert M. Arch Pharmacol 1989; 340: 633-638.
203 8. Timmerman H. J Med Chem 1990; 33: 4-11. 9. Van der Werf JF, Timmerman H. Trends Pharmacol Sci 1989; 10: 159-162. 10. Barnes JC, Clapham J, Dennes RP, Kilpatrick GL, Marshall FH, O'Shaughnessy CT, Cavicchini E. Soc Neurosci Abstr 1993; 19: P1813. 11. Lin JC, Sakai K, Louvet M. C R Acad Sci 1986; 303: 469-474. 12. Lin JS, Sakai K, Jouvet M. Eur J Neurosci 1994; 6: 618-625. 13. Lin JS, Sakai K, Vannimercier G, Arrang JM, Garbarg M, Schwartz JC, Jouvet M. Brain Res 1990; 523: 325-330. 14. Smith CPS, Hunter AJ, Bennett GW. Psychopharmacology 1994; 114: 651656. 15. Yokoyama H, Onodera K, Iinuma K, Watanabe T. Eur J Pharmacol 1993; 234: 129-133. 16. Yokoyama H, Onodera K, Maeyama K, Sakurai E, Iinuma K, Leurs R, Timmerman H, Watanabe T. Eur J Pharmacol 1994; 260: 23-28. 17. Leurs R, Timmerman H. Prog Drug Res 1992; 39: 127-165. 18. Leurs R, Van der Goot H, Timmerman H. In: Testa B, ed. Advances in Drug Research, Vol. 20. London: Academic Press, 1991; 217-304. 19. Lipp R, Arrang JM, Garbarg M, Luger P, Schwartz JC, Schunack W. J Med Chem 1992; 35: 4434-4441. 20. Arrang JM, Garbarg M, Lancelot JC, Lecomte JM, Pollard H, Robba M, Schunack W, Schwartz JC. Nature 1987; 327: 117-123. 21. Leurs R, Smit MJ, Timmerman H. Pharmacol Ther 1995; 66: 413-463. 22. Arrang JM, Garbarg M, Schwartz JC. Nature 1983; 302: 832-837. 23. Arrang JM, Schwartz JC, Schunack W. Eur J Pharmacol 1985; 117: 109114. 24. Lipp R, Stark H, Schunack W. In: Schwartz JC, Haas HL, eds. The Histamine Receptor. New York: Wiley-Liss, Inc., 1992; 57-72. 25. Garbarg M, Arrang JM, Rouleau A, Ligneau X, Tuong MDT, Schwartz JC, Ganellin CR. J Pharmacol Exp Ther 1992; 263: 304-310. 26. Howson W, Parsons ME, Raval P, Swayne GTG. Bioorg Med Chem Lett 1992; 2: 77-78. 27. Van der Goot H, Schepers MJP, Sterk GJ, Timmerman H. Eur J Med Chem 1992; 27:511-517. 28. Vollinga RJ, De Koning JP, Jansen FP, Leurs R, Menge WMPB, T~mmerman H. J Med Chem 1994; 37: 332-333. 29. Arrang JM, Roy J, Morgat J, Schunack W, Schwartz JC. Eur J Pharmacol 1990; 188: 219-227. 30. Korte A, Myers J, Shih N, Egan R, Clark M. Biochem Biophys Res Commun 1990; 168: 979-986.
204 31. Kathmann M, Schlicker E, Detzner M, Timmerman H. Arch Pharmacol 1993; 348: 498-503. 32. West RE, Zweig A, Shih N, Siegel MI, Egan RW. Mol Pharmacol 1990; 38: 610-613. 33. Kilpatrick GJ, Michel AD. In: Timmerman H, Van der Goot H, eds. New Perspectives in Histamine Research. Basel: Birkh~iuser Verlag, 1991; 69-75. 34. Menge WMPB, Van der Goot H, Timmerman H, Eersels JLH, Herscheid JDM. J Labelled Comp Radiopharm 1992; 31: 781-786. 35. Jansen FP, Rademaker B, Bast A, Timmerman H. Eur J Pharmacol 1992; 217: 203-205. 36. Jansen FP, Wu TS, Voss HP, Steinbusch HWM, VoUinga RC, Rademaker B, Bast A, Timmerman H. Br J Pharmacol 1994; 113: 335-362. 37. Ligneau X, Garbarg M, Vizuette ML, Diaz J, Purand K, Stark H, Schunack W, Schwartz JC. J Pharmacol Exp Ther 1994; 271: 452-459. 38. Vollinga RC, Menge WMPB, Leurs R, Timmerman H. J Med Chem 1995; 38: 266-271. 39. Leurs R, Kathmann M, Vollinga RC, Menge WMPB, Schlicker E, Timmerman H. J Pharmacol Exp Ther 1996; in press.
Perspective in Receptor Research D. Giardin~t, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
Three-dimensional structure f r o m s p e c u l a t i o n s to facts.*
205
of G p r o t e i n - c o u p l e d r e c e p t o r s :
M. Hibert a, J. Hoflack a, S. Trumpp-Kallmeyer a, J.-L. Paquet a, R. Leppik a, B. Mouillac b, B. Chini b, C. Barberis b, S. Jard b aMarion Merrell Dow Research Institute, Strasbourg, France.
16 rue d'Ankara, 67000
bCCIPE, rue de la Cardonille, 34000 Montpellier Cedex.
INTRODUCTION G protein-coupled receptors (GPCR) represent very important targets for drug design in most therapeutic areas. More than 200 different GPCRs have been cloned and expressed over the last 8 years, allowing a better characterization of their structure and function. We review here briefly the biochemical features of GPCRs and the three-dimensional model of their complex with different ligands. The experimental validation of these models and their putative usefulness for drug design are discussed.
GPCRs : TARGETS FOR DRUG DESIGN G protein-coupled receptors belong to the same functional and structural protein family. These receptors are found in cell membrane (Figure 1). They are activated upon binding of an extracellular endogeneous ligand. This most probably induces a receptor conformational change leading to the activation of an intracellular protein, the G protein. The G protein is a heterotrimer which dissociates into alpha and beta-gamma subunits upon activation. The alpha subunit can subsequently interact with ion channels or enzymes to generate intracellular second messengers. Overall, GPCRs act as selective signal transmitters and amplifiers. GPCR endogenous ligands present a striking structural diversity (Figure 1). The challenge for the medicinal chemist is to understand how molecules as different as small cationic neorotransmitters, small cyclic or non-cyclic peptides, proteins and glycoproteins can interact with structurally related receptors.The access to a three-dimensional structure of GPCRs would represent a major step towards the understanding of receptor structure-function and ligand structure-activity relationships. Since access to an experimental 3D structure of a GPCR still remains a challenge, homology protein modelling is currently the only alternative strategy to gain some structural insight into receptor activation and function.
206 ~F
HORMONE
~kA
Io. IArach.
Second
G protein
PAF GABA
glutamate cyclic AMP pheromone M prostaglandin thromboxane
IP G
PEPTIDE
SMALL MOLECULE
adrenaline acetylcholine dopamine histamine serotonin adenosine cannabinol
Efl'ector
ACTH
angiotensin II bradyklnin CCK endothelin N formyl peplide born besin substance P substance K neuromedin neurotensin
Messengers
PROTEIN NPY
opiold oxytocin parathyroid hormone secrelln somatostatln vasopressin
VIP
TRH thrombln
C5A chemotaclic ca,atonln glucagon
SENSORY
olfactory opsins
IL--8
melanotrop|n
TSH FSH LH
Figure 1 9G protein-coupled receptors and some of their ligands.
T H R E E - D I M E N S I O N A L M O D E L S OF G P C R s Structural information derived from sequence analysis, chemical labelling, fluorescence, circular dichroism, accessibility to enzymes and antibodies, ligand structure-activity relationship, chimeric receptors, site-directed mutagenesis, etc.. has been taken into consideration to propose generic 2D and 3D models of GPCRs. In a first step, such studies had led to the approximate localisation of the ligand binding site (transmembrane region) and of the G protein coupling domain [1,2]. The next step consisted in proposing a more detailed 3D model. This has been achieved as follows [3,4]. The reference protein chosen is bacteriorhodopsin. This protein shows no sequence or functional homology with GPCRs. However, it presents an activation s y s t e m very similar to mammalian opsins (which are GPCR members) and contains seven transmembrane, a-helical domains as postulated for GPCRs [5]. A low resolution (10 A) crystallographic structure of bacteriorhodopsin [5] has been used as a scaffold to position the seven ahelical domains of GPCR relative to each other. For a given receptor, each of the seven hydrophobic stretches of aminoacids was built as an a-helix with the SYBYL programme and energy minimized with AMBER [6] and TRIPOS [7] force fields. The seven helices have then been assembled using bacteriorhodopsin as a template. The relative orientation of helices fulfills three independent criteria : the helix hydrophobic surface points to the outside, towards the hydrophobic membrane environment ; the most conserved residues are found in the core
n
Figure 2 : Three-dimensional model of acetylcholine in the muscarinic m l receptor.
Figure 3 : Three-dimensional model of vasopressin in the VIA vasopressin receptor.
208 of the protein ; the ligand binding residues converge to the inside of the protein. Loops were built with the loop search programme in SYBYL. The whole structure has then been extensively energy minimized without or with ligands. More recently, the model has been refined (eg Figures 2 and 3) in order to obtain a perfect match between the theoretical projection map of the model and the experimental projection map of bovine opsin, the first crystallographic data related to a GPCR [8]. The optimised model differs from the original one by the relative tilt of transmembrane a-helices. However the two models are qualitatively similar. These models led to the proposal of a number of novel hypotheses : 9The activation site of GPCR appears to be buried in the structure at about 15 .~ from the extracellular surface of the receptor. 9 The putative binding sites of several important neurotransmitters have been described [3,4] (Figures 4-6). For instance, residues likely to interact directly with dopamine, adrenaline, serotonin, histamine, acetylcholine, adenosine, substance P, TSH, etc.., have been identified [3,4,9-11]. 9All endogenous ligands (small or huge, cationic or not, peptidic or not) can interact with a homologous region of the receptor transmembrane domain. 9 A number of hypotheses regarding the receptor activation mechanisms and its dynamics have been proposed [3,4]. The proposed model was in good qualitative agreement with the existing experimental results. However, the next crucial step was to further support this model with new experiments and to examine their predictive value.
FROM SPECUI~TION
TO FACTS: E X P E R I M E N T A L V A L I D A T I O N
Four years after the first disclosure of GPCR 3D models, it is interesting and necessary to examine their relevance and level of predictability in the light of more recent experimental results. Briefly : 9 The predicted backbone architecture is in very good agreement with the first crystallography data on a GPCR (bovine opsin) published recently [8]. GPCR have indeed seven alpha-helical transmembrane domains, bundled very similarly to the bacteriorhodopsin transmembrane domain. 9 The depth of the binding site of small cationic ligands is at least 11 .~ as predicted and as demonstrated by fluorescence quenching experiments [12]. 9 The binding site of a number of neurotransmitters has been probed by site directed mutagenesis. More than 100 mutants have been studied [13]. The observed shifts in ligand affinity are in most cases in good qualitative agreement with the models. 9 Structure-activity relationships are in good agreement with the 3D models and have been better understood [14-18]. Although one should be cautious in interpreting binding affinity shifts in terms of direct ligand-protein interaction changes, site directed mutagenesis remains the easiest way to probe a model. We have u n d e r t a k e n to probe the proposed models of GPCR in exchanging systematically residues predicted to be responsible for ligand binding [1922]. We decided to probe two classes of GPCRs: receptors of cationic
209
~'
/
#/" : i' ' ' P~',3~ Phe61:'
B
I I
9
Trp3o~
/
,\
h
Serso5 I As I "" " L~,' I[" ' OH P3" I HO IN"" Lr HO s~,o~--',~." 5 r
1/
-:: \
4
3
; 1i"'17
"HOWl S,ersos"',,,~k-" I/ Phe5~~~ ~ - - - ~ ] 5
6
I
O NH2
Lr
Pr~
4
D ,/
Se r~o~......
"~, Ho]
I
Phe617
I
Phe616
'
Pr~ TrP6'3
9
3
6
I TrP307
Phe617
\
I ffp.~,,
Tyr6~ 6
I t -i-. ~.
Phesog~Alas~ 5
4
3
Pr~
TT,~,,~ 6
Alasas ~ Phe5o9T ~ x ~
N---El/_ H'--.;er4o6I
5
4
3
Pro615 TrP613 6
Figure 4 : S c h e m a t i c r e p r e s e n t a t i o n of predicted n e u r o t r a n s m i t t e r binding sites : A : d o p a m i n e ; B : a d r e n a l i n e ; C : acetylcholine ; D : serotonin. Helix axes are r e p r e s e n t e d as v e r t i c a l lines and n u m b e r e d 1 to 7. R e s i d u e s s u r r o u n d i n g t h e n e u r o t r a n s m i t t e r are s c h e m a t i c a l l y d i s p l a y e d a n d n u m b e r e d in a coded m a n n e r : the first digit indicates the t r a n s m e m b r a n e domain an the next two digits correspond to the rank of the residue in this domain (reproduced w i t h the a u t h o r i s a t i o n of the authors and publishers, from Medecine/Science 1993, 9, 31-40.).
210 neurotransmitters and receptors of neuropeptides. Hence, the binding of acetylcholine to the human m l muscarinic receptor has been studied. A schematic model of the acetylcholine binding site had been proposed (Figure 2 and 4). Most amino acids in the putative direct neighbourhood of the ligand have been mutated. The binding affinity of a number of agonists, partial agonists and antagonists for these mutants has been measured. The binding shifts observed have already been reported [19] and suggest comments important for structure-activity relationship understanding and drug design. In brief: 9 Agonist, partial agonist and antagonist binding is generally affected by mutations around this region of the receptor while it is not by similar mutations in other regions. This indicates that the considered cholinergic ligands bind indeed to the postulated binding domain. 9 Highly homologous receptor subtypes (ml, m2, m3; more than 95 % similarity) interact in a different way with the same ligand. These results suggest that in spite of their sequence similarity and binding site identity, these receptor subtypes differ in some way : in their extracellular loops, in their folding, in the packing of helices or, most probably, in their dynamics. This result also highlights the limits of homology protein modelling if one wants to account for subtle receptorligand interactions. 9 The same receptor residue contributes to a different extent to the binding affinity of ligands binding to the same receptor region. 9 Compounds with highly similar structures bind differently to the same receptor. This observation raises the issue of the relevance of any structureactivity relationship study based on ligand comparison. 9 Taken together, these experimental results demonstrate that the ligand binding sites and the residues likely to be involved in the recognition process had been correctly identified from the model. However, as anticipated [23], the model could not account for the subtle differences in interaction reported above. Similarly, a three-dimensional model of the vasopressin/oxytocin receptor has been defined and the binding mode of the neuropeptides has been analysed (Figure 3 and 5). Residues likely to contribute to the affinity, efficacy and selectivity have been proposed from the model. These qualitative predictions have subsequently been confirmed by site directed mutagenesis [20-22]. A number of general comments can be made : 9A large cyclic neuropeptide can indeed penetrate the core transmembrane domain of its receptor. 9The bottom of the binding pocket is in a position equivalent to the binding site of small cationic neurotransmitters (dopamine, adrenaline, serotonin, acetylcholine,etc.) and to the ionone ring of retinal in opsins. 9 Peptidic and non-peptidic antagonists binding is not affected by the mutations which led to a decrease in agonist affinity. 9The selectivity of vasopressin for its receptor is due to a single interaction between the side-chain of Arg8 on the ligand and T y r l l 5 on the receptor. 9 Primary sequence analysis of neuropeptide receptors and SAR study of their ligands suggest t h a t the residue in the first extracellular loop corresponding to Tyr115 of the vasopressin receptor might contribute to binding affinity and selectivity. This would seem to be the case notably for angiotensin, cholecystokinin, neuropeptide Y and neurokinin receptors [22].
211 YII5 L
60
5~
K3eSA
.. N"2 .
Q 620A
~ o
10
I
I
Qt~A
H6
o
~
o%
i
L'/
"s.2 ......
.~ ,~~ .._
" - . ? - - - T..~
)=;>
I
H4
~/~" "~, A
~
~ ~,~-o
-..
"~
//-'~
Q 218A
3OO
"
H2 N
.,'
o~._.__ -
Q 2'1,i A
6
.~/,'--"
H3
H2
Figure 5 : S c h e m a t i c r e p r e s e n t a t i o n of vasopressin binding to the V1A receptor. Dotted lines indicate the postulated interactions between the ligand and the receptor residues located in the t r a n s m e m b r a n e domain of the receptor. The residue n u m b e r i n g is coded as in Figure 4, with the exception of Tyr115 which corresponds to the official VIA receptor label. The site directed m u t a t i o n and the decrease (x fold) in affinity of vasopressin for this m u t a n t are indicated near each residue.
Figure 6 9Similarity in the binding mode of GPCR ligands, as predicted from the model [9]. Reproduced from reference [9] with permission of the authors and publishers.
212 QUALITATIVE VERSUS QUANTITATIVE VALUE OF THE MODEL The experimental results reported and discussed above and many others not reported here indicate that GPCR 3D models are QUALITATIVELY excellent. We moved from the "Lock and Key" model to an atomic representation which led to novel working hypotheses. Many of these hypotheses have a posteriori received support from experiments. However, as feared [23], these models have in general little, if any, QUANTITATIVE predictive value. It seems indeed unlikely t h a t the site directed mutagenesis results reported above could have been predicted from the model. In order to avoid to expect too much from these models, it is important to understand how complex GPCRs are, compared to soluble enzymes for instance. This complexity is illustrated in Figure 7. We are in fact dealing with a multimolecular system and many inter-dependent equilibria : ligandreceptor ; waterreceptor ; ligandwater ;resting r e c e p t o r < - > a c t i v a t e d receptor ; receptorG protein ; receptor i n t r a c e l l u l a r allosteric cation ; r e c e p t o r < - > p h o s p h o r y l a s e ; G proteinGDP ; alpha subunitbeta-gamma subunit ; etc. Additional parameters such as the nature of the phospholipidic constituents of the membrane, receptor concentration, G protein nature and concentration, acidity of the extracellular membrane surface can modulate the measured ligand binding affinity. Finally, the intrinsic dynamics of the receptor is most probably determinant for ligand binding and function. The hurdle thus appears very high for those aiming at predicting quantitatively subtle molecular interactions in these systems.
9 9
Ligand
H20 Sialic acid
/\
,
,,
Ro (~
(,_jb Kinase
,o p Y
--'~ ' %
G protein
e
cation
A
[1
GTP
GDP
Figure 7 : Schematic representation of the molecular equilibria involved in G protein-coupled receptor activation ( reproduced with permission from publishers and authors of Hibert M, et al. Sanz F, ed. Trends in QSAR and Molecular Modelling '94, Barcelona : Prous Sciences, 1995).
213 CONCLUSION For one century, medicinal chemists tried to design ligands for receptors without knowing how these receptors look like. With the recent progress in molecular biology, molecular pharmacology, biophysics, SAR, a n d theoretical chemistry, it became possible to propose a threedimensional representation of GPCRs and to predict qualitatively the binding mode of very structurally different ligands. Originally based on very w e a k hypotheses, these models could progressively be refined and have received support from experimental data. In fact, they proved to be a very v a l u a b l e tool to g e n e r a t e novel h y p o t h e s e s on receptor functional architecture and led to conceive and perform new interesting experiments. The next important milestone will probably be the access to a high resolution experimental structure of GPCRs. However, these static pictures might not even be sufficient to provide scientists with definitive answers on receptor-ligand interaction since recent studies have already demonstrated the importance of the kinetic parameters for the neurotransmission process and the extraordinary complexity of this system.
REFERENCES
1 Lefkowitz EJ, Caron MG. J Biol Chem 1988; 263: 4993-4996. 2 Dixon RAF, Strader CD. Annu Rep Med Chem 1988; 23: 221-223. 3 Hibert MF, Trumpp-Kallmeyer S, Bruinvels A, Hoflack J. Mol Pharmacol 1991; 40: 8-15. 4 Trumpp-Kallmeyer S, Hoflack J, Bruinvels A, Hibert M. J Med Chem 1992; 35: 3448-3462. 5 Henderson R, Baldwin JM, Ceska TA, Zemlin F, Beckmann E, Downing KH. J Mol Biol 1990 ; 213: 899-929. 6. Weiner SJ, Kollman PA. J Comput Chem 1986; 7: 230-252. 7 Sybyl Molecular Modelling Software, Tripos Associates inc., St Louis MO, USA 8 Schertler GFX, Villa C, Henderson R. Nature 1993; 362: 770-772. 9 Hoflack J, Trumpp-Kallmeyer S, Hibert M. In: Kubinyi H, eds. 3D QSAR and Drug Design. Theory, Methods and Applications. Leiden: ESCOM, 1993; 355-372 10 Hoflack J, Hibert MF, Trumpp-Kallmeyer S. Drug Des Discov 1993; 10: 157-171. 11 Trumpp-Kallmeyer S, Hoflack J, Hibert M. In: Buck SH, eds. The Tachykinin Receptors. New York: Humana Press, 1993; 237-255. 12 Tota M, Candelore M, Dixon RAF, Strader C. Trends Pharmacol. Sci. 1991; 12: 4-6. 13 Savarese TM, Fraser CM. Biochem J 1992; 283: 1-19. 14 Lewell XQ.Drug Des Discov 1992; 9: 29-48.
214 15 IJzerman AP, Van Galen PJ, Jacobson KA. Drug Des Discov 1992" 9: 49-67. 16 Nordvall G, Hacksell UJ. J Med Chem 1993; 36: 967-976. 17 Fanelli F, Menziani M, Carotti A, De Benedetti. Bioorg Med Chem 1994; 2: 195211. 18 Yamamoto Y, Kamiya K, Terao S. J Med Chem 1993; 36: 820-825. 19 Hibert M, Hoflack J, Trumpp-Kallmeyer S, Paquet J'L, Leppik R, Barberis C, Mouillac B, Chini B, Jard S. Eur J Med Chem 1995; 30:189-199. 20 Chini B, Mouillac B, Ala Y, Balestre MN, Trumpp-Kallmeyer S, Hoflack J, Elands J, Hibert M, Manning M, Jard S, Barberis C. EMBO J 1995; 14: 2176-2182. 21 Mouillac B, Chini B, Balestre MN, Elands J, Trumpp-Kallmeyer S, Hoflack J, Hibert M, Jard S, Barberis C. J Biol Chem 1995; 270: 25771-25777.. 22 Trumpp-Kallmeyer S, Chini B, Mouillac B, Barberis C, Hoflack J, Hibert M. Pharm Acta Helvetiae 1995; 70: 255-262. 23 Hibert MF, Trumpp-Kallmeyer S, Hoflack J, Bruinvels A. Trends Pharmacol Sci 1993; 14: 7-12. *This article is adapted and incremented from reference [19].
Perspective in Receptor Research D. Giardinb., A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved. ION
CHANNELS
David J. School 14260
AS
TARGETS
FOR
DRUG
215 DESIGN
Triggle
of Pharmacy,
State University
of New York,
Buffalo,
NY
Abstract
Ion channels are a heterogeneous group of p h a r m a c o l o g i c receptors that respond to a variety of i n f o r m a t i o n a l inputs by controlling in rapid and specific fashion the m o v e m e n t s of ions across cell membranes. Under p a t h o l o g i c conditions the disorderly movements of ions may c o n t r i b u t e to cell pathology and death. Drugs that regulate ion channel function in both voltage- and l i g a n d - g a t e d c a t e g o r i e s represent both molecular tools w i t h w h i c h to dissect channel function and therapeutic agents for, inter alia, c a r d i o v a s c u l a r and neuronal disorders. These aspects are well i l l u s t r a t e d by the Ca 2+ channel antagonist class of drugs. GENERAL
PROPERTIES
OF
ION CHANNELS
Introduction Ion channels are one group of p h a r m a c o l o g i c receptors. As such ion channels: I. Exist as homologous protein families 2. Possess specific drug binding sites which define s t r u c t u r e - a c t i v i t y relationships for both activators and antagonists 3. Have binding sites for both endogenous and exogenous ligands 4. May be regulated by homologous and h e t e r o l o g o u s influences including disease states. Ion channels are one important cell signalling process by w h i c h e x c i t a b l e cells respond to a variety of informational signals i n c l u d i n g neurotransmitters, hormones and sensory inputs [ 1 ] .
The cell maintains an asymmetric distribution of ions t h r o u g h the operation of a variety of pumps and p e r m e a b i l i t y barriers. The net result is the m a i n t e n a n c e of a m e m b r a n e p o t e n t i a l that is controlled by the selective and c o o r d i n a t e d o p e n i n g and closing of discrete ion channels [ Figure 1 ]. Quite generally, the selective opening of an ion channel will drive the m e m b r a n e potential towards the e q u i l i b r i u m p o t e n t i a l for that ion. Thus, the opening of Na § and Ca 2§ channels or the c l o s i n g of K § channels will depolarize the cell and m e d i a t e e x c i t a t i o n and, the opening of K § and Cl-channels will move the resting membrane potential in a h y p e r p o l a r i z i n g d i r e c t i o n to m e d i a t e inhibitory responses. These c o n s i d e r a t i o n s underlie the actions of drugs that may act s e l e c t i v e l y at one or other channel class. It should be e m p h a s i z e d that under physiological conditions ion channels c o o p e r a t e to regulate membrane potential and i n t r a c e l l u l a r ionic c o n c e n t r a t i o n s . Thus, the cardiac action potential is made up of a dozen or more ionic conductances functioning in h i g h l y c o o r d i n a t e d fashion.
216
Antagonists
K.=--90mV
Ec~§=+120mY
ENa+=+50mV~~
[K+in,] = - 1 5 5 m M
I'Ca2§]=--10-4mM [Na+j =-12mM Ca2§ L
0
intJ
~// Na+O
Agonists
Figure I. Ionic concentrations and equilibrium potentials Na +, K + and Ca 2+ and the effects of drugs [ a n t a g o n i s t / a g o n i s t s ] that regulate the ion channels.
IT T Lipid bilayer
TTT
for
Remote Sensor
G Pr
Intermediate
Figure 2. A schematic representation of ion channel structure depicting the principal components that determine channel function.
217 D y s r e g u l a t i o n of one or more of these conductances may p e r t u r b c r i t i c a l l y this coordination leading to a variety of arrhythmic situations. The
S t r u c t u r e a n d C l a s s i f i c a t i o n of Ion C h a n n e l s The schematic r e p r e s e n t a t i o n of Figure 2 depicts ion channels as m e m b r a n e p r o t e i n s that possess regulatory sensors, both e x t r a c e l l u l a r and intracellular, that respond to i n f o r m a t i o n a l inputs, gates that open and close and a selectivity filter a s s o c i a t e d w i t h the channel pore and which controls ion permeation. The sensors may be components of the channel protein[s] itself or may be remote and linked to channel function by c y t o s o l i c or membrane signalling devices. The u n d e r l y i n g m o l e c u l a r bases of channel structures are i n c r e a s i n g l y well defined through biochemical and m o l e c u l a r biology studies [ 2,3 ]. Ion channels may be c l a s s i f i e d by several criteria including the nature of the permeant ion, the e l e c t r o p h y s i o l o g i c a l characteristics including c o n d u c t a n c e and opening and closing rates, the principal informational input as a ligand- or v o l t a g e - g a t e d channel and the p h a r m a c o l o g i c a l s e n s i t i v i t y of the channel [ Figure 3 ]. The latter c l a s s i f i c a t i o n is of particular importance to issues of drug development. Ion Channels
As
Targets
For D r u g s
As m a j o r
classes
of
p h a r m a c o l o g i c a l receptors ion channels are automatic loci for drug action. However, ion channels are p a r t i c u l a r l y a t t r a c t i v e targets for several additional reasons [ 4 ] : i. They are loci for integrated cellular c o m m u n i c a t i o n 2. They are extremely rapid and efficient signalling devices 3. They exist in several major classes and subclasses 4. They are multiple pharmacologic receptors, each channel typically possessing several specific drug b i n d i n g sites 5. The p r o p e r t i e s of drug binding are often m o d u l a t e d by state-dependent interactions whereby the a f f i n i t y or access of a drug for its binding site is d e t e r m i n e d by the state - open, closed or i n a c t i v a t e d - of the channel. Collectively, these properties define o p p o r t u n i t i e s for m u l t i p l e control mechanisms of a single physiological or p a t h o l o g i c a l process. Thus, pathologies that require n e u r o p r o t e c t i v e agents may find pharmacological s a t i s f a c t i o n in at least four discrete groups of agents - e x c i t a t o r y amino acid antagonists, Ca 2§ channel antagonists, Na + channel a n t a g o n i s t s and activators of ATP-dependent K § channels [ Figure 4 ]. Additionally, each channel typically p o s s e s s e s several d i f f e r e n t drug binding sites or receptors that amplify c o n s i d e r a b l y the potential for pharmacologic control [ Figure 5 ]. These o p p o r t u n i t i e s have been explored and i n c r e a s i n g l y realized w i t h several ion channel types including voltagegated Ca 2+ channels [ 5 ].
218
O"
-"
H O ' / ~ " ~ ~ ' / ~ ~ NH > I~H2 OH OH
H2Nco/O~3 H
H21~"~Y ~HN" ~--NH
/10 12~ OH 11 OH
a
+
NEt 4
b
~NO2 MeOOC~ COOMe d
F i g u r e 3. D r u g s t h a t s e l e c t i v e l y i n t e r a c t w i t h ion c h a n n e l s a,b - tetrodotoxin a n d s a x i t o x i n for Na § c h a n n e l s ; c tetraethylammonium for K § c h a n n e l s ; d - n i f e d i p i n e for Ca 2+ channels.
EAA Antagonists I NMDAJ AMPA I
Ca 2+ Antagonists
Phenytoin BW 1003C87
Cromakalin Pinacidil
Na + Antagonists
K+ATPActivators
CNS 1102 NBQX Remacemide GYKI52466 HA966
F i g u r e 4. The c l a s s e s of ion c h a n n e l in n e u r o p r o t e c t i o n strategies.
Nimodipine Conotoxins
drugs
that
are
effective
219
CGP 40116 LY 233053
6.5 mmol/L were compared with those from a placebo group, where all the lipid parameters measured (see Table 1) increased over the 8 week period of administration. Encouragingly, in patients treated with levcromakalim 2, significant reductions in total cholesterol, LDL and triglycerides were observed and the HDL:total cholesterol ratio, which is a good indicator of atherogenic potential, was improved. These data are indicative of a potential for improvement of two risk factors in hypertensive patients with hypercholesterolemia, not generally observed with other antihypertensive agents. Table 1 Effects of levcromakalim 2 or placebo (once daily oral doses for up to 8 weeks) on plasma lipid profile (% change) in patients with baseline total cholesterol > 6.5mmoU1
Total Cholesterol HDL LDL Triglycerides HDL/cholesterol * p P5-NH2 > P5 > P7. In contrast in the rat gastric longitudinal muscle contraction assay (reflecting the gastric smooth muscle receptor), the potency series for the same four peptides was: P7-NH2 > P5-NHa > P7 > P5 [103]. In a similar fashion, the potency series for a human placental artery contractile preparation was: P7-NH2 > P7 > P5-NH2 > P5, whereas in the human umbilical vein contraction assay, the potency order was P5-NH2 >> P7-NH2 = P7 >> P5; and these two vascular potency series differed in turn from the one observed for a human platelet aggregation assay: P5-NH2 > P7-NH2 > P7 >> P5 [ 109]. Taken together, these distinct orders of agonist potencies for the same set of agonists in the different assay systems, as summarized in Table 3, point to distinct
Table 3 Relative potencies of TRAPs in different assay systemsa
Tissue
Response
Potency series (P7-NH2, P7, P5-NH2, P5)
Rat aorta Rat gastric LM Human placental artery Human umbilical vein Human platelets
relaxation contraction contraction contraction aggregation
P7-NH2 > P5-NH2 > P5 _>P7 P7-NH2 > P5-NH2 > P7 > P5 P7-NH2 > P7 > P5-NH2 > P5 P5-NH2 >> P7-NH2 = P7 >> P5 P5-NH2 > P7-NH2 > P7 > P5
a
Data from Hollenberg et al. [ 103,109]
functional receptor subtypes, according to classical criteria [ 110]. The question to be asked, however is: do the functionally distinct receptor subtypes characterized by different structure-activity profiles correspond to different thrombin receptor sequences in the different tissues? To deal with this question, we used a reverse-transcriptase/polymerase chain reaction (RT-PCR) approach to analyze thrombin receptor mRNA present in human placental artery and umbilical vein tissues, which exhibited very distinct structure-activity profiles (Table 3). Somewhat to our surprise, the two tissues yielded exactly the same cDNA sequence for the thrombin receptor. Our finding of the same thrombin receptor mRNA in tissues that possess pharmacologically distinct receptors according to their structure-activity profiles may be rationalized by a number of possibilities. First, it may be that distinct posttranslational processing of the product of the same receptor mRNA in different tissues may yield functionally distinct receptor subtypes. Such a situation appears to be the case for the
283 murine bradykinin receptor expressed by transfection in COS cells [111]. A second possibility is that the expression in different tissues of markedly different types and amounts of the G-protein, to which the thrombin receptor couples could, in theory, alter relative agonist potency, as has been discussed by Kenakin and Morgan [ 112]. A third alternative to consider is that the TRAPs, such as P5-NH2, in addition to activating the thrombin receptor, might also be able to activate the recently cloned protease-activated receptor, PAR-2 [113,114], which could coexist along with the thrombin receptor in a number of tissues. Further work willbe required to evaluate these several possibilities. From a practical point of view, nonetheless, the distinct structure-activity profiles we have observed in different assay systems suggest that it might be possible to design selective agonists or antagonists for specific tissue targets. In this regard it can be pointed out that TRAPs, such as P5-NH2, which are relatively potent in activating the thrombin receptor in rat gastric and vascular preparations [98,99,103] are not able to activate rat platelets, which are otherwise stimulated by thrombin via an as-yet uncharacterized thrombin receptor [ 115]. Similarly, the TRAP, P5, has been reported to regulate canine coronary artery contractility, but is unable to aggregate canine platelets that are otherwise responsive to thrombin [ 116]. Such absolute selectivity for the TRAPs between vascular and platelet assay systems has not been observed for human tissue samples.
Thrombin receptor antagonists Given the pathophysiologic importance of thrombin and its receptor, as outlined above, it is no surprise that considerable effort has been aimed at developing thrombin receptor antagonists. To date however, only a few peptide antagonists have been described, primarily resulting from a human platelet assay screen of peptides based on the TRAP, P 14, sequence. One of these peptides, YFLLRNP, can antagonize the aggregation of human platelets stimulated by low concentrations of ct-thrombin or the TRAP, P7; however, this peptide was a partial agonist in the platelet assay [117]. An alternative novel series of antagonists based on the TRAP motif, beginning with mercaptopropionic acid (Mpr) instead of serine and having the two leucine side chains replaced with a cyclohexylalanyl residue (Cha), (Mpr F Cha Cha R...) have been observed to inhibit both thrombin and P5-induced platelet aggregation, with ICs0s ranging from about 5 to 80 ~tM [118,119]. One of these Mpr analogues (Mpr F Cha Cha RKPNKD-NH2 or Mpr-P 10-NH2), exhibiting an IC50 of 5 ~tM in a thrombin-induced human platelet aggregation assay [ 118] has also been reported to inhibit the relaxant and contractile effects of thrombin and P7 in porcine coronary artery ring preparations [120]. Nonetheless, the data reported by Tesfamariam [120] would indicate that in the porcine coronary preparations Mpr-P10-NH2 was a desensitizing partial agonist. A peptide based on the P5 sequence, SFLVR-NH2 (or VR-NH2) has also been reported to act as a TRAP antagonist in a coronary artery preparation, shifting the concentration-effect curve for P5 to the right [121]. Another intriguing independent method for the discovery of thrombin receptor antagonists has used a phage display approach, coupled with a platelet binding recovery procedure to identify the sequence, MSRPACPNDKYE, as an inhibitor of TRAP- and thrombin-induced platelet aggregation [ 122]. In our own work, we wished to evaluate the actions of the putative thrombin receptor antagonist, Mpr-P 10-NH2 in the rat gastric longitudinal muscle contraction assay; and we wished to assess the actions of VR-NH2 both in the gastric contractile assay and in the platelet aggregation assay. As indicated by the data in Figure 7, Mpr-P10-NH2 which was an
284
RAT STOMACH L M 140
...I 0 ~-120
& 9P5-NHz Mpr-PIO-NH
l
z
ElO0 o 143 80
z O 60 D I-O .~ 40 I--z 20 O O o
9
!
0.1
,
i
1
,
10
i
|
100
CONCENTRATION
Figure 7. Contractile activity of MprP10-NH2 compared with P5-NH2 in the rat gastric LM assay. The contractile activities of Mpr-P10-NH2 (&) and PSNH2 (A) in the rat gastric longitudinal muscle strip were expressed as a percentage (% 50 mM KC1) of the contractile response to 50 mM KC1.
IOO0
(pM)
antagonist in the platelet aggregation assay, was a relatively potent contractile agonist in the rat gastric LM assay, with an activity comparable to that of P5-NH2. In the rat aorta relaxation assay, Mpr-P 10-NH2 was a weak agonist, without antagonist activity (not shown). In contrast, VR-NH2, which was a desensitizing partial agonist in the canine coronary assay, was a full agonist in the platelet assay and was a non-desensitizing full agonist in the rat gastric LM assay (Figure 8). Thus, the main findings of our work with the peptides reported to be thrombin receptor antagonists were that the observation of antagonist activity depends heavily on the type of assay system used for the evaluation. Clearly, in the search for thrombin receptor antagonists, it will be necessary to use a matrix of bioassay systems to evaluate in depth the complete pharmacologic properties of any lead compound.
120 ~. ~---100 ~
120
P5-NH 2 (&.A} VR-NH z (O,e)
~00
80
8O
x 3;
~ 8o
60
~ ,o
40 ~
:~ 20
20
O
Z O u,l
0
0
0.1
1
10
CONCENTRATION
100 (pM}
1000
O
Figure 8. Agonist activity of SFLVRNH2 in the rat gastric LM and human platelet assays. The contractile (relative to 50 mM KC1) and platelet aggregation (% max) activities of PS-NH2 (A, &) and VR-NH2 (0, O) were measured using rat gastric longitudinal muscle (open symbols) and washed human platelets (solid symbols) essentially as described elsewhere [103,28].
285 ACKNOWLEDGEMENTS
Work reported in this article was supported in part by a Medical Research Council of Canada University-Industry grant awarded to M.D.H. in conjunction with BioChem Therapeutic Inc. The authors are grateful to the staff of the Pharmaceutical sector of the Biotechnology Research Institute of Montreal (NRC) without whom part of the work described in this article would not have been possible. We are also indebted to Drs. Ren6 Martel and Arshad Siddiqui as well as to Annie St-Pierre, C~line Locas, Micheline Tarazi and Nicole Landry for expert assistance. We are also grateful to Mrs. Sylvie Ouellet for secretarial help. ABBREVIATIONS USED
TRAP, thrombin receptor-activating peptide; P14, SFLLRNPNDKYEPF; P7, SFLLRNP; P7-NH2, SFLLRNP-NH2; P5, SFLLR; P5-NH2, SFLLR-NH2; P5VR-NH2, SFLVR-NH2; Cha, cyclohexylalanyl; Mpr, mercaptopropionyl; LM, gastric longitudinal muscle. REFERENCES
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21
Kaplan NM. J Cardiovasc Pharmacol 1982; 4: S 187-S 189. Ker6nyi T, Schneider F. In: Machovich R, ed. Blood Vessel Wall and Thrombosis. Boca Raton: CRC Press, 1988; vol. II: 51-69. Shepard LY. Trends Biotechnol 1991; 11: 80-85 Cook NS, Ubben D. Trends Pharmacol Sci 1990; 11: 444-451. SharfRE, Harker LA. Blut 1987; 55: 131-144. Furie B, Furie BC. Cell 1988; 53: 505-518; Altieri DC. Blood 1993; 81: 569-579. Mann KG, Jenny ILl, Krishnaswamy S. Ann Rev Biochem 1988; 57: 915-956. Machovic R. In: Machovich R, ed. Blood vessel wall and Thrombosis. Boca Raton: CRC Press, 1988; Vol. I: 128-140. Andrews RK, Fox JEB. Curr Opinion Cell Biol 1990; 2:894-901. Kunicki TJ. Blut 1989; 59: 30-34. Jonasson L, Holm J, Skalli O, Bondjers G, et al. Arteriosclerosis 1986; 6:131. Schwartz SM, Gajdusek CM. Prog Thromb Hemost 1983; 6:85-112. Fitzgerald DJ, Fitzgerald GA. Proc Natl Acad Sci USA 1989; 86: 7585-7589. Gast A, Tschopp TB, Baumgartner HR. Arterioscler Thromb 1994; 14: 1466-1474. Fenton JW. Ann NY Acad Sci 1981; 370: 468-485; Magnusson S. In: Boyer PD, ed. Enzymes. New York: Academic Press, 1971; 277-321. Fenton JW, Landis BH, Walz DA, Bing DH, et al. In: Bing DH, ed. The Chemistry and Physiology of Human Plasma proteins, Oxford: Pergamon Press, 1978; 151-183. Chang JY. Biochem J 1986; 240: 787-802. Charles S, Greenberg K, Achyuthan KE, Hiraglia CC et al. Ann NY Acad Sci 1986; 485: 140-143. Hernker HC, Beguin S. In: Jolles G, Legrand Y, Nurden A, eds. London: Academic Press, 1986; 219-226.
286 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54
Phillips DR, Charo IF, Parise LV, Fitzgerald LA. Blood 1988; 71" 831-843. Hynes RO. Cell 1987; 48: 549-554. Nesheim ME, Hibbard LS, Tracy PB. In: Mann KG, Taylor FB, eds. The regulation of coagulation, New York: Elsevier-North Holland, 1980; 145-159. Nawroth PP, Kisiel W, Stem DM. Clin Haematol 1985; 14: 531-546. KroU MH, Schafer AI. Blood 1989; 74:1181-1195. Vu TKH, Hung DT, Wheaton FI, Coughlin SR. Cell 1991; 64:1057-1068. Bahou WF, Coller BS, Potter CL, Norton KJ, et al. J Clin Invest 1993; 91: 14051413. Rasmussen UB, Vouret-Craviari V, Jallat S, Schlesinger Y, et al. FEBS Lett 1991; 288: 123-128. Hung DT, Vu TKH, Nelken NA, Coughlin SR. J Cell Biol 1992; 116: 827-832. Zhong C, Hayzer DJ, Corson MA, Runger MS. J Biol Chem 1992; 267: 1697516979. Howells GL, Macey M, Curtis MA, Stone SR. Brit J Haematol 1993; 84:156-160. Dihanich M, Kaser M, Reinhard E, Cunningham D, et al. Neuron 1991; 6: 575581. Deschepper CF, Bigomia V, Berens ME, Lapointe MC. Mol Brain Res 1991; 11: 355-358. Weinstein JR, Gold SJ, Cunningham DS, Gall CM. J Neurosci 1995; 15" 29062919. Jalink K, Moolenar WH. J Cell Biol 1992; 118:411-419. Suiclan HS, Stone SR, Hemmings BA, Monard D. Neuron 1992; 8: 363-375. Smith GF, Neubauer BL, Soundboom JL, Best KL, et al. Thrombosis Res 1988; 50:163-174. Honn KV, Tang DG, Chen YQ. Sem Thromb Haemostas 1992; 18:392-415. Nierodzik ML, Plotldn A, Kajumo F. J Clin Invest 1991; 87: 229-236. Klepfish A, Greco MA, Karpatkin S. Int J Cancer 1993; 53: 978-982. Vu TKH, Wheaton VI, Hung DT, Coughlin SR. Nature 1991; 353: 674-677. Liu L, Vu TKH, Esmon CT, Coughlin SR. J Biol Chem 1991; 266: 19697-19680. Rosenberg RD, Damus PS. J Biol Chem 1973; 248: 6490-6505. Carrell RW, Boswell DR. In: Barret AJ, Salvesen G, eds. North Holland: Elsevier, 1986; 403-418. Travis J, Salvesen GS. Ann Rev Biochem 1983; 52: 655-709. Ware JA, Salzman EW. In: Lane DA, Lindahl U, eds. Heparin Chemical and Biological Properties, Clinical Applications. London: Edward Arnold, 1989; 475494. Esmon CT. Science 1987; 235: 1348-1352. Sawyer RT. Biopapers J 1990; 10:11-13. Markwardt F. Methods Enzymol 1970; 19: 925-932. Tuszynski GP, Gasic TB, Gasic GJ. J Biol Chem 1987; 262: 9718-9723. Connolly TM, Jacobs JW, Condra C. J Biol Chem 1992; 267: 6893-6898. Budzynski AZ, Olexa SA, Sawyer RT. Proc Soc Exp Biol Med 1981; 168: 259265. Keller P, Schultz LD, Condra C, Karczewski J, et al. J Biol Chem 1992; 267: 6899-6904.
287 55 56 57 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87
Dodt J, Machleidt N, Seemuller U, Maschler R, et al. Biol Chem Hoppe-Seyler 1986; 367: 803-811. Trippier D. Folia Haematol 1988; 115: 30-35. Dodt J, K6hler S, Smitz B, Wilhelm B. J Biol Chem 1990; 265: 713-718. Stone StL Braun PJ, Hofsteenge J. Biochemistry 1987; 26:4617-4624. No6 G, Hofsteenge J, Rovelli G, Stone SR. J Biol Chem 1988; 263:11729-11735. Wallace A, Dennis S, Hofsteenge J, Stone SR. Biochemistry 1989; 28: 100-7910084. GrfitterMG, Priestle JP, Rahuel J, Grossenbacher H, Bode W, et al. Embo J 1990; 9:2361-2363. Rydel TJ, Ravichandran KG, Tulinsky A, Bode W, et al. Science 1990; 249: 277280. Braun PJ, Dennis S, Hofsteenge J, Stone SR. Biochemistry 1988; 6517-6522. Lipowski AW. Pol J Pharmacol Pharm 1987; 39: 585-596. Krstenansky JL, Mao JJT. FEBS Lett 1987; 211: 10-16. Maraganore JM, Chao B, Joseph ML, Jablonski J, et al. J Biol Chem 1989; 264: 8692-8698. DiMaio J, Gibbs B, Munn D, Lefebvre J, Ni F, et al. J Biol Chem 1990; 265: 21698-21703. Bajusz S, Barabas E, Tolnay P, Szell E, et al. Int J Peptide Protein Res 1978; 12: 217-221. Maraganore JM, Bourdon P, Jablonski J, Ramachandran KL, et al. Biochemistry 1990; 29:7095-7071. DiMaio J, Ni F, Gibbs B, Konishi Y. FEBS Lett 1991; 282: 47-52. DiMaio J, Gibbs B, Lefebvre J, Konishi Y, et al. J Med Chem 1992; 35: 33313341). Zdanov A, Wu S, DiMaio & Konishi Y, et al. Proteins Struct Funct Gen 1993; 17: 252-265). Hanson SR, Harker LA. Proc Natl Acad Sci USA 1988; 85:3184-3188. Kelly AB, Marzec UM, Krupski W, Bass A, et al. Blood 1991; 77: 1006-1012. Maraganore JM. In: Anderson PS, Kenyon GL, Marshall GR, eds. Perspectives in Drug Discovery and Design. Leiden: Escom, 1994; 461-478. Shand RA, Smith JR, Wallis RB. Thromb Res 1984; 36: 223-232. Carone FA, Peterson DR, Flouret G. J Lab Clin Med 1982; 100: 1-14. Markwardt F. Cardiovasc Drug Rev 1992; 10:211-232. Kurz KD, Main BW, Sandusky GE. Thrombosis Res 1990; 60: 269-280. Walz DA, Anderson GF, Fenton JW II, Shuman MA. Ann NY Acad Sci 1986; 485: 323-334. Fenton JW, II. Ann NY Acad Sci 1986; 485: 5-15. Shuman MA. Ann NY Acad Sci 1986; 485: 228-229. Bar-Shavit R, Kahn AJ, Mann KG, Wilner GD. Proc Nat Acad Sci 1986; 83: 976980. Bar-Shavit R, Benezra M, Sabbah V, Bode W. Amer J Resp Cell Mol Biol 1992; 6: 123-130. Davey MG, Luscher EF. Nature (Lond) 1967; 216: 857-858. Chen L-B' Buchanan JM" Pr~ Nat Acad Sci 1975; 72: 131-135. White RP, Chapleau CE, Dugdale M, Robertson JT Stroke 1980; 11: 363-368.
288 88 89 90 91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106
Cunningham DD, Farrell DH. Ann NY Acad Sci 1986; 485: 240-248. Bar-Shavit R, Eldor A, Vlodavsky I. J Clin Invest 1989; 84:1096-1104. Cuatrecasas P and Hollenberg MD. Adv Protein Chem 1976; 30:251-451. Coughlin StL Vu T-K, Hung DT, Wheaton VI. J Clin Invest 1992; 89:351-355. Ishii K, Hein L, Kobilka B, Coughlin SR. J Biol Chem 1993; 268: 9780-9786. Scarborough RM, Naughton MA, Teng W, Hung DT, et al. J Biol Chem 1992; 267: 13146-13149. Vassallo RR, Jr, Kieber-Emmons T, Cichowski K, Brass LF. J Biol Chem 1992; 267: 6081-6085. Mummatsu I, Laniyonu AA, Moore GJ, Hollenberg MD. Can J Physiol Pharmacol 1992: 70: 996-1003. Yang S-G, Laniyonu AA, Saifeddine M, Moore GJ, et al. Life Sciences 1992; 51: 1325-1332. DeBlois D, Drapeau G, Petitclerc E, Marceau F. Brit J Pharmacol 1992; 105: 959967. Antonaccio MJ, Normandin D, Serafino R, Moreland S. J Pharmacol Exp Ther 1993; 266: 125-132. Laniyonu AA, Hollenberg MD. Brit J Pharmacol 1995; 114:1680-1686. Hung DT, Wong YH, Vu T-KH, Coughlin SR. J Biol Chem 1992; 267: 2083120834. Vouret-Craviari V, Obberghen-Schilling, Rasmussen UB, Pavirani A, et al. Mol Biol Cell 1992; 3: 95-102. Glen KC, Frost GH, Bergmann JS, Carney DH. Pept Res 1988; 1: 65-73. Hollenberg MD, Laniyonu AA, Saifeddine M, Moore GJ. Mol Pharmacol 1993; 43: 921-930. Chao BH, Kalkunte S, Maraganore JM, Stone SR. Biochemistry 1992; 31: 61756178. Hui KY, Jakubowski JA, Wyss VL, Angleton EL. Biochem Biophys Res Comm 1992; 184: 790-796. Sabo T, Gurwitz D, Motola L, Brodt P, et al. Biochem Biophys Res Commun 1992; 188:604-610.
107 108 109 110 111 112 113 114 115 116
Hollenberg MD, Yang SG, Laniyonu AA, Moore GJ, et al. Mol Pharrnacol 1992; 42: 186-191. Natarajan S, Riexinger D, Peluso M, Seiler SM. Int J Peptide Protein Res 1995; 45: 145-151. Tay-Uyboco J, Poon M-C, Ahmad S, Hollenberg MD. Brit J Pharmacol 1995; 115: 569-578. Ahlquist RP. Am J Physiol 1948; 153: 586-600. Mclntyre P, Phillips E, Skidmore E, Brown M, et al. Mol Pharmacol 1993; 44: 346355. Kenakin TP, Morgan PH. Mol Pharmacol 1989; 35:214-222. Nystedt S, Emilsson K, Wahlestedt C, Sundelin J. Proc Natl Acad Sci USA 1994; 91" 9208-9212. Nystedt S, Larsson A-K, Aberg H, Sundelin J. J Biol Chem 1995; 270: 5950-5955. Kinlough-Rathbone RL, Rand ML, Packham MA. Blood 1993; 82: 103-106. Walker RM, Dai J, Ku DD. Faseb J 1995; 9: A57.
289 117 118 119 120 121 122
Rasmussen UB, Gachet C, Schlesinger Y, Hanau D et al. J. Biol. Chem 1993; 268: 14322-14328. Coughlin StL Scarborough RM. Intemational Patent Application, Publication No WO92/14750, 3 Sept. 1992. Seiler SM, Peluso M, Michel IM, Goldenberg H, et al. Biochem Pharmacol 1995; 49: 519-528. Tesfamariam B. Am J Physiol 1994; 267: H1962-H1967. Ku DD, Zaleski JK, Palgunachari MN. Faseb J 1994; 8: A328. Doorbar J, Winter G. J Mol Biol 1994; 244: 361-369.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardinb., A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All fights reserved.
291
Structure-Activity-Relationships of Non-Peptide Kappa-Opioid Analgesics: A Perspective of the Last 10 Years D.C. Rees Organon Laboratories, Newhouse, Scotland, ML1 5SH, UK
INTRODUCTION Opium, obtained from the ripe seed capsule of the poppy plant, Papaver somniferum, has been used to alleviate pain and induce sleep in humans for over two thousand years. During the last two decades research into the pharmacology of opioid receptors has led to the identification of at least three opioid receptor subtypes, designated mu, kappa and delta. Recently the human forms of these three receptor subtypes have been cloned. They are characteristic of the seven transmembrane domain structure of the G-protein coupled receptor superfamily [la]. Activation of mu receptors is associated with classical morphine-like properties such as centrally mediated analgesia, constipation, respiratory depression and addiction. Activation of kappa receptors appears to be associated with centrally mediated analgesia, sedation and diuresis. This has provided the impetus for the discovery of selective kappa opioid agonists for evaluation as clinically useful analgesic drugs without the limiting side-effects that characterise morphine and all morphine-related pain relieving drugs in use today. Some kappa selective compounds have recently been progressed into clinical trials, although to date results describing their analgesic properties in humans have not been published. Potential side-effects include diuresis, sedation and possibly dysphoria. The discovery of highly selective non-peptide kappa agonists has assisted the pharmacological study of this system and it has become apparent that these compounds also behave as neuroprotective agents in animal models of stroke/cerebral ischaemia. This is of particular current interest because of the lack of any effective medicine for these patients. Further developments in the kappa field have led to patents and publications disclosing selective antagonists and also the identification of agonists with limited access to the central nervous system. The potential therapeutic utility of these agents is currently being investigated. AIM
The aim of this project was to synthesise non-peptide selective kappa opioid agonists for evaluation as new analgesic drugs.
292
KAPPA OPIOID AGONISTS:
EXAMPLES OF PEPTIDOMIMETICS
During the last decade several medicinal chemistry projects have aimed at discovering low molecular-weight, non-peptide ligands which modulate the actions of membrane bound receptors of endogenous peptide neurotransmitters [l b]. The opioids (e.g. morphine) are sometimes regarded as prototype "peptidomimetics" since they act as receptor agonists of the endogenous opioid peptides (e.g. endorphins, enkephalins). In a similar way the small molecule kappa agonists described in this talk can be regarded as peptidomimetics of the dynorphin peptides. Three distinct medicinal chemistry strategies have emerged for the discovery of these non-peptides, firstly broad screening followed by lead optimisation, secondly design incorporating elements of a peptide structure, and thirdly modification of an existing non-peptide lead [1]. The opioids discussed here exemplify the third of these strategies. EARLY SYNTHETIC ACHIEVEMENTS
Ten years ago, when this project began in the Parke-Davis Laboratories there were a number of chemical structures reported in the literature as possessing high affinity for the kappa opioid receptor. Some of these are shown below (fig. 1). They include the endogenous peptide, dynorphin, the benzomorphan derivative EKC, the benzodiazepine tifluadom, and the 1,2 aminoamide U-50488. At this point ! would like to emphasise .the importance of the prototype Upjohn compound, U-50488 [2]. It represents a milestone in kappa opioid research because of its high selectivity for this receptor and its in vivo activity as a non mu-opioid analgesic agent. This compound was selected by the Parke-Davis group (and subsequently by many others) as an attractive chemical lead for synthetic modification. STRUCTURE-ACTIVITY-RELATIONSHIPS OF THE AROMATIC RING IN THE U-50488 SERIES
The dichlorophenyl aromatic ring of U-50488 was extensively modified to probe the SAR of this fragment. Replacement with a bicyclic 10-pi aromatic group rapidly led to the identification of the benzo[b]thiophene series. Five regioisomers of benzo[b]thiophene acetic acid were prepared. Substitution at the 4-position was shown to be optimal in terms of in vitro kappa receptor binding affinity and mu-kappa binding selectivity [3] (see fig. 2). A 4-benzo[b]furanyl ring retains greater than 100-fold mu-kappa selectivity and this group also found favour in later structures. The 4-benzo[b]thiophene derivative PD117302, is typical of these compounds in that it causes naloxone reversible inhibition of electrically evoked contractions of isolated guinea-pig ileum (IC50=l.lnM) and behaves as a naloxone reversible analgesic in a variety of rodent models, for example, mouse tail clip MPE50=2.2 mg/kg sc [4]. Other pharmacological effects are characteristic of a kappa agonist, i.e. naloxone reversible diuresis and locomotor impairment, but no sign of mu-receptor mediated
293 Dynorphin (1 - 17) = Tyr-Gly-Gly-Phe-Leu-Arg-Arg-Ile-Arg--Pro--Lys--Leu-Lys-Trp-Asp-Asn-GI n CH 3 CH2NHCO
o
7
HO
Ethyl ketocyclazocine
Tifiuadom
Cl
u-50488 Figure 1. "Kappa-Selective" Ligands (1985).
~
NCO - Ar
Opiotd __l~c~__~lorBinding
~nnny~ (nu)
4
Kappa
(+.)
Mu Mu/kappa Rat Paw i~ouun~ ratio Assay (iv) MPE,aiomg4cg ,
Substitution Poslton X 3 4 5 6 7
Me
OPIOID BINDING Ki (nM) Kappa Mu Mu/Kappa 69 3.9
$
94
$ S
ClCz # ~
3.4
864
258
HT
~C/-~~
12.7
4340
341
NT
127
>104
0.37
244
859
0.014
1.2
273
227
0.43
,
S S
146 17
1,400 410 475 425 650
21 110 5 3 39
Figure 2. Structure-affinity relationships of aromatic ring.
,
~C/~~
NT
Figure 3. Conforrnationally restricted aromatic side chain.
294 respiratory depression, inhibition of gastrointestinal motility or naloxone induced withdrawal jumping in mice. Having concluded that steric properties of the aromatic ring are an important factor in determining receptor selectivity the study was extended by incorporating a methyl group on the o~-carbon between the aromatic ring and the amide carbonyl. This significantly reduced kappa receptor affinity and after consulting molecular models it was suggested that this inactivity may be caused by the methyl group disfavouring the biologically active conformation. One such conformation is when the methyl group interacts unfavourably with the C-3 proton on the aromatic ring. If this is the conformation required for kappa receptor binding then it should be possible to increase affinity by locking the aromatic group in this orientation. To test this hypothesis a rigid tricyclic acenaphthene moiety was prepared. The results, given in the table below (fig. 3), show that one of the acenaphthene diastereoisomers has 10fold greater affinity than the corresponding conformationally flexible napthalene analogue and behaves as a potent analgesic in the rat paw pressure test [5].
CI-977 (ENADOLINE)
Credit has already been given to the Upjohn compound, U50488 as being the prototype 1,2-amino amide selective kappa agonist: A second development in the SAR of these compounds discovered by Upjohn involved incorporating a spiroether group at the C-4 position of the cyclohexane ring to increase the kappa analgesic activity [6] (fig. 4). The compound with which Upjohn performed clinical trials, U62066 (spiradoline), contained this structural modification. However, U 62066 is a racemic mixture of two enantiomers and has only a modest mu/kappa selectivity ratio (approx. 10-fold) [7]. The compound which Parke-Davis selected for clinical evaluation was CI-977 (enadoline), a single enantiomer of the 4-benzofuranyl derivative of spiradoline and a very selective, potent kappa agonist [8], see (fig. 5,6). The diuretic effects, pharmacokinetic effects and safety of CI-977 in 16 healthy human volunteers have been reported over a dose range of 5-25 l~g intramuscular injection [9]. CI-977 Crnaxand AUC(0-oo) values increased in proportion to dose and significant dose-related decreases in negative free water clearance and urine osmolality were observed, in accordance with the diuretic effects previously seen in rodents. Adverse effects include dizziness and fatigue. In another rising, singledose tolerance study in humans doses up to 40 ~g were administered intramuscularly. The highest well tolerated dose was 15 ~g. No euphoria or dysphoria occurred [10].
295
Upjohn's Spiroether Derivatives .=
(_+)
CH31 _ ..,N- ~1" CH=
CI
~
CI
U-62066 (Spiradoline) 913 x more potent analgesic than U-50488 9(-) enantiomer more potent and more selective 9
chloro derivative is highly kappa selective (U-69593) Vonvoigtlanderand Lewis,1988
Figure 4. Upjohn's spiroether derivatives.
Receptor binding affinity (K, nM) Compound kappa
mu
delta
CI-97"/ Spiradoline U-69593 EKC
100 44 2460 1.02
1040 4530 9900 7.0
0.11 0.35 0.67 0.21
Smooth muscle (ICs0,nM)
mu/kappa 905 125 3670 4.9
GP!
LVD
0.087 0.88 3.4 0.26
3.3 29 89 22
Figure 5. Opioid activity in vitro for CI-977 and reference compounds.
296
MPEs0 (mg/kg) (tail clip, s.c.)
Mouse
(paw pressure,i.v.) (tooth pulp, i.m.)
Rat
Dog
(tail immersion i.m)
C I-977
0.03
Oo02
0.00026
0.001
Spiradoline
0.81
0.38
0.0022
NT
Morphine
2.4
1.3
NT
1.0
Compound
Monkey
Figure 6. Antinociceptive activity of CI-977 and reference compounds.
Me I
MeO2CN,,~
~
( . . ~ . ~ NCOCH," - ' ~ o
,/~CI NCOCH~ - - ~ - - C l
.H I. CI-977
2. GR 103545
O
v
"N~
~
"CI
Me HO
3.
4.
5. EKC
Figure 7. Kappa Opioid Agonists used in molecular modelling study.
297
DESIGN OF NOVEL STRUCTURES (UNRELATED TO U-50488) During the late 1980's a number of laboratories reported preliminary data on a variety of kappa agonists based on the 1,2-amino amide, U-50488. Some of these are shown in fig. 7. We decided to use a molecular modelling approach to probe the chemical similarities of these non-peptide agonists together with the benzomorphan, EKC, in order to construct a template which could be used to design novel structures for evaluation as kappa ligands [11]. The compounds that were overlayed with each other according to a 3-point pharmacophore model (basic nitrogen atom and two aromatic ring centroids) are shown in fig. 8. This generated a proposed binding model for these kappa agonists. Novel structures could then be examined via computer modelling to see which fitted the model. In this design three factors were considered. Firstly, the designed molecules should have appropriately orientated aromatic and amino groups. Secondly they should as far as possible occupy space within that occupied by the known kappa ligands. Thirdly, they should be as simple as possible in terms of chemical synthesis. Three series, a pyrrolizidine, a macrocyclic iactam and an aryl amine, were then selected for synthesis (fig. 9). The pyrrolizidine (PD 146795) (kappa K~=940 nM, mu/kappa ratio=3.5) and the arylamine (kappa K~=160 nM, mu/kappa ratio >60) represent novel chemical leads for kappa opioids and they illustrate the success of this molecular modelling strategy for designing new receptor ligands. Despite the macrocyclic lactam (PD 146884) fitting the model quite well it is essentially devoid of affinity indicating that this criterion alone is not sufficient for activity. During this study we synthesised and tested fewer than ten compounds based on our receptor binding model yet one of these (PD 148282) has 160 nM affinity for the kappa opioids receptor and is selective with respect to the mu receptor. Unlike almost all other kappa selective ligands reported at the time it is not a 1,2-aminoamide. Since this study was performed a number of other kappa ligands have been reported. A selection of some of these are shown in figs. 10 and 11. ACKNOWLEDGEMENTS i would like to acknowledge the numerous scientists at the Parke-Davis Research Unit who contributed this project over many years. In particular I note the senior staff who initiated the project, Dr. David Horweil (Medicinal Chemistry), Dr. Ray Hill and Professor John Hughes (Pharmacology). Dr. John Hunter (Pharmacology) and Dr. Geoff Woodruff (Pharmacology) also made leading contributions.
298 !
Figure 8. Relaxed stereoview of the spatial overlay of compounds 1-5. N and N 1 are the basic nitrogens of EKC and the aryl acetamides respectively. The three points which define our binding model are labelled p.
kappa
mu
mu/kappa ratio ,
940
3350
,,
3.5
6 Oploid receptor btndlng affinity
(KI, .M)
0
CONMe
~
~
>10 000
>10 000
160
>10 000
>60
Figure 9. Novel structures designed to fit binding model.
299
OyMe
C02Me I
OH O~~~--
Cl
SO=Me peripherally selective agonist LVD ICso= 0.9nM (Glaxo, 1992)
O
CI
GR 89696 LVD IC~ = 0.041nM (Glaxo, 1993)
CH3 ~' CHPh2
~ N (
0 ' ~ 0 H .HCI
kappa binding K~= 47nM (SB-Italy, 1994)
EMD 61753 kappa binding = 2nM (E. Merck, 1994)
Figure 10. Recentlyreportedselective kappa agonists based on U-50488.
300
HO
N~COOCH3 CCB (Ronsisvaile, 1993)
HN
O NMe CO2Me
(Borsodi, 1994)
Figure 11. Recently reported selective kappa ligands not based on U-50488.
301
REFERENCES
l(a) Knapp RJ, Malatynska E, Collins N, Fang L, Wang JY, Hruby V J, Roeske WR and Yamamura HI, Faseb J, 1995; 9: 516-525. l(b) Rees D: In: Bristol J, ed. Ann Rep Med Chem 1993; 28:59-68 and references cited therein. 2(a) Szmuszkovicz J and Von Voigtlander PF, J Med Chem 1982; 25: 1126-1127; (b) Lahti RA, Von Voigtlander PF and Barsuhn, C Life Sci 1982; 31:, 22572260; (c) Katz JL, Woods JH, Winger GD, Jacobson AE, Life Sci 1982; 31: 2375-2378. 3. Clark CR, Halfpenny PR, Hill RG, Horwell DC, Hughes J, Jarvis TC, Rees DC and Schofield D, J Med Chem 1988; 31: 831-836. 4. Clark CR, Birchmore B, Sharif NA, Hunter JC, Hill RG and Hughes J, Br J Pharmacol 1988; 93: 618-626. Leighton GE, Johnson MA, Meecham KG, Hill RG and Hughes J, Br J Pharmacol 1987; 92: 915-922. 5. Halfpenny PR, Horwell DC, Hughes J, Humblet C, Hunter JC, Neuhaus D and Rees, DC J Med Chem 1991 ; 34: 190-194. 6. Lahti RA, Mickelson MM, McCall JM and Von Voigtlander PF, Eur J Pharmacol 1985; 109: 281-284. 7. Meecham KG, Boyle SJ, Hunter J and Hughes J, Eur J Pharmacol, 1989, 173, 151-158. Von Voigtlander PF and Lewis RA, J Pharmacol Exp Ther 1988; 246: 259-262. 8. Halfpenny PR, Horwell DC, Hughes J, Hunter JC and Rees DC, J Med Chem 1990; 33: 286-291. Hunter JC, Leighton GE, Meecham KG, Boyle SJ, Horwell DC, Rees DC and Hughes J, Br J Pharmacol 1990; 101: 183-189. Davis RE, Callahan MJ, Dickerson M, Downs D, J Pharmacol Exp Ther 1992; 261: 10441049. 9. Reece PA, Sedman A J, Rose S, Scott Wright D, Dawkins R, Rajagopalan R, J Clin Pharmacol 1994; 34:1126-1132. 10. Dawkins R, Lebsack ME, Scott Wright D, Posvar EL, J Clin Pharmacol, 1991; 31 : 841-871 (Abstr. 97). 11. Higginbottom M, Nolan W, O'Toole J, Ratcliffe GS, Rees DC, BioMed Chem Lett 1993; 3: 841-846.
This Page Intentionally Left Blank
Perspective in Receptor Research D. Giardinh, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
303
Selective N o n p e p t i d e L i g a n d s as Probes to Explore 8 Opioid R e c e p t o r Architecture P.S. Portoghese
Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, USA INTRODUCTION The relatively recent molecular cloning and sequencing of opioid receptors [1-5] has confirmed the mlmerous pharmacologic and structureactivity relationship studies that have suggested the existence of multiple opioid receptors. Presently, there appear to be at least three types of receptors, designated as 8, ~ and ~, in the opioid receptor family which have high homology and are members of the rhodopsin-like class within the superfamily of G protein-coupled receptors. Numerous ligands have been developed as potential opioid receptor probes in an effort to distinguish between different types of opioid receptors. These include peptide [6,7] and nonpeptide [8] agonists and antagonists. Here we discuss the design and structure-activity relationship studies of ~selective nonpeptide ligands and the role of conformation of the "address" moiety in conferring selectivity to the 81 and 82 subtypes of opioid receptors. THE MESSAGE ADDRESS CONCEFI" IN ~
DESIGN OF NALTRINI~!.E
The prototypical nonpeptide 8 opioid antagonist, naltrindole [9] 1 (NTI), is employed widely as a pharmacologic tool [10]. The rationale for the design of NTI was based on the "message-address" concept which was developed by Schwyzer [11] to analyze the structure-activity relationship of ACTH-related peptides. For agonists, the "message" component of the ligand was envisaged to be responsible for triggering signal transduction upon interaction with the receptor. The "address" was considered to target the ligand to a specific receptor.
OH
H
I
H
1 0~TD
This concept was tested initially in the design of selective opioid ligands by the attachment of putative 8 and ~ address fragments of leucine-enkephalin
304 and dynorphin to oxymorphone, which contains the opioid message component [12]. It was found that the enkepbalin-derived sequence conferred 8 selectivity while the sequence obtained from truncated dynorphin afforded a ~selective ligand. NTI contains an indolic benzene moiety that was envisaged to mimic a key element of the 8 address, the Phe 4 phenyl group of enkephalin. In this context the indolic pyrrole moiety was viewed as part of a molecular scaffold to hold the address. ROLE OF ~ MOLECIKAR SCAFFOLD IN DETERMINING DELTA B E C E F T O R SUBTYPE ~.]e~-L'IVI'I-L While the aforementioned studies are consistent with the concept that selective ligands interact with message and address subsites on the 8 opioid receptor, it was unclear whether the recognition sites for agonists and antagonists are the same, are overlapping, or are different. If agonists and antagonists interact with identical recognition sites, then it was expected that substitution of a methyl for the N-cyclopropylmethyl group in NTI should afford a 8 agonist by analogy with naltrexone 2 and oxymorphone 3. N.-R
N..CI'~
OH
H
2, R = CH2CH(CH2)2 3, R=CH3
4 (OMi)
H
The finding that the oxymorphone analogue, oxymorphindole [13] 4 (OMI), is not a 8 agonist and retains potent 8 opioid antagonist activity in vivo [14], raised the possibility that the molectdar scaffold that holds the address i n 4 might play an i m p o ~ t role in promoting 8 antagonism by fixing the address in a specific conformation. This would imply that Phe 4 in the enkephalins plays a dual role, in that it may serve both as a functional but nonessential part of the message and as an essential part of the 8 address. Molecular dynamics simulation of leucine-enkephalin has shown that the conformational space occupied by the phenyl group of Phe 4 is restricted to the region of the indolic benzene moiety [15] (Fig. 1). However, it appeared unlikely that the Phe 4 phenyl group can assume a conformation identical with that of the indolic benzene moiety because superposition of both rings was not observed during the simulations. In this regard, many of the Phe 4 phenyl conformations were nearly perpendicular to the plane of the indole system.
305
Figure 1.
Ten low energy conformations of leucine enkephalin superposed upon OMI. The indolic benzene moiety is indicated by bold lines.
These results contributed to the design of spiroindan-substituted opiates with a a address moiety that is restricted to a conformation that is perpendicular to that of NTI or OMI. This restriction is due to the spiro substitution which prevents torsion of the indan benzene moiety.
N-R OH
H 5. R = Ci-12CH(CH2)2 (SINTX) 6, R = C1-13 (SIOIVl)
7-Spiroindanylnaltrexone 5 (SINTX) was found to be a potent 8 antagonist with approYimately one-sixth the atrmity of NTI for 8 receptors, although its ~selectivity was considerably lower [16]. Significantly, replacement of the cyclopropylmethyl of SINTX with a methyl group afforded a ligand 6 (SIOM) with potent 5 agonist activity [17]. The fact that this is in contrast to OMI 4, which has its address moiety restricted to a conformation that is coplanar to ring C of the opiate, is consistent with the idea that agonists and antagonists bind either to different sites or to overlapping sites on the 5 receptor. Superposition of SIOM upon OMI illustrates the different orientations of the address moiety (Fig. 2).
306
.~OM
Figure 2.
A stereoview of OMI superposed upon SIOM.
INTERACTION OF LIGANDS W i t h CI~NED WH.n-TYPE AND MUTANT DI~.TA OPIOID RECEPTORS Studies of SIOM and NTI binding to mutant and wild type cloned 8 opioid receptors were consistent with different or overlapping recognition sites for agonists and antagonists [18]. The point mutation involved the replacement of Asp95 with Asn in tr~n~membrane s p a n n i n g region 2 (TM2), which is conserved in the rhodopsin class of G protein-coupled receptors. The fact that the Asn95 mutant exhibited a great decrease in at~inity for 8 agonists, while antagonists were unaffected, suggests that their interaction with 8 receptors is fundamentally different (Table 1). Table 1 Inhibition of [3H]NTI binding to wild-tTpe and Asp95Asn mutant 8 receptors ICso (nM) a Ligand 8 A~onists Deltorphin DPDPE Met-enkephalin SIOM (6) 8 Antagonists NTI (1) NTB (9) BNTX (8) ,,,,
aData from [18].
Wild Type 48 245 81
Asp95Asn 552 >I000
>1000
33 40
>1000 544
2.1 0.04 37
1.7 0.09 30
307 Also consistent with the possibility of different binding modes for agonists and antagonists is the finding that the Asp128Asn mutant in TM3 greatly reduced agonist binding to the S receptor, but did not substantially alter antagonist binding [19]. Asp128 is conserved among the subgroup of G protein coupled receptors that recognize aminergic ligands. It has been suggested that this conserved Asp serves as the anionic binding site for cationic protonated amine or quaternary ammonium groups in different pharmacologic classes of agonist ligands [19]. Since the Asn128 residue is incapable of ion-pairing with a cationic group, the mutant should have considerably lower affinity than the wild type 8 receptor for 8 agonists. While this was found to be the case for agonists, the antagonists exhibited minimal differences in binding between mutant and wild-type receptors. This once again suggests that Asp128 is not involved in the recognition of antagonists and that the recognition sites for agonists and antagonists differ. The fact that the Asn128 mutant bound 8 antagonists equally as well as the wild-type receptor might mean that a receptor-based anionic residue is not important for the binding of antagonists. However, another interpretation is that a counterion is necessary, but an anionic residue other than Asp128 is involved in the binding of antagonists. In an effort to distinguish between these possibilities, amide 7 corresponding to NTI was synthesized and found to possess little or no airmity for 8 receptors [20]. This suggested the latter interpretation of the data the more likely possibility. 0
H
I
H
DI~.TA OPIOID RECk--fOR SUBTYPES
Evidence for the existence of subtypes of the 8 opioid receptor (81 and 82) has been acquired through in vivo pharmacologic studies using selective opioid agonists and antagonists [21,22]. [I)-Pen2,D-Pen5]enkeph~lin [23] (DPDPE)is selective for 81, while [D-Ser2,LeuS]-enkephalin Thr 6 [24] (DSLET) and [DAla2]deltorphi~n II [25] are selective for 82. The antagonist ligands employed to distinguish between these 8-receptor subtypes were [D-Ala2,LeuS]enkephalin Cys 6 [26] for 81 and the NTI analogues, naltriben [15] 9 ( N ) and naltrindole5'-isothiocyanate [27] for 82.
308
H
8 (BN'~
9 (N'm)
[~ to
It appears that the conformation of the address may contribute to subtype selectivity, but it is dearly not the only factor. With regard to the role of conformation, the 81-selective antagonist, 7-benzylidenenaltrexone [28] 8 (BNTX), possesses a phenyl group that is orthogonally oriented with respect to ring C of the morphinan structure. The nonpeptide 8 agonist, SIOM 6, has been reported [17] to be al selective and also has a similarly oriented address. This is in contrast to. the coplanar arrangement of the address in the 82selective antagonist NTB 9 (Fig. 3).
Yi~u'e 3.
Superposition of SIOM, BNTX, and NTB.
If conformation were the only factor contributing to a subtype selectivity, NTI would be expected to be a 82-selective antagonist, as its address moiety is superimposable with that of NTB. However, we have found that NTI does not dearly distinguish between a subtypes. Thus, an additional factor may play a role in 8 subtype selectivity. In view of the fact that the 82 antagonists, NTB and N-benzyl-NTI [29] 10, are more lipophilic than NTI, their 8 subtype selectivity might be related in part to their greater lipid solubility. The pharmacologic evaluation of amino acid conjugates (11, 12) of NTI has provided some insight into this possibility, as these compounds are considerably more hydrophilic than NTI [30]. The finding that 11 and 12 are highly potent al-selective antagonists in vivo suggests that subtype selectivity of 8 antagonists may in part be a function of the greater hydrophilic character contributed by their amino acid residues. Thus, the hydrophilic-lipophilic
309 balance of the antagonist ligand might differentially modulate access to different compartments in the CNS that contain 81 and 82 receptors. For example, accessibility to tissue compartments that contain 81 receptors might be favored by more hydrophilic molecules. Conversely, hydrophobic antagonists might gain access to 52 receptors more readily, while access to 81 receptors is impeded.
~176
H
~C;OOH
i
HO
o
H ""R
11, R = H 12, R = CH2COOH
SUMMARY AND CONCLUSIONS Structure-activity relationship studies have suggested that agonist and antagonist ligands bind to separate or overlapping recognition sites on 8 opioid receptors. These sites eThibit different conformational requirements for the address component (phenyl or benzene moiety). In this regard, nonpeptide agonists (e.g., SIOM) require the plane of the aromatic group to be approximately orthogonal to the plane of ring C in the opiate. On the other hand, there are no specific conformational requirements for antagonists, as the aromatic group that comprises the address may be coplanar (e.g., NTI or NTB) to ring C or oriented orthogonally (e.g., SINTX, BNR~). However, an aromatic group that is coplanar with ring C possesses greater antagonist potency than an orthogonally oriented group. Is there any physiologic significance for separate agonist and antagonist binding sites? It seems within the realm of possibility that binding of endogenous opioids (e.g., enkephalin) to discrete recognition sites on the a receptor may play a modulatory role in signal transduction. A model for the existence of separate recognition sites for agonists and antagonists was proposed in 1983 [31] for the g opioid receptor and is presented here in a modified version for the 8 receptor in Figure 4. We suggest that the 8 receptor possesses two discrete recognition sites that are topographically similar, but not identical. The sites are proposed to possess different afAnities for enkephalin. For agonists, binding to the higher atrmity site (site 1) is associated with signal transduction, while occupation of the lower amnity binding site (site 2) decreases the binding to site 1. Site 2 therefore would function as a regulatory site to reduce signal transduction at high concentrations of endogenous peptide.
310 ml
~rrN~
~F
Figure 4.
A conceptual model for distinct agonist (site 1) and antagonist (site 2)recognition sites on opioid receptors. Binding of endogenous agonist to site 1 stabilizes the active state (a). The inactive state is stabilized by binding of agonist to site 2 (b). The efficacy of an agonist is reflected by the steady-state ratio of agonist-bound active and inactive states. A ~pure ~ antagonist binds selectively to site 2 to shift equilibrium almost entirely to the inactive state. The wavy arrow within the receptor signifies receptor activation of G protein, while the straight arrow symbolizes the stabilization of G protein in the inactive state.
The presence of two sites for agonists present some intriguing possibilities for the SAR of opioid ligands. For example, the efficacy of an agonist ligand would be determined by its relative ~fl~nity for sites 1 and 2. High efficacy would be due to much higher affinity for the agonist site (site 1) relative to the regulatory site (site 2). Conversely, low efficacy agonists or partial agonists would be ligands whose atrmity to site 2 relative to site I is greater than that of high efficacy agonists. An antagonist is envisaged to be a ligand whose atrmity for site 2 is much greater than for site 1. Antagonist binding to site 2 would also promote stabilization of the inactive state of the opioid receptor (32-35). Finally, pharmacologic studies point to the existence of 8 receptor subtypes. The antinociceptive effect of SIOM appears to be mediated through 81 opioid receptors. If DPDPE and SIOM bind to the same agonist recognition site on the 81 receptor, this would suggest a similar conformational relationship between the message and address components for these molecules. The conformation of the address in antagonist ligands plays a less i m p o ~ t role in 8 subtype selectivity, inasmuch as attachment of polar residues to NTI
311 changes the subtype selectivity to 51 receptors even though it retains its coplanar aromatic group which appears to favor 62. Thus, 6 subtype selectivity for antagonists may be determined by a combination of factors. While the coplanar orientation of the address and ring C (e.g., NTB) appears to confer ~2 selectivity, increasing the polarity of the molecule favors 81 selectivity. A possible explanation for this change in subtype selectivity may be the localization of 81 and 52 receptors in different tissues or cellular compartments having different accessibility to the antagonist ligands. REFERENCES
10 11 12 13 14 15 16
17 18 19 20 21
Evans CJ, Keith DE, Morrison H, Magendzo K, Edwards RH. Science 1992; 258; 1952-1955. Kieffer BL, Befort K, Gaveriaux-Ruff C, Hirth CG. Proc Natl Acad Sci USA 1992; 89; 12048-12052. Yasuda K, Raynor K, Kong H, Breder CD, Taketa J, Reisine T, Bell G I. Proc Natl Acad Sci USA 1993; 90; 6736-6740. Chen Y, Mestek A, Liu J, Hurley JA, Yu L. Mol Pharmacol 1993; 44; 8-12. Wang JB, IrnAi y, Eppler CM, Gregor P, Spivale CE, Uhl GR. Proc Natl Acad Sci USA 1993; 90; 10230-10234. Schiller PW. Prog Med Chem 1991; 28; 301-340. Hruby VJ and Gehrig C~, Med Chem Res Rev 1989; 9; 343-401. Portoghese PS. In: Herz A, ed. Opioids I. Berlin; Springer Verlag, 1993; 279-293. Portoghese PS, Sul~n~ M, Nagase H, Takemori AE. J Med Chem 1988; 31;281-282. Takemori AE, Portoghese PS. Annu Rev Pharmaeol Toxicol 1992; 32; 239-269. Schwyzer R. Ann NY Acad Sci. 1977; 297; 3-26. Lipkowski AW, Tam SW, Portoghese PS. J Med Chem 1986; 29; 12221225. Portoghese PS, Sultana M, Takemori AE. J Med Chem 1990; 33; 17141720. Takemori AE, Sultana M, Nagase H, Portoghese PS. Life Sci 1992; 50; 1491-1495. Portoghese PS, Nagase H, MaloneyHuss KE, Lin CE, Takemori AE. J Med Chem 1991; 34; 1715-1720. Portoghese PS, Sultana M, Moe ST, Takemori AE. J Med Chem 1994; 37; 579-585. Portoghese PS, Moe ST, Takemori AE. J Med Chem 1993; 36; 2572-2574. Kong H, Raynor K, Yasuda K, Moe ST, Portoghese PS, Bell GI, Reisine T. J Biol Chem 1993; 268; 23055-23058. Reisine T. Neuropharmacol 1995; 34; 463-472. Larson DL, Portoghese PS. Unpublished data. Sofuoglu M, Portoghese PS, Takemori AE. J Pharmacol Exp Ther 1991; 257; 676-680.
312 22 23 24 25 26
27 28 29
30 31 32 33 34 35
Jiang Q, Takemori AE, Sultana M, Portoghese PS, Bowen WD, Mosberg HJ, Porreca F. J Pharmacol Exp Ther 1991; 257; 1069-1075. Mosberg HI, Hurst R, Hruby VI, Gee K, Yamamura HI, Galligan JJ, Burks TF. Proc Nail Acad Sci U S A 1983; 80; 5871-5874. Fournie-Zaluski M-C, Gacel G, Maigret B, Premilat S, Roques BP. Mol Pharmacol 1981; 20; 484-491. Erspamer V, Melchiorri P, Falconieri-Erspamer G, Negri L, Corsi R, Severini C, Barra D, Simmaco M, Kreil G. Proc Nail Acad Sci U S A 1989; 86; 5188-5192. Bowen WD, HeUeweU SB, Keleman M, Huey R, Stewart D. J Biol C h e m 1~7; 262; 13434-13439. Portoghese PS, Sultana M, Takemori AE. J Med Chem 1990; 33; 15471548. Portoghese PS, Sultana M, Nagase H, Takemori AE. Eur J Pharmacol 1992;218; 195-196. Korlipara VL, Takemori AE, Portoghese PS. J Med Chem 1994;37; 18821885. Portoghese PS, Farouz-Grant F, Takemori AE. J Med Chem 1995; 38; 402-407. Portoghese PS, Takemori AE. J Med Chem 1983; 26; 1341-1343. Costa T, Herz A~ Proc Nail Acad Sci 1989; 86; 7321-7325. Costa T, Ogino Y, Munson PJ, Onaran HO, Rodbard D. Mol Pharmacol 1992; 41; 549-560. Sr W, Freissmuth M. Trends Pharmacol Sci 1992; 13; 376-379. IV[ilIigan G, Bond RA, Lee M. Trends Pharmacol Sci 1995; 16; 10-13.
Perspective in Receptor Research D. Giardinh, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
313
THE ROLE OF NOVEL LIGANDS IN THE BIOLOGICAL CHARACTERIZATION OF SIGMA RECEPTORS Brian R. de Costa, a Xiao-shu He,b Celia Dominguez, c Wanda Williams, d Kenner C. Riced and Wayne D. Bowend aMedical Sciences Building, University of Toronto, Toronto, Ontario M5S lAB, Canada. bNeurogen Corporation, 35 Northeast Industrial Road, Branford, CT 06405, U.S.A. CDuPont Merck Pharmaceutical Co., Experimental Station, Cardiovascular Diseases Research, PO Box 80402, Wilmington DE 19880-0353., U. S. A. dLaboratory of Medicinal Chemistry, National Institutes of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Building 8, Room B1-26, 9000 Rockville Pike, Bethesda, MD 20892., U. S. A. Investigation of the physiological and pharmacological role of sigma receptors is a relatively new area which has been facilitated by the development of high affinity and selective sigma receptor ligands including putative agonists and antagonists [1]. Many different classes of drugs, ranging from steroids such as progesterone to neuroleptics such as haloperidol, exhibit high affinity for sigma receptors. This binding activity of the neuroleptic drugs suggested that sigma receptors may be responsible, at least in part, for the motor sideeffects of these drugs. The involvement of sigma receptors in several biologic and psychiatric effects ranging from psychotic behavior to second messengers have been reviewed in detail elsewhere [1,2]. More recent studies have indicated uses for sigma receptor ligands in the development of neuroprotective and atypical antipsychotic agents [1 ]. Certain sigma ligands appear to inhibit the behavioral effects of cocaine, which suggests other promising uses for these compounds [ 1]. In general terms, sigma receptors can be summarized as follows: proposed in 1976 by Martin et al non-opioid non-dopaminergic high density found in brain areas that control motor behavior found also in periphery (e.g. liver) exist as subtypes found across many species (including reptiles) MW in the 20-30kDa range no known behavioral effects cross react with many different classes of CNS active drugs confused with many different receptor types
314 The diverse biological activities which add to the "sigma enigma" include: neuroprotective activity motor disturbances certain sigma ligands antagonize motor side effects of haloperidol in rats inhibition smooth muscle contraction may be involved in the cytotoxicity of some drugs may be involved in psychotic behavior modulate Pi turnover may represent modulatory sites for other receptors diversity across many different classes of CNS active drugs The existence of at least two sigma receptor subtypes (designated sigma-1 and sigma2) has been confirmed [1 ]. The vast majority of sigma ligands exhibit high selectivity for the sigma-1 subtype. The disubstituted guanidine DTG (1, Figure 1) displays roughly equal affinity for both sigma subtypes. The 5 , 6 , 7 , 8 , 9 , 1 0 - h e x a h y d r o - 7 , 1 0 iminocyclohept[b]indoles (2, Figure 1)[3] and certain benzomorphan derivatives [4] were shown to be selective and potent sigma-2 ligands. Compounds which bind selectively to sigma1 receptors include (+)-benzomorphans such as (+)-pentazocine, (+)-3-phenyl-1propylpiperidine ([3H](+)-3-PPP), haloperidol, (+)-morphinanssuch as dextrallorphan, cis cyclohexane-l,2-diamines and ethylenediamines [1] (see Figures 3-6). As part of our program to investigate the role of sigma-1 and -2 receptor subtypes, we focused our attention on the high affinity ethylenediamine sigma ligand 3 (Figure 1) and used it as a template [5]. Our interest in the development of sigma-2 selective ligands was spurred by evidence that this subtype may be responsible for the motor effects of sigma ligands (Bowen WD et al., personal communication). This is reinforced by the observation that DTG, which binds with lower affinity to sigma-1 receptors than (+)-pentazocine, is nonetheless more effective at causing motor disturbances in rats.
NyN NH~ 1
HN
N/R ~ ~ N 2 Figure 1
3
CI Cl
315
~N@NI
N~ N
n~Ar
I
~
N ~ Ar
I
8
NH2
?
4:n=0 3:n=l 5:n=2 6:n=3 7:n=4
N~
~NN/NAr 14
I
I
N ~
N ~ Ar
9 I
I
N~
I
N~/~ NAAr I
N ~ N ~ N @ A r Ar = ~ ~ [ ~ CI C1
12
~N
~
10
N
I
~
N
I
~
Ar
& N ~ N ~ N I
13 H2N~NH ~ 15
Ar
H
H2N~N~N
H~
16
Ar
H
~vjN/~/N~
~
Ar
H N~
Ar
I 11
17 Figure 2
Binding studies of polyamines 4-1 7 (Figure 2) indicated that several of these compounds were selective for the sigma-2 receptor subtype. For example, 1 3 is 10-fold selective for sigma-2 over sigma-1 whereas 3 is 4-fold selective for sigma-1 over sigma-2 receptor subtypes [5]. These and other compounds are proving invaluable in the biological characterization of sigma receptor subtypes. The vast array of structure activity studies that have been carried out from 1976-present (see below) have provided several potential therapeutic agents with a range of biological activities that can be summarized as follows: Neuroprotective agents Sigma receptor antagonists for reducing the motor side-effects of neuroleptics Antispasmodics, muscle relaxants Cytotoxic agents (high dose) Diagnostic tools Atypical antipsychotic agents (non-dopaminergic) Modulators for the effects of other drugs: e.g. cholinergic agents, opioids... Sigma ligand classes (1 8- 4 7) discovered and developed from 1976-94 are illustrated in the Figures 3-6 below. The vast array of chemical diversity accommodated by sigma receptors suggests that these receptors may be primordial. This is reinforced by high densities of sigma receptors found in more primitive species such as reptiles (Matsumoto RR, personal communication).
316
Figure 3: Sigma ligands (1976-1990). Z N__N
N "
f/"N
N
18
F
1
N,.
X 20
R
"Y
N.
X
19
~
R
21
:.
/R
N
22
R R
N 1~2R 3 23
24
26
27
CI"
v
25
28
Key to Figure 3:1 8: (+)-SKF10,047 [(+)-cis-benzomorphan opiates]; 1: N,N'-di(otolyl)guanidine; 1 9: 1-alkyl-4-arylpiperazines; 2 0: 3-phenylpiperidines; 2 1: octahydrobenzo[f]quinolines; 2 2: (+)-morphinans; 2 3: phenothiazines; 2 4: 4phenylpiperidines; 2 5: haloperidol (and other butyrophenones); 2 6: progesterone (and related steroids); 2 7: phencyclidine (arylcyclohexylamines); 2 8: tetrahydroisoquinolines and related benzylamines
317
Figure 4: Sigma ligands (1976-1990).
cI
MeHNm~~ 29
-
R
Cl
30
31
H
,~ N ~ N ~ H Cl
NI'I2
r~
~N 32
OH
N ,~,
35
34
33
~ .,~
N. R2 36
Key to Figure 4:2 9: cis-N-methyl-2-(1-pyrrolidinyl)arylacetamides; 3 0: sertraline and other 1-phenyl-4-aminotetralins; 3 1" clorgyline (MAOI's); 3 2. ifenprodil; 3 3: ciscyclohexanediamines; 3 4" polyamines; 3 5: tricyclic antidepressants; 3 6" 2-phenethylamines
318
Figure 5: Sigma ligands (1990-94)
~..~~1~ N ,
c'
Cl
3
37
~N/R 38
~ /H R N H 39
40
o
~ , ~ o
HO
41
L~.
42
c, CI
Key to Figure 5:3 : N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(1 -pyrrolidinyl)ethylamine and other substituted ethylenediamines; 3 7: N-substituted-5-phenylpentylamines; 3 8: spiropiperidines; 3 9: hexahydrobenz[f]isoquinolines; 4 O: octahydrobenz[f]isoquinolines; 4 1: (aminoalkoxy)chromones; 4 2: 1-[2-(3,4-dichlorophenyl)ethyl]-4-(1 -propyl)piperazine
319
Figure 6: Sigma ligands (1990-94)
N/• 43
R2
Cl
NR1
44
2
i N
Cl
I
N I
45
46
~.-" L ~ LI/.L
-.~-cI
Cl cl
Cl
47
Key to Figure 6:4 3: 1 -benzyl-4-[ 1 -aryl-(1 -ketoethyl)]piperidine; 4 4: 1 -[2-(3,4dichlorophenyl)ethyl]-3-(1-pyrrolidinyl)piperidine; 2: hexahydro-7,10iminocyclohept[b]indoles; 4 5: N,N'-dimethyi-3-[2-(3,4dichlorophenyl)ethylamino]piperidine; 4 6: N-(3,4-dichlorophenylethyl)-N,N'-dimethyl-2(aminomethyl)piperidine; 4 7 1-[2-(3,4-dichlorophenyl)ethyl-2-[(1pyrrolidinyl)methyl]pyrrolidine Sigma receptor research has been greatly facilitated by the development of chemical tools [1,2] ranging from antagonists which allow the probing of sigma receptor function to PET scanning iigands which have allowed visualization of sigma receptors in mammalian brain [6]. The radioligand [3H] azidoDTG has allowed identification of two receptor subtypes in brain [7]. Modeling Several models have been proposed to account for the chemical diversity of sigma iigands: eModels permit a large degree of steric tolerance. oBasic requirements for binding are aromatic ring separated from N by 2 carbon atoms with a bulky lipophilic N substituent. eModeling: (1) Manallack 1988: hypothetical receptor points; (2) Largent 1987: 3- and 4phenyipiperidines with bulky N-substituent; (3) Ablordeppey 1992: 2-phenylpropanolamine oRevision of the above models may be necessary to account for compounds such as the ~carbolines oModels may be inaccurate due the existence of subtypes The weak binding of polyamines (Table 1) to sigma receptors suggests that polyamines may be the biological modulators of sigma receptors [8]:
320 Table 1" Inhibition of [3H](+)-3-PPP binding by polyamines. IC50 (mM+SEM) IC50 (mM+SEM) Compound IC50 (mM+SEM) Rat Forebrain Adrenal Medulla Guinea Pi9 brain 104.8+20.1 82.1+14.4 Spermine 9.2+2.0 345.3+55.0 280.8+88.2 Spermidine 69.9+22.9 1018.2+147.9 716.0+221.8 Cadaverine 398.2+ 103.3 932.2+173.0 1740.8+420.3 Putrescine 410.8+ 180.1 Data extracted from Paul et aL 19 9 0
Conclusion Our own work in the sigma receptor field can be summarized as follows: ediscovered new sigma ligands starting with the kappa opioid agonist U50,488 edeveloped compounds with high potency and selectivity by conformational restriction of N-J2( 3,4-dichlorophenyl)ethyl]-N-methyl-2-( 1 -pyrrolidinyl) ethyla mine edeveloped subtype selective agents edeveloped sigma receptor antagonists oconfirmed the involvement of sigma receptors in the motor side-effects of neuroleptics ediscovered neuroprotective, antidystonic, and even cytotoxic effects among different sigma ligands from diverse chemical classes odesigned research tools to probe the sigma receptor The field of sigma receptor research continues to be an exciting area with a strong potential for the discovery of novel therapeutic agents. With continued and intensive chemical approaches to further characterize this receptor, the "sigma enigma" may soon be solved! References 1 For an update on the structure-activity-relationships of sigma receptor ligands see: de Costa BR, He XS in: Itzhak J, Chapman C, eds. The Sigma Receptors. London: Academic Press, 1994; 45-111. 2 Extensive reviews of sigma receptor pharmacology, biochemistry and physiology are presented in: Walker JM,.Bowen WD, Walker FO, Matsumoto RR, de Costa B Rice KC. PharmacoL Rev. 1990; 542: 355-402. 3 Mewshaw RE, Sherill RG, Mathew RM, Kaiser C, Bailey MA, Karbon EW. J. Med. Chem. 1993; 36: 343-352. 4 Bertha CM, Mattson MV, Flippen-Anderson JL, Rothman RB, Xu H, Cha XY, Becketts K.; Rice KC. J. Med. Chem. 1994; 37: 3163-3170. 5 de Costa BR, He XS, Dominguez C, Cutts J, Williams W, Bowen WD. J. Med. Chem. 1994; 37: 314-321. 6 Lee KS, Weinberger, D et al., unpublished. 7 Kavanaugh MP, Tester BC, Scherz MW, Keana JFW, Weber E. Proc. Natl. Acad. Sci. USA. 1988; 85: 2844-4288. 8 Paul IA, Kuypers G, Youdim M, Skolnick P. Eur. J. Pharmacol. 1990; 184: 203-204.
Perspective in Receptor Research D. Giardin~, A. Piergentili and M. Pigini (Editors) 9 1996 Elsevier Science B.V. All rights reserved.
321
T e t r a a m i n e s as lead c o m p o u n d s for the design of n e u r o t r a n s m i t t e r r e c e p t o r ligands: focus on a-adrenergic and m u s c a r i n i c receptors recognition C. Melchiorre a, P. Angelib, M.L. Bolognesia, R. Budriesi a, S. Cacciaguerra c, A. Chiarini a, M. Crucianelli b, D. Giardin~ b, U. Gulinib, G. Maruccib, A. Minarini a, S. Spampinato c and V. Tumiatti a a Department of Pharmaceutical Sciences, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy b Department of Chemical Sciences, University of Camerino, Via S. Agostino 1, 62032 Camerino (MC), Italy c Department of Pharmacology, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
A b s t r a c t . The concept that polyamines may represent a passe-partout (master key) for G-protein-coupled receptors recognition is illustrated. Using a linear t e t r a a m i n e as the focus, appropriate structural modifications have allowed to obtain novel tetraamines displaying selectivity for different receptor systems. It is shown that tetraamines have receptor subtype selectivity since they are site-directed, owing to different chain lengths separating the protonable nitrogens and to the presence of those peculiar structural elements which make them able of discriminating at the binding stage.
INTRODUCTION The advent of molecular biological techniques has made possible to identify and chemically characterize many receptor systems. It has been demonstrated that a high percentage of homology exists not only within receptor subtypes but also among different receptor families. For example, a comparison of the t r a n s m e m b r a n e domains of G - p r o t e i n - c o u p l e d receptors reveals t h a t muscarinic receptor subtypes have more than 70% identity, whereas aadrenoreceptor subtypes show a lower identity (about 40%). However, aadrenoreceptor subtypes share about 45% identity with serotoninergic and dopaminergic receptors [1]. It is evident t h a t the high homology among receptors accounts for the difficulty to achieve receptor selectivity which remains one of the most fascinating challenge to medicinal chemists. In fact, little is known about the structural elements that determine selectivity to one receptor subtype, rather than to another because only subtle and unpredictable differences in the binding pockets would account for selective binding for certain ligands.
322 TETRAAMINES AND R E C E P T O R R E C O G N I T I O N Although receptor homology makes inherently difficult to achieve receptor selectivity it may be used indeed to design molecular entities which are able, in principle, to recognize different receptor systems. This working hypothesis derives from the consideration that n e u r o t r a n s m i t t e r receptors are folded polypeptide chains which contain always the same amino acids, albeit in a different proportion and sequence. The sequence of the amino acids constitutes the primary structure which derives by peptide bonds linking the carboxylate groups to the amino groups. The resulting repeat units in the chain, i.e. the peptide bond and the a carbon, form the backbone. Since the backbone of a protein has only a structural role and cannot be considered a target for selectivity, it is evident that only lateral chains play a major role in drugreceptor binding. Among these functionalities aspartate and/or glutamate residues may acquire p a r a m o u n t importance for the binding with cationic ligands by way of a cation-anion interaction. Considering that proteins may bear several carboxylate residues, somewhere in their structure, in principle, it is possible to design a lead compound having a polyamine backbone which is able to recognize multiple anionic sites of a given receptor. Thus, such a ligand may interact with all receptor proteins, provided that the distance separating the amine functions of the ligand fits the distance between the carboxylate residues of the receptor. In other words a polyamine could be considered a "passe-partout" in the drug-receptor recognition process. The question is: What structural parameters of the polyamine backbone do receptor systems recognize in order to respond so differently to such subtle structural differences? Clearly, it is necessary to define the structural elements which cause selectivity for a given receptor rather than to another. It was reported t h a t polyamines, such as spermine, are highly protonated at physiological pH. It turned out that spermine is 97% tetracation, the remainder being the trication [2]. Thus, the ability of a polyamine to interact with biological counterions, that is a set of carboxylate anions fixed to the backbone of a receptor, could well be related to its cationic properties. Consequently, the distance between the cationic nitrogens of a polyamine becomes critical in the recognition step. It is known that increasing the number of interactions t h a t take place between a receptor and a ligand there is more chance to distinguish receptor systems. Thus, an appropriate modification of the chain length separating the nitrogens of a polyamine might give rise to an increase of selectivity, whereas the insertion of N - s u b s t i t u e n t s might improve the affinity by increasing the overall number of contacts between a dl~ug and a receptor. Many naturally occurring or synthetic polyamines with a linear tetraamine backbone are known. These ligands recognize different receptors which may be either activated or inhibited. For example, synthetic tetraamine disulfides proved to be a-adrenoreceptor or neuropeptide Y receptor antagonists [3-5], whereas synthetic polymethylene tetraamines resulted potent and selective muscarinic receptor subtype antagonists [4, 6, 7]. Furthermore, it has been demonstrated that the naturally occurring tetraamine spermine and analogues modulate NMDA receptor function [8]. The polyamine NMDA receptor site may have either agonistic or antagonistic modulatory activity depending on the
323 concentration and the chemical structure of the polyamine. In addition, N alkylated spermine analogues displayed antineoplastic activity against a number of tumor lines [9] and, in general, it can be stated t h a t natural polyamines are in many ways involved in the complex phenomena related to host defense mechanisms [10]. T E T R A A M I N E S VS a - A D R E N O R E C E P T O R S a l - A d r e n o r e c e p t o r s are not homogeneous as clearly evidenced by pharmacological tools and molecular biological techniques [11]. Two native aladrenoreceptor subtypes, that is alA and ~lS, have been so far characterized by functional and binding assays. The ~IA subtype has high affinity for antagonists such as WB 4101, 5-methylurapidil and (+)-niguldipine, and is insensitive to inactivation by chloroethylclonidine (CEC). The alB subtype displays low affinity for the above antagonists, but is preferentially inactivated by the alkylating agent CEC [11]. However, very recently it has been demonstrated that the aladrenoreceptor mediating contraction upon activation in isolated rat aorta may belong to the alp subtype [12, 13]. Cloning studies have confirmed the existence of at least three distinct al-adrenoreceptors, namely the alb, alc and ald subtypes [14-16]. The cloned alb-adrenoreceptor corresponds to the native alB type whereas the alc-adrenoreceptor is similar to the pharmacologically defined alAadrenoreceptor and for this reason it has been named Ula [17, 18]. Thus, aladrenoreceptors have been subdivided into alA (CY.la), (ZlS (alb) and O~ld subtypes [17]. Although the cloned ~qd-adrenoreceptor is not well characterized yet may find a correspondence with the pharmacologically defined aid subtype in rat aorta [12, 13]. Design of novel tetraamine disulfides based on benextramine and prazosin
Tetraamine disulfides, whose main structural feature is a cystamine moiety carrying amino alkyl substituents on the nitrogen atoms, represent a class of non-competitive antagonists of both al- and ~2-adrenoreceptors [3, 4]. It was shown that al-adrenoreceptor inhibition by tetraamine disulfides is the result of covalent bond formation between a receptor target thiol and the disulfide bridge of the antagonist through a disulfide-thiol interchange reaction [3, 4, 19, 20]. It was also found that optimum activity is associated with well defined chain lengths separating the inner from the outer nitrogens and the inner nitrogens from the sulphur atoms, and depends on the type of substituents on the terminal nitrogens [3, 4]. Benextramine resulted the prototype of this class of irreversible antagonists which bears no structural kinship with the other known a-adrenoreceptor antagonists. It has been fairly well investigated in both functional studies and binding assays [4]. Prazosin is a selective competitive al-adrenoreceptor antagonist widely used not only as a pharmacological tool for a l - a d r e n o r e c e p t o r subtypes c h a r a c t e r i z a t i o n but also as an effective agent in the m a n a g e m e n t of h y p e r t e n s i o n . Its a n t i h y p e r t e n s i v e activity depends on a p e r i p h e r a l vasodilatation mediated by a postjunctional ~l-adrenoreceptor blockade. Moreover, given its high al-selectivity, prazosin lacks side effects such as
324 t a c h y c a r d i a and hypereninemia, which are connected with a presynaptic a2adrenolytic action [21-24]. It has been advanced that benextramine and prazosin may interact with the very same site where norepinephrine binds. On the basis of substituents effects and t a k i n g advantage of the finding t h a t norepinephrine at a relatively low concentration (30 btM) afforded complete protection of aortic al-adrenoreceptors s
iI
ss ~
~%%
%
NH 2 ~
,
"r'f" 0.05), unless otherwise specified, pKi values were derived using the Cheng-Prusoff equation [30]. pKi values are not given for b e n e x t r a m i n e because the ICso values contain a kinetic t e r m for irreversible binding to the [3H]prazosin binding sites. Nonspecific binding was assessed in the presence of 10 pM phentolamine, aN.A. = not active up to a concentration of 50 ~M. eHill numer was significantly different from unity (see Table 2).
327 as the reference compounds, al-Adrenoreceptor blocking activity was assessed by antagonism of (-)-norepinephrine-induced contractions of the epidydimal portion of the vas deferens, whereas a2-blocking activity was determined by antagonism of the clonidine-induced depression of the twitch responses of the field-stimulated prostatic portion of vas deferens as reported previously [28, 31]. We chose to examine the receptor subtype selectivity of selected tetraamines 3, 5-9 by employing receptor binding assays. [3H]Prazosin was used to label alAand alB-adrenoreceptors binding sites of rat submaxillary gland and liver homogenates, respectively. Prazosin, benextramine and the alB-selective antagonist spiperone [11] were used as the standard compounds. The results obtained at rat vas deferens e.-adrenoreceptors are shown in Table 1. Surprisingly enough, all hybrid tetraamines 1-10, unlike benextramine, did not inhibit irreversibly ul-adrenoreceptors, r a t h e r they antagonized competitively n o r e p i n e p h r i n e - i n d u c e d responses like prazosin. However, t e t r a a m i n e s 1-4 inhibited irreversibly, although with a significantly lower potency than benextramine, clonidine-induced responses at a2-adrenoreceptors. This remarkable difference in the observed antagonism at al-adrenoreceptors may have two main sources: a) hybrid tetraamines 1-10 and benextramine may react with two distinct sets of sites or b) they may bind with the same set but t e t r a a m i n e s 1-10 are not able to unmask the buried receptor thiol thus preventing the disulfide-thiol interchange reaction which would lead to irreversible inactivation of the receptor. Unfortunately, it is not possible to distinguish yet between these two possibilities. An analysis of the results reveals t h a t t e t r a a m i n e s 1-10 displayed a significant potency, albeit lower t h a n t h a t of prazosin, toward a ladrenoreceptors whereas, again in contrast with benextramine, they showed only a modest, if any, affinity for a.2-adrenoreceptors. In fact, hybrid tetraamines displayed a significant selectivity (50-1000-fold) for a a - v e r s u s a2adrenoreceptors whereas benextramine showed an a2-selectivity (about 10-fold). Clearly, the replacement of the 2-methoxybenzyl group of benextramine with a quinazoline nucleus affording tetraamines 1-10 alters significantly the biological profile in comparison to benextramine. Concerning the chain length effects, it turned out that optimum potency at e.l-adrenoreceptors was associated with a six-carbon chain as in 3 although the homologues 2 and 5 retained the same level of activity. However, at a2-adrenoreceptors the chain length effects were more pronounced since either a shorter or a longer carbon chain t h a n six methylenes caused a significant drop in activity. Interestingly, N-methylation of the outer nitrogens of 3, 5, 7 and 9 affording the corresponding analogues 4, 6, 8 and 10 caused an increase in affinity for ~.1-but not ~2-adrenoreceptors. This finding parallels the results observed for prazosin analogues bearing a tertiary amine function at position 2 of the quinazoline moiety [28]. Tetraamines 4, 6, 8 and 10 resulted almost equiactive to each other suggesting that the chain length separating the inner from the outer nitrogens may not play an important role in affinity. Tetraamine 8 turned out to be even more selective, although 4 times less potent, than prazosin toward al- versus ~2-adrenoreceptors. Affinity constants, expressed as pKi values, for hybrid t e t r a a m i n e s and reference compounds at ~aa- and ~.lB-adrenoreceptor subtypes are shown in Table 1. These data indicate that, with the exception of 8, hybrid tetraamines
328 are alB-selective, whereas prazosin although being the most potent is not able to distinguish the two subtypes. The greatest selectivity was seen for tetraamine 5 that dispalyed an l l - f o l d selectivity for alB" versus alA-adrenoreceptors. This finding does not parallel the alA-selectivity shown by benextramine [27, this study] indicating that 2-methoxybenzyl and quinazolinyl groups play a different role on selectivity. Interestingly, spiperone, which is considered a selective alBadrenoreceptor antagonist [11], showed only a 4-fold higher affinity at alBadrenoreceptors and a slightly, higher (2-fold) alS/alA selectivity t h a n 5. However, spiperone may not represent a useful tool for the characterization of alB-adrenoreceptors because of the lack of receptor specificity owing to its high affinity for dopamine D2 and 5-HT1A receptors as well. The slopes of the competition curves obtained with hybrid tetraamines, prazosin and spiperone were not significantly different from unity, indicating t h a t the sites labeled with [3H]prazosin in rat submaxillary gland (alAadrenoreceptors) and liver (alB-adrenoreceptors) homogenates are essentially homogeneous. However, in the case of the binding competition curves with t e t r a a m i n e s 3, 6 and 9 in liver membranes, the slopes were significantly different from unity and the data were best fitted to a two-site model, revealing a high affinity site and a low affinity site (Table 2). Clearly, these results do not agree with the view that rat liver represents a pure als-adrenoreceptor tissue [32]. Using the rat alb-adrenoreceptor probe two mRNA species of similar sizes in liver were detected [16, 18, 33]. It has been advanced [18] that the detection of two different alB-adrenoreceptor subtypes in rat liver raises the possibility that such alb-adrenoreceptor isoforms could exhibit different pharmacological profiles. This hypothesis might explain the affinity profile displayed by tetraamines 3, 6 and 9 which are able to distinguish markedly between high and low affinity sites (Table 2). Table 2 Two-site analysis of the interaction of tetraamines 3, 6 and 9 with aladrenoreceptor binding sites in rat liver membranes a High
Low
no.
nH b
pKi
%
pKi
%
high I low c
3
1.19 1.35 0.58
8.12 8.31 7.81
72 47 62
7.30 7.19 6.65
28 53 38
7 13 14
6
9
aData from Table 1 and fitted to a two-site model of binding, bHill numbers (nil) were significantly different from unity (p < 0.05). cThe high/low selectivity ratio is the antilog of the difference between the pKi values at high and low affinity binding sites, respectively.
329 TETRAAMINES VS MUSCARINIC R E C E P T O R S The identification of multiple muscarinic receptor subtypes has stimulated the search for ligands with selectivity for a given receptor subtype [34-36]. Five different subtypes (ml-ms) have been identified so far by molecular cloning. Muscarinic receptors that have been characterized pharmacologically and classified as M1-M4, appear to correspond to cloned ma-n~ receptors. At present, little information is available about the nature and the cellular location of the m5 subtype. Achievement of selectivity is of paramount importance not only for receptor subtype characterization but also for the development of therapeutically useful muscarinic agonists and antagonists [35, 37, 38]. For instance, muscarinic M1 receptor agonists and muscarinic M2 receptor antagonists may be useful to enhance cognitive function [38], whereas muscarinic M3 receptor antagonists have potential for the treatment of airway obstruction. Furthermore, muscarinic M2 receptor antagonists may have application in the treatment of bradycardic disorders. Presently, several antagonists which bind selectively to the pharmacologically characterized muscarinic receptors are available. For example, pirenzepine [39] has high affinity for muscarinic M1 receptors which are mainly located in the central nervous system and peripheral ganglia; polymethylene tetraamines, exemplified by methoctramine [4, 6, 7], display high affinity for muscarinic M2 receptors of cardiac cells, whereas 4-DAMP [40], hexahydrosiladifenidol and its p-fluoro derivative [41-43] show high affinity for muscarinic M3 receptors located in smooth muscle and exocrine glands. Finally, MT3, a toxin isolated from the venom of green mamba, is highly selective for muscarinic M4 receptors [44]. D e s i g n of M1 o a n d Mz-selective t e t r a a m i n e s b a s e d on m e t h o c t r a m i n e Polymethylene tetraamines are a class of competitive antagonists of muscarinic receptors. Their development started from the observation that benextramine also displayed muscarinic antagonistic activity with a significant selectivity for cardiac muscarinic receptors. Using benextramine as the focus, polymethylene tetraamines lacking a disulfide moiety were designed to achieve specific recognition of muscarinic receptors. Detailed structure-activity relationship studies revealed that both affinity and selectivity for muscarinic receptor subtypes are heavily dependent on either the chain length separating the four amine functions or the type of substituent on the nitrogen atoms of polymethylene tetraamines. The development of polymethylene tetraamines, the prototype of which is methoctramine, as muscarinic M2 receptor antagonists has been the subject of review articles [4, 6, 7]. The finding that methoctramine, like all other M2 selective antagonists available, failed to discriminate between muscarinic M2 and M4 receptors, thus preventing a clear pharmacological characterization of muscarinic receptor subtypes, stimulated a research in order to improve affinity and selectivity for muscarinic M2 receptors by modifying methoctramine structure. The starting point of this investigation was the observation t h a t the two amine functions of AQ-RA 741 (11-[[4-[4(diethylamino)-butyl]- 1-piperidinyl]acetyl]-5,11-dihydro-6H-pyrido[2,3-b][ 1,4] benzodiazepin-6-one), an analogue of pirenzepine displaying selectivity for muscarinic M2 receptors [45], are located to such a distance that they can be
330 superimposed to one inner and one outer nitrogen of methoctramine when comparison is made between the extended conformations of the two prototypes. The observation t h a t diamines, obtained by truncating in two halves methoctramine structure and which are closely related to AQ-RA 741, resulted in analogues almost devoid of affinity and selectivity toward M2 muscarinic receptors was another consideration of paramount importance in the design strategy of our compounds [46, 47]. Clearly, an ll-acetyl-5,11-dihydro-6Hpyrido[2,3-b][1,4]-benzodiazepin-6-one moiety recognizes better t h a n a 2methoxybenzyl group the binding pocket of muscarinic M2 receptors. Thus, we thought that the insertion of a tricyclic system on the terminal nitrogens of a tetraamine would improve affinity for muscarinic M2 receptors. To prevent Nalkylation of the two inner nitrogens, we choose as a common backbone a tetraamine bearing on these nitrogens a methyl group. Furthermore, this choice was dictated by a previous finding that methylation of inner nitrogens of methoctramine affording N,N'-dimethyl methoctramine does not affect both affinity and selectivity for muscarinic M2 receptors [48]. Thus, a series of tetraamines were designed by replacing the terminal 2-methoxybenzyl groups of m e t h o c t r a m i n e with an ll-acetyl-5,11-dihydro-6H-pyrido[2,3-b][1,4]benzodiazepine-6-one moiety [49]. Tripitramine resulted the most potent and the most selective muscarinic M2 receptor antagonist so far available, able to discriminate also between muscarinic M2 and M4 receptor subtypes. The finding that the affinity profile of methoctramine-related tetraamines depends on the type of substituents on the terminal nitrogens of a tetraamine backbone, was the basis for a further modification of methoctramine structure, in order to improve the affinity and selectivity for different muscarinic receptor subtypes. Thus, we designed new tetraamines by substituting the 2methoxybenzyl groups of methoctramine with the structural features of 4DAMP, a relatively selective muscarinic M1/M3 receptor antagonist, and of its analogues spiro-DAMP and hydroxy-DAMP [50]. This study afforded spirotramine that displayed an inverse selectivity profile in comparison to both methoctramine and tripitramine owing to a higher affinity for muscarinic M1 receptors and a significantly lower affinity for all other muscarinic receptor subtypes. The structures of these novel tetraamines are shown in Figure 2 together with those of the parent compounds. Biological profile of novel tetraamines at muscarinic receptor subtypes Functional activity at muscarinic receptor subtypes was determined by the use of the muscarinic M2 receptor-mediated negative inotropism in driven guinea pig left atria (1 Hz) and muscarinic M3 receptor-mediated contraction of guinea pig ileum longitudinal muscle. The muscarinic receptor subtype selectivity was assessed by employing receptor binding assays [49, 51]. [3H]N-methylscopolamine was used to label native muscarinic M2, M3 and M4 receptors binding sites of rat heart, submaxillary gland, and NG 108-15 cell homogenates, respectively, and to label cloned human muscarinic ml-m5 receptors expressed in chinese hamster ovary cells. [3H]Pirenzepine was the tracer to label muscarinic M1 receptors binding sites of rat cerebral cortex.
331
O -COCH 2-N~
~~-'--(CH2)4NEt2
H'N ~ N AQ-RA 741 I~ CH2NH(CH2)6NH(CH2)sNH(CH2)6NHCH2 OMe
MeO Methoctramine
O
Me Me / -C OCH2NH(CH2)6N(CH2)sN(CH2)6N~ CH2CO--N
Tripitramine 0
d\/
Me (CH2)6N(CH2)4
Spirotramine Figure 2. Chemical structures of AQ-RA 741 and of tetraamines methoctramine, tripitramine and spirotramine.
332 Table 3 Antagonist affinities of t r i p i t r a m i n e and spirotramine in comparison to reference compounds a) at muscarinic receptor subtypes in guinea pig left atria (Ma) and ileum longitudinal muscle (M3), and b ) f o r inhibiting [3H]Nmethylscopolamine binding at native muscarinic receptors of rat heart (M2), rat submaxillary gland (M3), NG 108-15 cell (M4) and at cloned human muscarinic m 1-ms receptors, and for inhibiting [3H]pirenzepine binding at native muscarinic M1 receptors of rat cerebral cortex
pA2a
pKi b
M2
M3
M1/ ml
M~ m2
M3/ m3
MJ m4
Methoctramine c
7.82
6.32
Tripitramine c
9.75
6.55
Spirotramine a AQ-RA 741 c 4-DAMPa Pirenzepine c
5.36 8.03 8.53