DRUG DISCOVERY AND DESIGN: MEDICAL ASPECTS
Biomedical and Health Research Volume 55 Earlier published in this series Vol. 22. J. Matsoukas and T. Mavromoustakos (Eds.), Bioactive Peptides in Drug Discovery and Design: Medical Aspects Vol. 23. M. Hallen (Ed.), Human Genome Analysis Vol. 24. S.S. Baig (Ed.), Cancer Research Supported under BIOMED 1 Vol. 25. N.J. Gooderham (Ed.), Drug Metabolism: Towards the Next Millennium Vol. 26. P. Jenner (Ed.), A Molecular Biology Approach to Parkinson's Disease Vol. 27. P.A. Frey and D.B. Northrop (Eds.), Enzymatic Mechanisms Vol. 28. A.M.N. Gardner and R.H. Fox, The Venous System in Health and Disease Vol. 29. G. Pawelec (Ed.), EUCAMBIS: Immunology and Ageing in Europe Vol. 30. J.-F. Stoltz, M. Singh and P. Riha, Hemorheology in Practice Vol. 31. B.J. Njio, A. Stenvik, R.S. Ireland and B. Prahl-Andersen (Eds.), EURO-QUAL Vol. 32. B.J. Njio, B. Prahl-Andersen, G. ter Heege, A. Stenvik and R.S. Ireland (Eds.), Quality of Orthodontic Care - A Concept for Collaboration and Responsibilities Vol. 33. H.H. Goebel, S.E. Mole and B.D. Lake (Eds.), The Neuronal Ceroid Lipofuscinoses (Batten Disease) Vol. 34. G.J. Bellingan and G.J. Laurent (Eds.), Acute Lung Injury: From Inflammation to Repair Vol. 35. M. Schlaud (Ed.), Comparison and Harmonisation of Denominator Data for Primary Health Care Research in Countries of the European Community Vol. 36. F.F. Parl, Estrogens, Estrogen Receptor and Breast Cancer Vol. 37. J.M. Ntambi (Ed.), Adipocyte Biology and Hormone Signaling Vol. 38. N. Yoganandan and F.A. Pintar (Eds.), Frontiers in Whiplash Trauma Vol. 39. J.-M. Graf von der Schulenburg (Ed.), The Influence of Economic Evaluation Studies on Health Care Decision-Making Vol. 40. H. Leino-Kilpi, M. Valimaki, M. Arndt, T. Dassen, M. Gasull, C. Lemonidou, P.A. Scott, G. Bansemir, E. Cabrera, H. Papaevangelou and J. Mc Parland, Patient's Autonomy, Privacy and Informed Consent Vol. 41. T.M. Gress (Ed.), Molecular Pathogenesis of Pancreatic Cancer Vol. 42. J.-F. Stoltz (Ed.), Mechanobiology: Cartilage and Chondrocyte Vol. 43. B. Shaw, G. Semb, P. Nelson, V. Brattstrom, K. M01sted and B. Prahl-Andersen, The Eurocleft Project 1996-2000 Vol. 44. R. Coppo and Dr. L. Peruzzi (Eds.), Moderately Proteinuric IgA Nephropathy in the Young Vol. 45. L. Turski, D.D. Schoepp and E.A. Cavalheiro (Eds.), Excitatory Amino Acids: Ten Years Later Vol. 46. I. Philp (Ed.), Family Care of Older People in Europe Vol. 47. H. Aldskogius and J. Fraher (Eds.), Glial Interfaces in the Nervous System - Role in Repair and Plasticity Vol. 48. H. ten Have & R. Janssens (Eds.), Palliative Care in Europe - Concepts and Policies Vol. 49. T. Reilly (Ed.), Musculoskeletal Disorders in Health-Related Occupations Vol. 50. R. Busse, M. Wismar and P.C. Berman (Eds.), The European Union and Health Services Vol. 51. G. Lebeer (Ed.), Ethical Function in Hospital Ethics Committees Vol. 52. J.-F. Stoltz (Ed.), Mechanobiology: Cartilage and Chondrocyte, Vol. 2 Vol. 53. In production Vol. 54. I.M. Shapiro, B.D. Boyan and H.C. Anderson (Eds.), The Growth Plate ISSN: 0929-6743
Drug Discovery and Design Medical Aspects Edited by
J. Matsoukas Department of Chemistry, University of Patras, Patras, Greece
and
T. Mavromoustakos Institute of Organic and Pharmaceutical Chemistry, National Hellenic Research Foundation, Athens, Greece
IOS
Press
•11 Ohrrisha
Amsterdam * Berlin • Oxford • Tokyo • Washington, DC
© 2002, The authors mentioned in the Table of Contents All rights reserved. No part of this book may be reproduced, stored in a retrieval system, or transmitted, in any form or by any means, without the prior written permission from the publisher. ISBN 1 58603 261 5 (IOS Press) ISBN 4 274 90522 5 C3047 (Ohmsha) Library of Congress Control Number: 2002106945
Publisher IOS Press Nieuwe Hemweg 6B 1013 BG Amsterdam The Netherlands fax: +31 20 620 3419 e-mail:
[email protected]
Distributor in the UK and Ireland IOS Press/Lavis Marketing 73 Lime Walk Headington Oxford OX3 7AD England fax: +44 1865 75 0079
Distributor in the USA and Canada IOS Press, Inc. 5795-G Burke Centre Parkway Burke, VA 22015 USA fax: +1 703 323 3668 e-mail:
[email protected]
Distributor in Germany, Austria and Switzerland IOS Press/LSL.de Gerichtsweg 28 D-04103 Leipzig Germany fax: +49 341 995 4255
Distributor in Japan Ohmsha, Ltd. 3-1 Kanda Nishiki-cho Chiyoda-ku, Tokyo 101-8460 Japan fax: +81 3 3233 2426
LEGAL NOTICE The publisher is not responsible for the use which might be made of the following information. PRINTED IN THE NETHERLANDS
Preface This volume is a potpourri of review articles and research articles related to the "Bioactive Drugs in Drug Discovery and Design". It can be regarded as a continuation of Volume 22 published by IOS Press in the series Biomedical and Health Research. Most of the articles are part of the lectures given in the course of "Medicinal Chemistry" for graduate students. The book, like the first volume, is divided into four different topics in order to help the reader locate easily the manuscript of his interest. The first topic covers the synthesis and properties of bioactive molecules and enzymes. The second part examines the role of membranes in drug activity and formulation. The book continues with the third part that covers the conformational analysis of bioactive drugs. The final part mainly touches aspects of the molecular targets and drug design. This part is actually broader in scope and covers biological aspects of medicinal chemistry. The logic of classification of these parts is related to the route that medicinal chemistry follows. Novel compounds are synthesized which their conformations, and their interactions with the active site are studied. Finally, these compounds are undertaken for biological test. Meanwhile, new aspects like proteomics are developing in parallel to enhance the knowledge of the molecular basis of drug action. While the major part of the book is written by Greek specialists in their field several other articles come from researchers from different countries in order to assist the reader to get a global understanding of the recent activities in the field of Drug Discovery and Design. The editors wish to express their deep appreciation to all contributors in this book. In particular, to Rafarm Pharmaceutical Company, to the authors for their collaboration, to Panagiotis Zoumpoulakis for his assistance in editing the book, to George Zoumpoulakis for his artistic cover and IOS Press for providing the suitable and high-standard forum through which important findings of this research will become available to the scientific community. The Editors J. Matsoukas T. Mavromoustakos
The cover illustration was painted by the artist G. Zoumpoulakis. The revival of the plant is a metaphor for the healing of humans with the help of drugs.
Contents Preface
v
Part I: Synthesis and Properties of Bioactive Compounds Design, Synthesis and Biological Evaluation of Cyclic Angiotensin II Analogues with 3,5 Side-Chain Bridges: Role of C-Terminal Aromatic Residue and Positions 3,5 for Activity, L. Polevaya, P. Roumelioti, T. Mavromoustakos, P. Zoumpoulakis, N. Giatas, I. Mutule, T. Keivish, A. Zoga, D. Vlahakos, E. Iliodromitis, D. Kremastinos and J. Matsoukas 3 Synthesis of RGD Analogs Incorporating Moiety of Salicylic Acid Derivatives and their Effect on Human Platelet Aggregation In Vitro, Y. Sarigiannis, G. Stavropoulos and M. Liakopoulou-Kyriakides 13 Synthesis of Substance P C-terminal Analogs Incorporating D-Amino Acids and Studies of their Antineoplastic Properties in Vitro, P. Vakalopoulou, G. Stavropoulos, M. Liakopoulou-Kyriakides, Z. lakovidou and E. Mioglou 20 Synthesis of Chiral Tetramic Acids Using Activated Derivatives of N-protected a-Amino Acids as Precursors. Structural Characterization of Tetramic Acid Complexes with Metal Ions, J. Markopoulos, A. Detsi, E. Gavrielatos, G. Athanasellis and O. Igglessi-Markopoulou 25 L-Asparaginase: Structure, Properties and Anti-Tumor Activity, A.A. Pritsa and D.A. Kyriakidis 33 Growth Inhibitory and Apoptotic Effects of Gonadotropin-Releasing Hormone (GnRH) on Human Hepatocellular Carcinoma, C. V. Andreu-Vieyra and H.R. Habibi 41 Recent Applications of the Amide Approach to the Synthesis of Medicinally Interesting Polyamine Analogues, N. Tsiakopoulos, S. Vassis, M. Militsopoulou, C. Damianakos, P. Gatos, K. Voyiatzi, E. Pantazaka and D. Papaioannou 53 Design and Synthesis of Fluorescent Steroidal Lipopolyamine Conjugates for Monitoring Gene Delivery, I.S. Blagbrough and A.P.Neal 64 Synthesis of (S)-2-Amino-oleic Acid and Other Bioactive Molecules through Glutamate Semialdehydes, V. Constantinou-Kokotou, V. Magrioti, T. Makridis and G. Kokotos 76 Development of New Methods for Fluorometric Assays of Enzymes, C. Tzougraki 83 Enhancement of the Cytogenetic and Cytostatic Activity of Chlorambucil after its Esteric Connection with a Hecogenin Lactam, V. Karayianni, Ch. Camoutsis, V. Soulakis, D. Mourelatos, E. Mioglou and Z. lakovidou 97 In Searching for New Anti-Influenza A Drags: Heterocyclic and Carbocyclic Aminoadamantanes Bearing a 2-Adamantyl Group with Potent Activity Against Influenza A Virus, N. Kolocouris, A. Kolocouris, G.B. Foscolos, G. Fytas, E. Padalko, J. Neyts and E. De Clercq 103 Design, Synthesis and Conformational Properties of Linear Analogues of Human Myelin Basic Protein Epitope MBP1-11, S. Deraos, T. Tselios, I. Daliani, P. Zoumpoulakis, L. Probert, A. Troganis, P. Papathanassopoulos, T. Mavromoustakos and J. Matsoukas \ 16
Part II: Role of Membranes in Drug Activity The Effects of Vinblastine Sulfate on Dipalmitoylphosphatidylcholine Single and Multiple Bilayer Membranes, H. Maswadeh, C. Demetzos, I. Daliani, T. Mavromoustakos, G. Nounesis and A. Tsortos Stealth Liposomal Vinca Alkaloids (Vinorelbine and Vincrastine) and In Vitro Studies on Release by Buffer and Rat Plasma, C. Demetzos Structural Elucidation, Conformational Properties and Effects in Membrane Bilayers of the Toxin Paralysin, -Ala-Tyr from Larvae of the Gray Flesh Fly, /. Kyrikou, T. Mavromoustakos and C. Poulos Effects of NSAIDs in Membrane Bilayers, T. Mavromoustakos, I. Kyrikou, A. Kapou and D. Kovala-Demertzi
\ 25 131
142 150
Part III: Conformational Analysis of Bioactive Compounds The Structure of Endotoxin-Neutralizing Peptides Bound to LPS, P. Pristovsek and J. Kidric Synthesis and Conformational Analysis of Steroidal Carriers of Alkylating Agents with the Use of Two-Dimensional Nuclear Magnetic Resonance, A. Kapou, T. Mavromoustakos, S.G. Grdadolnik and S. Nikolaropoulos Structure Elucidation and Conformational Properties of Irbesartan, P. Zoumpoulakis, M. Zervou, S.G. Grdadolnik and T. Mavromoustakos 1 H and 13CNMR Assignment and Conformational Studies of a Myelin Basic Protein Epitope 74–85 Implicated in Multiple Sclerosis, A. Tzakos, A.N. Troganis, T. Tselios, A. Bonvin, N. van Nuland, S. Deraos, J. Matsoukas and IP. Gerothanassis Conformational Analysis of Biomolecules in Solution: NMR - 3D Structures Biological Implications, G.A. Spyroulias, S. Papazacharias, A. Galanis and P. Cordopatis Synthesis and Structural Investigation of a Synthetic LHRH Analogue in Solution, A.A. Zompra, G.A. Spyroulias, V. Magafa and P. Cordopatis Synthesis, Biological Evaluation & NMR Solution Structural Models of New Oxytocin Analogues, M. Fragiadaki, S. Koumentakos, D. Raptis, G.A. Spyroulias, V. Magafa, J. Slaninova and P. Cordopatis
161 167 174
180 188 205 217
Part IV. Molecular Targets and Drug Design Molecular Aspects of Human Abdominal Aortic Aneurysm: Differential Expression of Genes Coding for Extracellular Matrix Proteoglycans, A.D. Theocharis and N.K. Karamanos Advances in Botulinum Neurotoxin Inhibitors Based on the SNARE Motif, G.J. Moore, D. Moore, S. Roy, L.J. Hayden and G. Murray Effect of Linear and Cyclic Peptide Analogues of Myelin Basic Protein Epitope MBP72-85 on Human T-cell Activation, S. Thymianou, K. Chatzantoni, M. Karakantza, T. Tselios, P. Papathanassopoulos, J. Matsoukas and A. Mouzaki Use of Synthetic Peptides for the Identification of the Biologically Active Domains of the Growth Factor HARP, E. Papadimitriou, A. Polykratis, E. Karestou, J. Courty and P. Katsoris Unusual Interactions between MHC, Peptide and the T cell Receptor, V. Apostolopoulos and M. Plebanski
229 236
241 251 258
Effects of Angiotensin II, III and IV on Memory Retention of Rats: Interaction with Adenosine A1 Receptor Related Ligands, J. Tchekalarova and V. Georgiev Molecular Targets and Compounds for Anti-HIV Therapy, E. De Clercq Proteomics in Drug Discovery: Potential and Limitations, M. Fountoulakis Evaluation of Tumor Capabilities for Recurrence in Patients with Larynx and Pharynx Malignancies on the Basis of DNA Criterion, S. Andreychenko Significance of G-protein-coupled Receptor Polymorphisms: The Case of tt2-adrenergic Receptor, A. S. Manolis, A. Lymperopoulos, E.A. Bouga, M. Scheinin and C.S. Flordellis Computer Graphics Applications on Molecular Biology and Drug Design, K. Perdikuri and A. Tsakalidis Investigation of Novel DNA Gyrase Inhibitors Using the High Resolution NMR Spectroscopy, S.G. Grdadolnik, M. Oblak, T. Solmajer and R. Jerala Author Index
267 272 279 294 299 305 312 323
This page intentionally left blank
Part I.
Synthesis and Properties of Bioactive Compounds
This page intentionally left blank
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Design, Synthesis and Biological Evaluation of Cyclic Angiotensin II Analogues with 3,5 Side-Chain Bridges: Role of C-Terminal Aromatic Residue and Positions 3,5 for Activity L Polevaya1, P. Roumelioti2, T. Mavromoustakos3, P. Zoumpoulakis3, N. Giatas2, I. Mutule 1 , T. Keivish1, A. Zoga4, D. Vlahakos4, E. Iliodromitis4, D. Kremastinos4, J. Matsoukas2 Laboratory of Peptide Chemistry, Latvian Institute of Organic Synthesis, Riga, LV-1006, Latvia 2 Department of Chemistry, University of Patras, 26500 Patras, Greece 3 Section of Organic and Pharmaceutical Section, National Research Institute, Athens, Greece 4 Onassis Cardiac Surgery Center, 17674 Athens, Greece Abstract. The novel amide linked Angiotensin II cyclic analogues: y, e - cyclo(3, 5) -[Sar1Glu3-Lys5] ANG II (1) and y, e - cyclo(3, 5) -[Sar1-Glu3-Lys5-Ile8] ANG II (2) have been designed, synthesized and bioassayed in anesthetized rabbits in order to further unravel structural ring cluster characteristics important for receptor activation. The two analogues were synthesized in particular to investigate differences in activity and conformation upon 1) replacement of aromatic residue Phe with aliphatic Ile at position 8 and 2) reversing the position of the Lys, Glu linking residues at positions 3 and 5. Design was based on previous SAR and NMR studies in which residues 3 and 5 are the least important for activity and therefore could be used to create a link without disturbing the ring cluster conformation needed for activity (4, 5). The constrained cyclic analogues with a lactam amide bridge linking Glu, Lys residues at positions 3 and 5 and with Phe or Ile at position 8, were synthesized by solution procedure using the maximum protection strategy. Analogue 1 with Phe at position 8 was found to be a potent agonist while analogue 2 with He at position 8 was found to be an inhibitor of Angiotensin II. It appears that the aromatic ring cluster (Tyr-His-Phe) in agonist peptides is an essential stereo- electronic feature for Angiotensin II to exert its
L. Polevaya et al. / Cyclic Angiotensin II Analogues
biological activity. A 15th membered ring in the central X3-Tyr4-*5 core (X=Glu, Lys, vF=Lys, Glu) allows this cluster to exert its receptor action.
Introduction The octapeptide Angiotensin II (ANG II, Asp-Arg-Val-Tyr-Ile-His-Pro-Phe) is the main pressor component of the Renin-Angiotensin System (RAS) [1,2]. Accumulated experimental evidence for AngiotensinII supports a bioactive conformation characterized by a charge relay system between Tyr hydroxyl, His imidazole and Phe carboxylate, analogous to that found in serine proteases [3], as well as a ring cluster of the triad key aminoacids Tyr4-His6- Phe8 which appears to be responsible for activity [4,5]. Thus conformational analysis using modern 2D NMR techniques in receptor-simulating environments has shown proximity of the three key aminoacids sidechains and the formation of tyrosinate has been demonstrated by nanosecond time resolved tyrosinate fluorescence studies [6-7]. Comparative nuclear magnetic resonance studies of the backbone structure between peptide agonists and antagonists have shown that only agonists display ring clustering and form a change relay system [8]. In addition the proposed conformation overlays the recently discovered nonpeptide ANG II receptor antagonist Losartan and analogs when molecular modeling techniques and superimposition studies are applied [9,10]. Furthermore, the ring cluster conformation was recently supported by the design and synthesis of a novel constrained ANG II cyclic analogue, [Sar1-Lys3-Glu5] ANG anesthesized rabbits [5]. This potent cyclic analog was designed to have as a major molecular feature the integrity of the ring cluster. Other structure- activity studies have illustrated the importance of the C- terminal aromatic residue Phenylalanine (Phe) for agonist activity. Replacement of residue Phe at position 8 with an aliphatic one, as Ile, results in an antagonist [Sar1-Ile8] ANG II (Sarilesin) [1,2,11]. In this work two cyclic ANG II analogues 1, 2 with Glu, Lys residues at positions 3 and 5 and with Phe or Ile respectively at position 8 have been synthesized. The aims of this work were to investigate furthermore the role of a ring cluster receptor conformation in agonist activity and shed light to intriguing differences in activity and conformation upon 1) replacement of aromatic residue Phe with aliphatic Ile 2) reversing the position of the Lys-Glu sequence in the central amide bridge. While replacement of Phe with Ile at position 8 produces an antagonist, the reverse of the Glu-Lys order to a Lys-Glu one has a minimal effect in the agonist or antagonist activity of cyclic ANG II analogues. A 15th membered ring in the central core X-Tyr-^F allows a ring cluster and activity regardless of the Glu-Tyr-Lys or Lys-Tyr-Glu sequence in the agonist cyclic peptide. Based on these structure activity relationships which demand the presence of Phe, Tyr and His residues in ANG II as well as in linear and constrained ANG II analogues to possess biological activity, it can be inferred that the ability to form a ring cluster and consequently a charge relay system may be the key stereoelectronic molecular features of ANG II for exerting biological activity. These studies show also that cyclization at 3 and 5 positions does not affect the activity of the counterparts linear peptides Results and discussion The limited stability of peptides often severely restricts their medical and industrial application. Therefore, the engineering of stable proteins is of great technological and economic importance. If designed carefully without causing drastic changes in the
L. Polevaya et al. / Cyclic Angiotensin II Analogues
5
conformation of active peptides, the rigid geometry of the cyclic peptides enhances the binding affinity towards a selected target molecule compared to their linear counterparts [12, 13]. Furthermore cyclic analogues are important intermediates in the design and synthesis of non-peptide mimetics with the potential to be used as drugs [14,15]. With this aim, our group has been involved for several years in the design and synthesis of cyclic analogues for important peptides such as, Angiotensin [5, 16, 33], Thrombin Receptor Peptides [17] and Myelin Basic Protein [15] involved respectively in hypertension, cancer and multiple sclerosis. So far, a limited number of conformationally restricted via cyclization Angiotensin II analogues have been reported by us and others. In these studies cyclization was achieved either by the disulfide method using cysteine moieties at various locations of the peptide molecule or by the amide-linkage method [18-30]. Our interest in the conformational model of Angiotensin II, which could be used as a basis for the synthesis of non-peptide receptor antagonists, prompted us to design and synthesize two novel cyclic amide linked Angiotensin II analogues cyclo(3, 5)-[Sar1-Glu3Lys5] ANG II (1) and cyclo(3, 5)-[Sar1-Glu3-Lys5-Ile8] ANG II (2) (Fig. 1) that differ only in residue at position 8. The two analogues furthermore differ from cyclo(3, 5)-[Sar1-Lys3Glu 5 -X 8 ] ANG II (X=Phe or Ile) [31] only in the sequence of the Lys-Glu bridging residues. Both analogues can-potentially pertain the Angiotensin II conformational characteristics (i.e. backbone bend and aromatic ring clustering). It seems that reversal of the Lys-Glu order to Glu-Lys does not affect activity. The synthesis of counterpart novel analogue 2 with Ile of position 8 was prompted to further explore the role of the aromatic residue at position 8 for agonist activity. Cyclization was achieved by forming an amidelinkage between the -NH2 and -COOH side chain groups of Glu and Lys residues at positions 3 and 5, respectively, which are the least important for activity. With this study a confirmation of our Angiotensin II model was sought and further evidence to support the importance of the suggested ring cluster for receptor activation in which the Phe residue at position 8 plays a key role. In addition, it is studied the role of 3,5 aminoacids to affect the agonist or antagonist activity. Our findings indicate the importance of a Phe ring at position 8 for integration of a ring cluster receptor conformation and activity. On the contrary, the positioning of bridging residues Lys-Glu at positions 3 and 5 or 5 and 3 does not affect substantially the degree of potency in the two agonist and two antagonist cyclic peptides. Synthesis of central Glu3 -Tyr 4 -Lys 5 rings: The two cyclic analogues of angiotensin II (AIIand cyclo (3, 5)-Sar-Arg Glu-Tyr-Lys-His-Pro-Ile(I)) and cyclo (3, 5)-Sar-Arg-GluTyr-Lys-His-Pro-Phe (II) were synthesized by solution methods (Figure 1). Design of compounds was based on previous SAR and NMR studies, which has shown that residues 3 and 5 are the least important for activity and therefore could be used to create an amide link without disturbing the ring cluster conformation (4, 5). Peptide Synthesis: In particular, we have prepared two conformationally constrained analogues of AII with y,e-amide bridge between side chains of the glutamic acid and lysine residues at positions 3 and 5, containing an aromatic phenylalanine or aliphatic isoleucine residues in the position 8, respectively. The 3,5-cyclic AII analogues were synthesized by the solution procedure in combination with the maximum protection strategy. These compounds were constructed from N- and C-terminal linear fragments and the central a 15mer cyclic (Glu-Tyr-Lys) part, to produce the fully protected octapeptides, as it is shown in Scheme 1 for cyclo (3, 5)-Sar-Arg-Glu-Tyr-Lys-His-Pro-Ile (I). The same procedure was used to synthesize peptide cyclo (3, 5)-Sar-Arg-Glu-Tyr-Lys-His-Pro-Phe (II) except that tripeptide H-His(Bzl)-Pro-PheONb.HCI (18) was used (see Scheme 1), to obtain the
6
L.
Polevaya et al. / Cyclic Angiotensin II Analogues
protected octapeptide cyclo (3, 5)-Z-Sar-Arg(NO2)-Glu-Tyr-Lys-His(Bzl)-Pro-Phe-ONb (19). The key step in the preparation of each of these analogues was cyclization performed at the tripeptide stage by connecting the side chains of lysine and glutamic acid residues separated by tyrosine residue in the linear Glu3-Tyr4-Lys5 resulting in the formation of a cyclic amide (lactam) bond. The protected linear tripeptide 7 was synthesized stepwise using commercially available HCl.Lys(Z)-OtBu as the starting material.
H3C—NH H
O
HA
O
V
*
H3C—NH
Figure 1. Chemical structures of the Sar-Arg-{y,£-cyclo(Glu-Tyr-Lys)]-His-Pro-Phe (I), Sar-Arg-(y,£-cyclo(Glu-Tyr-Lys)]-His-Pro-Ile (II) analogues of AII, Sar-Arg-[y,e-cyclo(Lys-Tyr-Glu)]-His-Pro-Phe (III) and Sar-Arg-[y,E-cyclo-(Lys-Tyr-Glu)]-His-Pro-Ile(II)
L. Polevaya et al. / Cyclic Angiotensin II Analogues
CENTRAL FRAGMENT C-TERMINAL FRAGMENT
HCl .Lys(Z)-OtBu I Boc-Tyr(Bzl)-OH DCC/HOBt/DMF 1 DIPEA / EtOAc
Boc-Pro-OH HONB / EtOAc DCC HCl, H-Ile-Onb DIPEA
I
AcOH / tert-ButOH 3.7M HCl / tert-ButOH
Z-Sar-Arg(NO2)-Ome (11)
Boc-Pro-Ile-ONb (1) I
Z-Sar-OH
Tyr(Bzl)-Lys(Z)-OtBu (5)
H-Tyr(Bzl)-Lys(Z)-OtBu.HCl (6) I DMF/EtOAc I DIPEA / DMF * Boc-Glu(Bzl)-OPfp Boc-Glu(OBzl>Tyr(Bzl)-Lys(Z)-OtBu (7)
TFA
H-Pro-Ile-ONb.TFA (2)
Boc-His(Bzl)-OH / HOBt D1C/DIPEA
I 1.0M NaOH Z-Sar-Arg(NO)-OH (12)
MeOH / AcOH / H2O Pd black / H2
Boc-Glu-Tyr-Lys-OtBu.AcOH (8) Boc-His(Bzl)-Pro-Ile-ONb (3)
I HOBt / DIC / DIPEA
2.6M HCl / EtOAc
Boc-[ cyclo-(Glu-Tyr-Lys) ]-OtBu (9) Boc-His(Bzl)-Pro-Ile-ONb.HCl (4)
3M HCl / dioxane
[ cyclo-(Glu-Tyr-Lys) ]-OtBu (10) II
12
DIC / HOBt / DIPEA
Z-Sar-Arg(NO2)-[ cyclo-(Glu-Tyr-Lys) ]-OtBu (13) I TFA / DCM
Z-Sar-Arg(NO2)-[ cyclo-(Glu-Tyr-Lys) ]-OH (14) T DCC/HATU / DIPEA Z-Sar-Arg(NO2)-[ cyclo-(Glu-Tyr-Lys) ]-His(Bzl)-Pro-IIe-ONb (15) MeOH / AcOH / H2O H2 / Pd black 1M HCl / dioxane
H-Sar-Arg-[ cyclo-(Glu-Tyr-Lys) ]-His-Pro-Ile-OH.2HCl (I)
L. Polevaya et al. / Cyclic Angiotensin II Analogues Boc-His(Bzl)-OH (16) HBr. H-Pro-Phe-ONb
I
Boc-His(Bzl)-Pro-Phe-ONb (17) 2.9M HCl / EtOAc
I
H-His(Bzl)-Pro-Phe-ONb.HCl (18) 14 , DCC / HATU DIPEA Z-Sar-Arg(NO2)-[ cyclo (Glu-Tyr-Lys)]-His(Bzl)-Pro-Phe-ONb (19) MeOH / AcOH / H2O H2 / Pd black 1M HCl / dioxane
H-Sar-Arg-[ cyclo (Glu-Tyr-Lys)]-His-Pro-Phe-OH.2HCl (II)
Scheme 1. Synthetic routes for the peptides I and II.
The protected cyclic tripeptide Boc-[cyclo(Glu-Tyr-Lys)]-OtBu (9) was prepared after de-protection of the side chains of the bridgehead residues from 7 by coupling the linear precursor 8 via N-hydroxybenzotriazole active ester in situ using DIC/HOBt/ DIPEA. Cyclic dimer (-10%), which formed as a by-product during the cyclization, was removed by HPLC chromatography in the last stage of purification. Selective acidolysis of the N-terminal tert-butyl protecting group in compounds 5 and 9 and de-protection was monitored by regular TLC control in solvent system D. The tert-butyloxycarbonyl group present in 5 was selectively removed by treatment with 3.7 M HCl/tert-ButOH for 25 min at 45 °C in AcOH/tert-ButOH (5:3), whereas tertbutyloxycarbonyl group of 9 was cleaved during treatment with 3M HCl/dioxane for 25 min at room temperature to give the cyclic peptide H-[cyclo(Glu-Tyr-Lys)]-OtBu.HCl (10) in moderate yield (56,7%). tert-Butyloxycarbonyl group de-protection under these conditions, resulted in the formation of small amount of by-product as a result of the simultaneous acidolysis of N- tert-butyloxycarbonyl and tert-butyl ester protections. The N- and C-terminal fragments were synthesized step by step from their respective C-amino acid esters. Z-Sar-Arg(NO2)-OH (12) was obtained by saponification from methyl ester 11. The protected tripeptides Boc-His(Bz)-Pro-Ile-ONb (3) and Boc-His (Bz)-Pro-Phe-ONb (17) were synthesized with p-nitrobenzyl ester protection for the Cterminals and Boc group for the a-amino functions. After tert-butyloxycarbonyl group acidolytic cleavage by HCl/EtOAc, hydrochlorides of the C-terminal tripeptides 4 and 18 were used for preparation of the protected octapeptides 15 and 19, respectively. The fully protected cyclo (3,5)-Z-Sar-Arg(NO2)-Glu-Tyr-Lys-OtBu (13) was prepared by fragment condensation between Z-Sar-Arg(NO2)-OH (12) and cyclo(Glu-TyrLys)-OtBu.HCl (10) using DIC/HOBt carboxyl activation induced by DIPEA. The tertbutyl ester group of pentapeptide 13 was then converted to the free carboxyl by treatment with TFA in DCM for 40 min; The resulting peptide cyclo (3,5)-Z-Sar-Arg(NO2)-Glu-Tyr-
L. Polevaya et al. / Cyclic Angiotensin II Analogues
9
Lys-OH (14) reacted with either H-His(Bz)-Pro-Ile-ONb. HC1 (4) to generate the protected octapeptide 15 or with H-His(Bz)-Pro-Phe-ONb. HC1 (18) to produce octapeptide 19. The preparations of the Y,£-cyclo(glutamyl-tyrosyl-lysyl)-containing analogues I and II were completed by simultaneous catalytic hydrogenolysis of all protection groups from 15 and 19, further purification by reversed-phase HPLC and conversation of the hygroscopic trifluoracetate salts to the hydrochlorides. Crystallization from iso-propanol gave a desired compound cyclo (3, 5)-Sar-Arg-Glu-Tyr-Lys-His-Pro-Ile (I) and cyclo (3, 5)-Sar-Arg-GluTyr-Lys-His-Pro-Phe (II), whereas I was additionally purified by double re-precipitation from rnethanol-diethyl ether. Biological activity: The constrained amide linked cyclic Angiotensin II analogues were found to be active in both ex vivo and in vivo experiments. In the rat uterus assay, the agonist activity of the cyclic analogues were determined by matching its response with the equivalent response to human Angiotensin II and it was found to be -15% of that observed with the intact octapeptide. In anesthetized rabbits, previous testing had revealed that Angiotensin II infused at a rate of 2 mg/min exerts a significant and submaximal hypertensive response (+105 ± 13 mmHg). Thus, in each animal a comparison was made between blood pressure changes produced by 2 (ug/min of Angiotensin II with those produced by 10, 20, 40 and 80 ug/min of the cyclic analogues. As shown in Figure 2, c-[Sar1-Lys3-Glu5] ANG II and c-[Sar1Glu3-Lys5] ANG II are potent agonists producing significant and dose-dependent blood pressure increases. The blood pressure started rising within approximately 1 min after the commencement of drug infusion, reached a plateau within 1-2 min and returned to preinjection levels within 2-4 min after discontinuation of the drug. Of note, the magnitude of the blood pressure elevation was much higher with Angiotensin II than with equimolar quantities of the two potent cyclic analogues. In contrast, the other two cyclic Angiotensin II analogues with Ile at position 8 namely [Sar1-Lys3-Glu5-Ile8] ANG II and [Sar1-Glu3Lys5-Ile80] ANG II failed as agonists but caused a dose-dependent antagonism of the hypertensive response induced by Angiotensin II infusion.
Figure 2. Changes of Blood Pressure in mmHg for the four cyclic peptides under study.
10
L.
Polevaya et al. / Cyclic Angiotensin II Analogues
Conclusion This research was aiming at establishing differences between Angiotensin II agonist and antagonist peptides and at confirming the aromatic ring clustering conformational model for Angiotensin II which has been recently proposed on the basis of structureactivity relationships, NMR and fluorescence life time studies. A strong Tyr4-Ile5-His6 bend exists in cyclic agonist and antagonist cyclic analogues which both bear the same central Glu-Tyr-Lys or Lys-Tyr-Glu ring moiety, indicating that the difference in activity is resulting mainly from the nature of the C-terminal residue. An aromatic residue such as Phe allowing the formation of a ring cluster is necessary for exertion of agonist activity in ANG II. The constrained cyclic amide linked Angiotensin II analogues c-[Sar1-Lys3-Glu5] ANG II and c-[Sar1-Glu3-Lys5] ANG II were designed to keep intact the clustering and backbone bend characteristics of the peptide hormone Angiotensin II. On the contrary, the cyclic antagonists [Sar1-Lys3-Glu5-Ile8] ANG II and [Sar1-Glu3-Lys5-Ile8] ANG II without an aromatic residue at position 8, lack the ring cluster conformation. The molecules were designed with the hypothesis that residues 3 and 5 are not important for activity and exist on the other side of the molecule from the functionally important aromatic side chains and this structure can be accommodated in the charge relay conformation proposed for Angiotensin II. The lower potency of both agonist and antagonist cyclic analogues compared to ANG II and Sarilesin points out that the achieved conformation by cyclic analogues is not the optimal in inducing or suppressing ANG II induced hypertension. In the potent constrained agonist analogues the three rings are closely spaced at the same side of the cyclic ring and this has been shown by NOE interactions and molecular modeling. Activity is similar regardless of Lys-Tyr-Glu or Glu-Tyr-Lys order in cyclic agonist or antagonist peptides. The obtained data confirm our hypothesis that the aromatic side chains together with the C-terminal carboxylate are the essential pharmacophoric groups for receptor activation. These data also emphasize the role of closely spaced residues 4, 6 and 8 to form a possible relay system, which is not possible in Sarilesin and cyclic ANG II antagonists, lacking a C-terminal aromatic residue. In addition, the data show that the way of cyclisation at 3 and 5 positions does not interfere with the activity of the resulting peptide. The following figure shows models of the four peptides under study. These were derived using a combination of NMR spectroscopy and Molecular Modeling.
L. Polevaya et al. / Cyclic Angiotensin II Analogues
Figure 3. Models of the four analogs analogs. The models show proximity of the three aminoacids (Tyr, His and Phe or Ile) and that cyclization through Lys3-Glu5 or Glu5-Lys3 does not affect the activity of the peptides.
Acknowledgments This work was supported by the Ministry of Development, General Secretariat of Research and Technology of Greece (EPET II/115, PENED 1999), a NATO Linkage Grant (974548), the Latvian Science Council grant (96.0726) and the Latvian National Scientific Programme (ZP-10). References [1] H. Gavras, H.R. Brunner, J.H. Laraph, J.E. Sealey, I. Gavras, R.A. Vukovich, N. Engl. J. Med. 291 (1974) 817. [2] H. Gavras, H.R. Brunner, G.A. Turini, G.R. Kershaw, C.P. Tifft, S. Cuttelod, I. Gavras, R.A. Vukovich, D.N. McKinstry, N. Engl. J. Med. 298 (1978) 991. (3] Blow et. al., Nature 221 (1969) 337. [4] J. Matsoukas, J. Hondrelis, M. Keramida, T. Mavromoustakos, A. Makriyannis, R. Yamdagni, Q. Wu and G.J. Moore, 7. Biol. Chem. 269 (1994) 5303. [5] J. Matsoukas, J. Ancans, T. Mavromoustakos, P. Roumelioti, D.V. Vlahakos, R. Yamdagni, Q. Wu, G.J. Moore, Bioorg. Med. Chem. 8 (2000) 1. J. Matsoukas, G. Bigam, N. Zhou and G. Moore, Peptides 11 (1990) 359. J. Matsoukas, R. Yamdagni, G. Moore, Peptides 11 (1990) 367. J. Matsoukas, G. Agelis, A. Wahhab, J. Hondrelis, D. Panagiotopoulos, R. Yamdagni, Q. Wu, T. Mavromoustakos, H. Maia, R. Ganter, G.J. Moore, J. Med. Chem. 38 (1995) 4660. [9] T. Mavromoustakos, A. Kolocouris, M. Zervou, P. Roumelioti, J. Matsoukas, R. Weisemann, J. Med. Chem. 42 (1999) 1714. [10] P. Roumelioti, T. Tselios et. al., Bioorg. And Med. Chem. Letters 10 (2000) 1. [ 1 1 ] H . Gavras and I. Gavras, Bioactive Peptides in Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press. 1999. [ 12]K. Alexopoulos, P. Fatseas, E. Melissari, D. Vlahakos, J. Smith, T. Mavromoustakos, M. Saiteddine, G. Moore, M. Hollenberg, J. Matsoukas, Bioorganic and Medicinal Chemistry 1 (1999) 1033.
12
L.
Polevaya et al. / Cyclic Angiotensin II Analogues
[13] A Wahhab, J.R. Smith, R.C. Ganter, D.M. Moore, J. Hondrelis, J. Matsoukas and G.J. Moore, Drug Research 43 (1993) 1157. [14]J. Matsoukas, D. Panagiotopoulos, M. Keramida, T. Mavromoustakos, R. Yamdagni, Q. Wu, G.J. Moore, M. Saifeddine, M.D. Hollenberg, J. Med. Chem. 39 (1996) 3585. [15]T. Tselios, L Probert, I. Daliani, E. Matsoukas, A. Troganis, I. Gerothanassis, T. Mavromoustakos, G. Moore and J. Matsoukas, 7. Med. Chem. 42 (1999) 170. [16]J. Matsoukas, J. Hondrelis, G. Agelis, K. Barlos, R. Ganter, D. Moore and G.J. Moore, J. Med. Chem. 37 (1994) 2958. [17]K. Alexopoulos, D. Panagiotopoulos, T. Mavromoustakos, P. Fatseas. MC. Paredes-Carbajal, D. Mascher, S. Mihailescu, J. Matsoukas, J Med Chem. 44 (2001) 328. [18]K,L. Spear, M.S. Brown, E.J. Reinhard, E.G. McMahon, G.M. Olins, M.A. Palomo, D.R. Patton, J. Med. Chem. 33 (1990) 1935. [19]E. Sugg, C. Dolan, A. Patchett, R. Chang, K. Faust, V. Lotti, In Peptides: Chemistry, Structure and Biology, ESCOM Science: Leiden, The Netherlands, 1990, pp 305-306. [20] J. Matsoukas, M. Scanlon, G.J. Moore, J. Med. Chem. 27 (1984) 404. [21]G.V. Nikiforovich, G.R. Marshall, Biochem. Biophys. Res. Commun. 195 (1993) 222. [22]G.V. Nikiforovich, J.L.-F. Kao, K. Plucinska., W.J Zhang, G.R. Marshall, Biochemistry 33 (1994) 3591. [23] K. Plucinska, T. Kataoka, M. Yodo, W.L. Cody, J.X. He, C. Humblet, G.H. Lu, E. Lunney, T.C. Major, R.L. Panek, P. Schelkun, R. Skeen, G.R. Marshall, 7. Med. Chem. 36 (1990) 1902. [24] W.J. Zhang, G.V. Nikiforovich, J. Perodin, D.E. Richard, E. Escher, G.R. Marshall, J. Med. Chem. 39 (1996) 2738. [25] E. Jorgensen, W. Patton, J. Med. Chem. 12 (1969) 935. [26] J. De Coen, E. Ralston, J. Durieux, In Peptides: Chemistry, Structure and Biology, Ann Arbpr. Ml, 1975, pp.553-558. [27] J.Ancans, D. Biseniece, N. Myshliakova, E. Porunkevich, Bioorg. Khim. 12 (1986) 118. [28] J. Ancans, D. Biseniece, N. Myshliakova, G. Chipens, Bioorg. Khim. 16 (1990) 358. [29] J. Matsoukas, J. Hondrelis, G. Agelis, K. Barlos, D. Gatos, R. Ganter, D. Moore, G.J. Moore, 7. Med. Chem. 37 (1994) 2958. [30] E. Gross, R. Meienhofer, J., G. Eds. Chipens, G. Nikiforovich, F. Mutulis, N. Veretennikova, I. Vosekalna, A. Sosnov, L. Polevaya, J. Ancans, N. Mishlyakova, E. Liepinsh, I. Sekacis, M Breslav, Cyclic analogs of linear peptides. In Peptides. Structure and Biological Function. Proceeding of the Sixth American Peptide Symposium, Pierce Chemical Company: Rockford, IL, 1979 pp. 567-570. [31]L. Polevaya, T. Mavromoustakos et. al., Bioorg. Med. Chem. 9 (2001) 1639.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
13
Synthesis of RGD AnalogsIncorporating Moiety of Salicylic Acid Derivatives and their Effect on Human Platelet Aggregation in vitro Y. Sarigiannis1, G. Stavropoulos1, M. Liakopoulou-Kyriakides2 1
2
Dept. of Chemistry, University of Patras, Patras, Greece, Dept. of Chemical Engineering, Aristotle University of Thessaloniki, Thessaloniki, Greece
Abstract. A series of RGD (Arg-Gly-Asp) analogs incorporating salicylic acid derivatives at their N-terminal amino group have been synthesized by conventional solution techniques and/or by solid phase synthesis. The synthesized compounds were tested for their inhibitory activity on human platelet aggregation in vitro. They exert their inhibitory activity by adding the aggregation reagent to citrated platelet rich plasma from venous blood. The analog 2-HO-C6H4-CO-Arg-Gly-Asp-NH2 (1) showed higher inhibitory activity than the parent peptide Arg-Gly-Asp-NH2, which means that the combination of salicylic acid and RGD-NH2 in the same molecule increases the antiplatelet activity. The analog 2-HO-C6H4-CO-Arg-Gly-Asp(OBzl)NH2 (3) was found to be the most potent inhibitor of platelet aggregation with IC50 0.05 mM. This ensures our previous observations about the requirements of a lipophile group at the C-terminal side of the peptidic sequence.
14
Y. Sarigiannis et al. / Synthesis of RGD Analogs
Introduction The development of new antithrombotic agents has been stimulated by clinical needs as long as the major cause of death and disablement, in developed countries, are cardiovascular diseases, mainly unstable angina, stroke and acute myocardial infraction. There are two classes of antithrombotic agents -anticoagulants and antiplatelet agentsthat are used in antithrombotic therapy. Anticoagulants inhibit thrombin generation and fibrin formation and antiplatelet agents block platelet aggregation. Aspirin, coumarins, low-molecular weight heparins and direct inhibitors of thrombin have been used as antithrombotic agents for many years with effectiveness but they have their limitations. Antagonists of platelet glycoprotein IIb/IIIa (GP IIb/IIIa) represent a new therapeutic approach in inhibiting platelet aggregation, thus providing a powerful form of antithrombotic therapy [1-4]. The minimum sequence on fibrinogen recognized by GP IIb/IIIa receptor is Arg-Gly-Asp (RGD) [5-7]. Extensive research on structure-activity as well as conformational studies have been performed on the pharmacofore groups [810] and have led to the development of peptidic [11,12] and not peptidic analogs [13,14]. Other researchers isolated RGD-containing peptides from snake venoms with effective activity on platelet aggregation [15-17]. In order to get more stable molecules, researchers synthesized cyclic analogs [18-21]. We have already reported that the combination in the same molecule of dipeptide amides, containing amino acid(s) of RGD sequence with salicylic or acetylsalicylic acid moiety 2-RO-C6H4-CO-, [where R: H or Ac] at their N-terminal amino group have shown inhibitory activity on human platelet aggregation stimulated by collagen, ADP or adrenaline [22-24]. Here, with this report we carry out our investigational effort. All the synthesized compounds contain amino acid(s) of RGD sequence incorporating moiety of salicylic acid or derivatives. Experimental Part Materials and Analytical Methods: Solid Phase Peptide Synthesis (SPPS) was carried out using the 2-chlorotrityl chloride resin [25] (CBL, Patras, Greece) as solid support on which the Rink linker was coupled. All amino acids and derivatives used in this work are of S-configuration and are purchased from CBL (Patras, Greece), while the aggregating agents collagen and ADP were purchased from Sigma Chem. Co (Germany). The used solvents were of analytical grade. Capillary melting points were determined on a Electrothermal apparatus and are reported uncorrected. IR spectra were recorded as KBr pellets on a Perkin-Elmer 16PC FT-IR spectrophotometer. 1H and 13C NMR spectra were obtained at 400 MHz on a Bruker (Avance) instrument using CDCl3 or DMSO-d6 as solvent and TMS as internal standard. Chemical shifts are reported as 6 values in parts per million. ESI-MS were recorded on a Micromass (Platform LC) mass spectrometer. Analysis by TLC was performed on precoated plates of silica gel 60 F254 (Merck) with the following solvent systems (ratio by volume) AcCN/H2O (5:1) (1), nBuOH/AcOH/H20 (4:1:1) (2), n-BuOH/AcOH/Pyr/H2O (4:1:1:2) (3). Spots on TLC plates were detected by UV light, with ninhydrin solution and/or by chlorination followed by solution of 1% starch/ 1% KI (1:1 v/v). For the cleaning of the synthesized crude compounds was used Flash Chromatography with silica gel 60, 230-400 mesh
Y. Sarigiannis et al. / Synthesis of RGD Analogs
(ASTM), whereas the collection was done with the Ultrorac 7000 fraction collector (LKB). Biological Assays: Platelet aggregation was studied by a conventional photometric technique with a Dual Channel Coulter Electronic Aggregometer, at 37°C, with continuous recording of light transmission, according to the method of Born [26]. The aggregation agents used were at a final concentration of 60u,g/ml for collagen and 10mM for ADP. Platelets were obtained from venous blood of healthy donors who had not taken any medication during the week preceding blood collection. The blood was immediately mixed at a 9:1 ratio with 3.8% sodium citrate solution and was centrifuged at 200g for 5min to yield platelet rich plasma. Chemistry Activation and coupling procedure: Fmoc amino acid (3mmol, threefold excess of the resin with linker substitution) and 1-hydroxybenzotriazole (HOBt, 4.5mmol) were dissolved in DMF (4 ml). Diisopropylcarbodiimide (DIC, 3.3mmol) was added and the solution was transferred to the solid phase reactor. After 3h, the mixture was washed (3x DMF, 2x 2-propanol, 2x DMF, 3x 2-propanol and 2x ether). The progress of the reaction was checked by Kaiser's test. In case of uncompleted reaction, the coupling was repeated by using half of the quantities of the Fmoc-AA-OH and the rest reagents.
H3CO
Rink Amide Linker
O-CH2COOH
2-CLTR
Cleavage of the Fmoc-group: The cleavage was performed at room temperature with 20% piperidine in DMF for 5 min followed by a second treatment for 25 min. After that, the mixture was washed with the same washing procedure, as above. Coupling of the salicylic-acid and its derivatives with the tripeptides:Salicylic-acid or its derivatives (3 mmol, three-fold excess of the resin with linker substitution) and PyBOP (3.63 mmol) were dissolved in DMF (4 ml). DIPEA (4.5 mmol) was added and the solution was transferred to the solid phase reactor. After 5h, the progress of the reaction was checked by Kaiser's test. Subsequently, the above washing procedure was applied. Cleavage of the salicyl-peptides from 2-Chlorotrityl resin: The salicylic-peptide-linker resin ester was suspended in a mixture of 25ml of DCM/TFE/AcOH (7:2:1) for 2.5h at room temperature. The resin was filtered off and the filtrate was concetrated in vacuo. The crude peptide was precipitated by addition of ether, collected by filtration, washed with ether on the filter and dried in vacuo.
15
16
Y. Sarigiannis et al. / Synthesis of RGD Analogs
Removal of the side-protected groups and linker's moiety: The salicylic-peptide-linker was treated in a solution of TFA/DCM/scavengers for 2.5h at room temperature.The solution was concetrated in vacuo. The crude salicylic-peptide was precipitated by addition of ether, collected by filtration, washed with ether on the filter and dried in vacuo
H3CO
I.Fmoc-X-OH
1
X1:Glyorl_eu
2. Fmoc-X'-OH
[oiC/HOBt
X : AspCOBut). Asp(OBzl).
3. Fmoc-Arg(Pbf)-OH Deprotection with 20% piperidine in DMF
Asp(OMe)
*• H-Arg(Pbf)-X1-X-Linker-O-Q PyBOP DIPEA, 5h
CO-Arg-X1-X-NH2 X1: GlyorLeu X : Asp, Asp(OBzl), Asp
Cleavage with DCWTFE/AcOH 7 : 2 : 1
.COArgfPbO-X'-X-Linker-O-Q
Deprotectkxi with TFA/DCM/EDT/l-fcO/Anisole 75 : 15 :3 : 4 : 3
Fig. 1: The synthetic route of RGD analogs
Figure 1 shows the synthetic route of RGD analogs. The incorporated in the peptide chain amino acids have either the sequence of RGD or the Gly has been replaced by Leu, an amino acid which has given salicylic-dipeptides with strong inhibitory activity. Also, the aspartyl residue is incorporated in some cases as p-methyl or (3-benzyl ester. The activated salicylic acid derivatives react at the N-teminal group of the peptidic sequence. Results and Discussion We have reported in the past, that the combination in the same molecule of dipeptide amides, containing amino acid(s) of RGD sequence, with salicylic-residue 2RO-C6H4-CO~, {where R= H or CH3CO} at their N-terminal amino group have shown inhibitory activity on human platelet aggregation. In an attempt to synthesize more potent inhibitors of platelet aggregation, we tried out the synthesis of RGD analogs incorporating salicylic acid derivatives and in some cases we used aspartic acid products (p-benzyl or p-methyl ester) in order to have at the C-terminal side of the analog a lipophile group, as it is suggested from many researchers and from our group too [22,23].
17
Y. Sarigiannis et al. / Synthesis of RGD Analogs
The crude synthesized analogs have been purified by crystallization and flash column chromatography. Table 1 summarizes some physical constants and other data of the synthesized analogs. Table 1: Physical constants of RGD analogs
RGD analogs
1. R1-Arg-Gly-Asp-NH2
MW
M+
m.p. (°C)
TLC Rfi
Rf2
0.23 0.11 0.34 0.49 522.5 241 (dec) 0.71 2. R1-Arg-Leu-Asp-NH2 0.66 0.14 3. R1-Arg-Gly-Asp(OBzl)-NH2 556.4 242 (dec) 0.37 0.54 0.19 612.5 244 (dec) 0.60 4. R1-Arg-Leu-Asp(OBzl)-NH2 0.67 480.4 0.52 5. R1-Arg-Gly-Asp(OMe)-NH2 246 (dec) 0.62 0.70 0.41 6. R1-Arg-Leu-Asp(OMe)-NH2 535.5 242 (dec) 0.46 0.62 544.5 7. R2-Arg-Gly-Asp-NH2 180 (dec) 0.42 0.12 0.36 8. R2-Arg-Gly-Asp(OMe)-NH2 558.5 559.3 183 (dec) 0.45 0.15 0.56 513.9 514.5 189 (dec) 9. R3-Arg-Gly-Asp(OMe)-NH2 0.46 0.19 0.58 10. R1-Arg(NO2)-Gly- 600.2 623.2 190 0.66 0.58 Asp(OBzl)-NH2 0.69 R1:2-HO-C6H4-CO, R2: 5-Br-2-HO-C6H3-CO, R3: 5-Cl-2-HO-C6H3-CO (1) AcCN/H20 (5:1, v/v), (2) BuOH/AcOH/H2O (4:1:1, v/v), (3) BuOH/AcOH/ Pyr/H 2 O(4:l:l:2,v/v) 466.4
466.2
240 (dec)
7 522.3 7 556.6 7 612.4 3 480.3 2 536.7 2 545.2
The synthesized RGD analogs were tested for inhibitory activity on human platelet aggregation in vitro, by adding the aggregation reagent (collagen) to citrated platelet rich plasma (PRP). Platelets were obtained from venous blood of healthy volunteers and the PRP was isolated by centrifugation at 200g for 5 min at 37° C. The aggregation was determined using a dual channel electronic aggregometer by recording the increase of light transmission. The compound 2-HO-C6H4-CO-Arg-Gly-Asp-NH2 (1) showed higher inhibitory activity than the compound HC1.RGD-NH2 (11). This result indicates that the combined molecule of salicylic acid with RGD may lead in a synergetic action, giving more potent compound against platelet aggregation. Comparing the side (3-ester group of aspartyl-residue it was found that the compound 2-HO-C6H4-CO-Arg-GlyAsp(OBzl)-NH2 (3) is more active with IC50<0.05 mM, indicating that a lipophile character of the side chain at this position is preferable by the receptor (Figure 2a and 2b).
18
Y. Sarigiannis et al. / Synthesis of RGD Analogs
0 2040
60803
4
5
6
Time (min)
Fig. 2a. Effect of compounds on human platelet aggregation induced by collagen: HO-C 6 H 4 COArg-Gly-Asp-NHJl), HCl.Arg-Gly-Asp-NH2 (11) at concentration of 1.9mM. C. control (normal serum + collagen)
4 Tinw
5 (min)
Fig. 2b. Effect of compounds (2), (3), (4) and (5) on human platelet aggregation induced by collagen at concentration of 0.5mM. C, control (normal serum + collagen
The next figures 3a and 3b show the inhibitory effect of RGD analogs, 7 and 10 respectively, on platelet aggregation induced by collagen at the concentration of 1.7 mM. As it is shown peptide 7 presents strong inhibitory effect with an IC50 value estimated at 0.4 mM for the case of collagen, whereas a mild effect is presented by peptide 10. The rest of the analogs didn't show any significant effect.
) 00
1 DO
4 OO
* OO
• I
Fig. 3a. Effect of compound (7) on human platelet aggregation induced by collagen at concentration of 1.7 mM. C, control (normal serum + collagen)
Fig. 3b. Effect of compound (10) on human platelet aggregation induced by collagen at concentration of 1.7 mM. C, control (normal serum + collagen)
Aknowledgment: The Research Committee of the University of Patras supports this project (Grant No 2468)
Y. Sarigiannis et al. / Synthesis of RGD Analogs
References [1] Fareed J., Hoppensteadt D., Walenga J. and Bick R., Med. Clin. of North America, 78 (1994) 7-13 [2] Fareed J., Callas D., Hoppensteadt D., Walenga J. and Bick R., Med. Clin. of North America, 82 (1998) 569 [3] Hirsh J., Weitz J., The Lancet, 353 (1999) 1431 [4] Clemetson K.J. and Clemetson J.M., Cell. Mol. Life Sci., 54 (1998) 502 [5] Pierschbacher M.D. and Ruoslahti E., Nature, 309 (1984) 30 [6] Gartner T. K. and Bennett J. S., The Journal of Biological Chemistry, 260 (1985) 11891 [7] Ruoslahti E. and Pierschbacher M.D., Science, 238 (1987) 491 [8] Kang Y.K., Jhon J.S., J. Peptide Res., 56 (2000) 360 [91 Dal Pozzo A., Fagnoni M., Bergonzi R., Vanini L., De Castiglione R., Aglio C., Colli S., J. Peptide Res., 55 (2000) 447 [10]Bognadowich-Knipp S.J., Jois D.S.S. and Siahaan T.J., J. Peptide Res., 54 (1999) 43 [11]Tzong-Ming W., Ming-Liang L. and Tz-Chong C., Thrombosis Res., 97 (2000) 191 [12]Ramesh C.V., Jayakumar R. and Puvanakrishnan R., Peptides, 19 (1998) 1695 [13]Gopalsamy A., Yang H., Ellingboe J.W., Kees L.K., Yoon J., and Murrills R., Bioorg. Med. Chem. Lett., 10 (2000) 1715 [14] Katada J., Takiguchi Y., Muramatsu M., Fujiyoshi T. and Uno I., Thrombosis Res., 88 (1997) 27 [15]Sheu J.R., Yen M., Peng H.C., Chang M.C., Huang T.F., European Journal of Pharmacology, 294 (1995) 231 [16]Yeh C.H., Peng H.C., Yih J.B., Huang T.F., Biochimica et Biophysica Acta, 1425 (1998) 493 [17]Sheu J.R., Ko W.C., Hung H.C., Peng H.C. and Huang T.F., Journal of Pharm. Pharmacol., 49 (1997) 78 [18] Schumann C., Seyfarth F., Greiner G., Reissmann S., J. Peptide Res., 55 (2000) 428 [19] Allen D., Helium O., and Jacobsen E., Angew. Chem. Int. Ed., 37 (1998) 1907 [20]Kessler H., Kantlehner M., Gibson C., Haubner R., Finsinger D., Dechantsreiter M., Plancker E., Wertmuth J., Schmitt J., Meyer J., Schaffner P., Holzemann G., Wiesner M., Goodman S., Hahn D., Jonczyk A., Wester H. and Schwaiger M., Peptides, (2000), p.235 [21] Dechantsreiter M.A., Plancker E., Matha B., Lohof E., Holzemann G., Jonczyk A., Goodman S.L. and Kessler H., J. Med. Chem., 42 (1999) 3033 [22]Stavropoulos G., Magafa V., Liakopoulou-Kyriakides M., Sinakos Z. and Aaberg A., Amino Acids, 13 (1997) 171 [23]Stavropoulos G., Magafa V., Sarigiannis Y. and Liakopoulou-Kyriakides M., Peptides, (1998), p.860 [24] Sarigiannis Y., Stavropoulos G., Magafa V., Liakopoulou-Kyriakides M. and Garypidou L., Peptides, (2000), p.727 [25]Barlos K., Chatzi O., Gatos D. and Stavropoulos G., Int. J. Peptide Protein Res., (1991), 37, 513 [26] Born GVR, Nature, 194 (1962) 927
19
20
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Synthesis of Substance P C-terminal analogs incorporating D-amino acids and studies of their antineoplastic properties in vitro P. Vakalopoulou1, G. Stavropoulos1*, M. Liakopoulou-Kyriakides2, Z. lakovidou3 and E. Mioglou3 1
Dept. of Chemistry, University of Patras, 26500 Patras, Greece Dept. of Chemical Engineering, Aristotle University of Thessaloniki, and 3 Dept. of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece 2
Abstract. Several small peptides are currently under investigation as possible antitumor agents. Neuropeptides such as substance P (SP) and neuropeptide Y (NPY), have been studied for their ability to prevent tumor growth or the proliferation of several cancer cell lines. These neuropeptides have been investigated for their effect to prostate cancer, small cell lung cancer (SCLC) and breast cancer. The synthetic SP analog (D-Arg1, D-Phe5, D-Trp7.9, Leu11]SP (antagonist D) and the C-terminal analog [Arg6, D-Trp7.9, MePhe8]SP6-11 (antagonist G) inhibit SCLC cell proliferation in vitro and in vivo, while the analogs [Glp6, Glu(But)11]SP6-11 and [Glp5, Glu(But)11]SP5-11 showed significant inhibition in the proliferation of the cancer cell lines HeLa and T47D. In the present study the C-terminal analogs of SP [Glp6, D-Trp7, Glu(But)11]SP6-11 ( 1 ) , [Glp6, D-Trp7.9, Glu(But)11]SP6-11 (2), [Glp6, D-Trp7.9, MePhe8, Glu(But)11]SP6-11 (3), [Glp6, D-Trp7, MePhe8, Glu(But)11]SP6-11 (4), [Glp6, Trp7, MePhe8, Glu(But)11]SP6-11 (5), [Glp6, MePhe7, D-Trp8, Glu(But)11]SP6-11 (6), [Glp6, D-Trp7, MePhe8, Glu(But)11-OH]SP6-11 (7), [Glp6, D-Trp7, Cys(Acm)11-OH]SP6-11 (8), [Glp6, D-Trp7, MePhe8, Cys(Acm)"-OH]SP6-11 (9), [Glp6, D-Trp7.9, MePhe8, Cys(Acm)11-OH]SP6-11 (10) have been synthesized and tested for their antineoplastic properties in several cancer cell lines and for their cytotoxicity to normal cells. The analogs were performed using the stepwise synthesis either in solution, using the method of mixed anhydrides with carbonic acids or in SPPS, using the Fmoc/Bu1 methodology. The fragment condensation method in solution, using phosphonium reagents, such as PyBOP, was also applied.
P. Vakalopoulou et al. / Synthesis of Substance P C-terminal Analogs
Introduction Substance P (SP) is naturally occurring and well-studied undecapeptide amide (H-Arg-Pro-Lys-Pro-Gln-Gln-Phe-Phe-Gly-Leu-Met-NH2), widely distributed in the central and peripheral nervous system and belonging to the family of mammalian tachykinins together with neurokinins A and B (NKA and NKB). They possess a very similar amino acid sequence at their C-terminal pentapeptide amide: H-Phe-X-Gly-LeuMet-NH2, where X=Phe in case of SP and Val in case of NKA and NKB. SP, NKA and NKB activate the G-protein-coupled receptors Neurokinin-1, Neurokinin-2 and Neurokinin-3, respectively. Tachykinin receptor selectivity can be altered by modifying residues in the divergent N-terminal region and position "X". N-terminal fragments do not retain tachykinin activity. The minimum sequence identified to date that maintains significant biological activity is the C-terminal pentapeptide SP7-11. However, this peptide binds poorly to NK-1 receptor, while the hexapeptide [pGlu]SP6-11 is functional and displays high affinity [1]. It is assumed that SP participates in a wide variety of physiological functions, like smooth-muscle contraction, vasodilatation, inflammation, secretion from glands, transmission of pain and regulation of immune responses [2,3]. Monocytes and macrophages regulate many aspects of immune function including antigen processing, T cell proliferation, antibody production, tumor defense and cytokine production [4,5]. Substance P stimulates human peripheral blood monocytes to produce these inflammatory cytokines including interleukin-1 (IL-1), IL-6, IL-10, BL-12 and tumor necrosis factor alpha (TNF-a) [6-9], which are important constituents of immune cell activation that act as physiological inductive signals in the regulation of immune responses. TNF-a is a major mediator of inflammation which upregulates in T cells and monocytes in vitro [10]. Substance P enhances TNF-a [7] and IL-10 [8] production from monocytes and macrophases derived from adult human peripheral and placental cord blood and modulates HIV-1 replication in human peripheral blood monocyte-derived macrophases (MDM) [11], supporting the concept that neuropeptides such as substance P are important regulators of cytokine expression in human bloodderived monocytes and MDM. Monocytes and macrophases treated with the Substance P C-terminal octapeptide analogs [Pro4, Glu(OBzl)11]SP4-11, [Hyp4, Glu(OBzl)11]SP4-11, [cHyp4, Glu(OBzl) 11 ]SP 4-11 and [kPro4, Glu(OBzl)11]SP4-11 alone increased TNF-a secretion at a magnitude similar to the effect of entire undecapeptide SP. Treatment of monocytes and macrophages with the SP analogs in combination with LPS, however, showed no synergistic interaction in upregulation of TNF-a. These data indicate the biological effect of SP on TNF-a production by human monocytes and macrophages depends mainly on the sequence of the C-terminal region of the molecule [12]. The synthetic SP analog [D-Arg1, D-Phe5, D-Trp7.9, Leu11]SP (antagonist D) and the C-terminal analog [Arg6, D-Trp7.9, MePhe8]SP6-11 (antagonist G) are the most potent antagonists of bombesin stimulated by Ca21 mobilizing neuropeptides. These peptides inhibit the growth of small cell neuroendocrine cancer [13-15]. It was found that they inhibit SCLC cell proliferation in vitro and in vivo [14-17]. As a result, antagonist G is in a Phase II clinical trial [18]. Recently we found that the analogs [Glp6,
21
22
P. Vakalopoulou et al. / Synthesis of Substance P C-terminal Analogs
Glu(But)11]SP6-11 and [Glp5, Glu(But)11]SP5-11 showed significant inhibition in the proliferation of the cancer cell lines HeLa and T47D [19]. Chemistry The analogs 1-10 were performed using the stepwise synthesis either in solution, using the method of mixed anhydrides with carbonic acids or in SPPS using the Fmoc/But methodology on the solid support of 2-chlorotrityl chloride resin [20]. The fragment condensation method in solution, using phosphonium reagents, such as PyBOP, was also applied. SYNTHESIS IN SOLUTION
SOLID PHASE SYNTHESIS
Z-Leu-OH + HCl Glu(But)-NH2
2-Clt-resin
Step by Step
1. X=D-Trp, Y=Phe, 2. X=D-Trp, Y=Phe, 3. X=D-Trp, Y=MePhe, 4. X=D-Trp, Y=MePhe, 5. X=L-Trp, Y=MePhe, 6. X=MePhe,Y=L-Trp,
Z=Gly Z=D-Trp Z=D-Trp Z=Gly Z=Gly Z=Gly
l. Method of mixed anhydrides with carbonic acids 2. H2-Pd/C
H-Leu-Glu(But)-NH2
Glp-X-Y-Z-OH
Method of phosphonium reagents Glp-X-Y-Z-Leu-Glu(But)-NH2 (1-6) Step by Step Glp-D-Trp-MePhe-Gly-Leu-GluCBuVOH (7)
2-Clt-resin Step by Step 2-Clt-resin
Glp-D-Trp-X-Y-Leu-Cys(Acm)-OH 8. X=Phe, Y=GIy 9. X=MePhe, Y=Gly 10. X=MePhe, Y=D-Trp
(8-10)
P. Vakalopoulou et al. / Synthesis of Substance P C-terminal Analogs
23
Biological tests The biological assays of the synthesized analogs are under investigation. The applied methods concern Sister Chromatide Exchange (SCE) and the Proliferation Rate Index (PRI) [21]. The SCE method was applied on cultured human lymphocytes from healthy donors and the effect of the analogs of the chromatide exchanges and of the mitotic cycle was studied. The preparations were scored for cells in their first, second, third and subsequent divisions and second division cells that were suitably spread, were scored blindly for SCEs and lymphocyte proliferation kinetics. A minimum of 30 cells were scored for each culture in order to establish mean SCE values. The method of PRI is a sensitive marker of cytotoxicity. For PRIs, at least 100 cells were scored. The PRI values were calculated according to the formula, PRI=(M1+M2+M3)/N, where M1, M2, M3 indicate the metaphases corresponding to first, second, third or subsequent division and N is the total number of metaphases scored.
Results and Discussion In the present study the SP C-terminal analogs [Glp6, D-Trp7, (1), [Glp6, D-Trp7.9, Glu(But)11]SP6-11 (2), [Glp6, D-Trp7.9, MePhe8, (3), [Glp6, D-Trp7, MePhe8, Glu(But)11]SP6-11 (4), [Glp6, Trp7, MePhe8, 6 7 8 t 11 6 7 8 t 11 11 (5), [Glp , MePhe , D-Trp , Glu(Bu ) ]SP6-11 (6), [Glp , D-Trp , MePhe , Glu(Bu ) 6 7 11 6 7 OH]SP6-11 (7), [Glp , D-Trp , Cys(Acm) -OH]SP6-11 (8), [Glp , D-Trp , MePhe8, Cys(Acm)11-OH]SP6-11 (9) and [Glp6, D-Trp7.9, MePhe8, Cys(Acm)11-OH]SP6-11 (10) have been synthesized and tested for their antineoplastic properties in several cancer cell lines. They were also examined for their cytotoxicity to normal cells. The analogs 1-6 are peptide amides whereas the analogs 7-70 are peptide acids. They were purified (HPLC) and identified (ESI-MS) (Table 1). Table 1: Physical constants of SP C-terminal analogs. HPLC ESI-MS TLC [M+H]+ M.W. m.p. (°C) tR (min) Rf(b) R f(a) Rf(c) 0.81 818 0.67 172-174 / 817.06 0.63 15.8(I) 0.78 0.68 137-140 14.5 (II) 948 2 947.26 0.67 0.85 15.1 (II) 962 0.77 176-178 961.26 0.76 3 0.82 4 831.09 14.9 (II) 832 0.76 0.78 181-183 0.71 0.80 0.76 179-183 14.7 (II) 832 5 831.09 0.78 831.09 0.76 178-181 832 0.71 6 14.8 (II) 174-177 7 832.07 833 0.70 0.75 ,_ 0.71 12.8 (II) 0.77 807.02 0.77 808 0.78 224-226 8 ,_ 19.8(I) 0.62 9 0.68 0.66 217-220 822 821.05 10.3 (II) 10 951.15 0.50 0.48 0.64 167-169 952 20.6 (I) TLC: a: l-BuOH/AcOH/H2O (4:1:1, v/v), b: !-BuOH/AcOH/Py/H2O (4:1:1:2, v/v), c: MeCN/H2O (3:1, v/v). HPLC: I: 20%MeCN, 80%H2O =*> 100%MeCN, 0%H2O, //: 40%MeCN, 60%H2O =* 100%MeCN, 0%H2O.
The biological results showed (Table 2) that the higher SCE values were obtained with peptides /, 3 and 9 at the concentration of 10uM. The PRI value, obtained for these compounds were found close to the control values. Peptides 4-7 are still under
P. Vakalopoulou et al. / Synthesis of Substance P C-terminal Analogs
24
investigation, therefore no results are mentioned. The rest of the peptides gave values close to the control levels. Table 2: SCEs and PR1 values for peptides 1-3 and 8-10. Concentration (uM) Substances SCEs+SE Control 7.32 ±0.53 Peptide 1 2 8.18 ±0.68 » 10 9.34 ± 0.74 Peptide 2 2 8.08 ± 0.56 » 10 7.86 ± 0.56 Peptide 3 2 9.18 ±0.68 » 10 9.31 ± 0.68 Peptide 8 2 8.99 ± 0.63 » 10 8.12 ±0.41 Peptide 9 2 7.65 ± 0.40 » 10 9.62 ± 0.58 2 Peptide 10 8.33 ± 0.79 10 » 9.00 ± 0.65
PRI 1.98 2.05 2.01 1.97 1.96 1.90 2.00 2.25 2.14 2.06 2.01 1.89 2.03
References [1] Cascieri M.A., Huang R.R., Fong T.M., Cheung A.H., Sadowsky S., Ber E. and Strader C.D., Mol. Pharmacol, 41 (1992) 1096. [2] Giannis A. and Kolter T., Angew. Chem. Int. Ed. Engl., 32 (1993) 1244 and references cited therein. [3] Karagiannis, K., Manolopoulou, A., Poulos, C., Stavropoulos, G., Eur. J. Med. Chem., 29 (1994) 667. [4] Auger, M.J., Ross, G.A., (1992), In: Lewis, C.E., O' D McGee, J. (Ed), The Macrophage, Oxford University Press, New York, pp1-74. [5] Ho, W.-Z., Douglas, S.D., (1995), In: Beutler, E., Lichman, M.A., Coller, B.S., Kipps. T.J. (Eds), Hematology, 5th ed., Williams Hematology. McGraw-Hill, New York, ch.86. [6] Laurenzi, M.A., Persson, M.A.A., Dalsgaard, C.J., Haegerstrand, A., Scand. J. Immunol., 31 (1990) 529. [7] Lee, H.R., Ho, W.-J., Douglas, S.D., Clin. Diagn. Lab. Immun., 1 (1994) 419. [8] Ho, W.-Z., Kaufman, D., Uvaydova, M., Douglas, S.D., J. Neuroimmu., % (1996) 132. [9] Kincy-Cain, T., Bost, K.L., J. Immunol., 158 (1997) 2334. [10] Rosenberg, Z.F., Fauci, A.S., Immunol. Today, 11 (1992) 176. [11]Ho, W.-Z., Cnaan, A., Li, Y.-H., Zhao, H.-Q.. L., H.-R., Song, L.. Douglas, S.D., AIDS Hum. Retroviruses, 12 (1996) 195. [12]Ho W.Z., Stavropoulos G., Lai J.P., Hu B.F., Magafa V., Anagnostides S., Douglas S.D.. J. Neuroimmu., 82 (1998) 126. [13] Woll, P.J., Rosengurt, E., Proc. Natl. Acad. Sci. USA, 85 (1988) 1859. [14] Woll, P.J., Rosengurt, E., Cancer Res., 50 (1990) 3968. [15]Langdon, S., Sethi, T., Ritchie, A., Muir, M., Smyth, J., Rosengurt, E., Cancer Res., 52 (1992) 4554. [16] Sethi, T., Langdon, S.P., Smyth, J.F. and Rosengurt, E., Cancer Res., (1992), 52 (Suppl.), 2737. [17]Cummings, J., MacLellan, A.J., Jones, D.A., Langdon, S.P., Rozengurt, E., Ritchie. A.A. and Smyth, J.F., Annals Oncol., 6 (1995) 595. [18]MacKinnon, A.C., Armstrong, R.A., Waters, C.M.. Cummings, J., Smyth, J.F., Haslett. C. and Sethi, T., British Journal of Cancer, 80 (1999) 1026. [19]Korakakis, G., Vakalopoulou, P., Magafa, V., Liakopoulou-Kyriakides, M. and Stavropoulos, G., (2000), Peptides 661. [20]Barlos, K., Chatzi, O., Gatos, D. and Stavropoulos. G., Int. J. Pept. Prot. Res., 37 (1991) 513. [21]Mourelatos, D., J. A.,(1996) 136.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
25
Synthesis of Chiral Tetramic Acids Using Activated Derivatives of N-protected aAmino Acids as Precursors. Structural Characterization of Tetramic Acid Complexes With Metal Ions J. Markopoulos,1 A. Detsi,2 E. Gavrielatos,2 G. Athanasellis, 2 and O. IgglessiMarkopoulou2 1
Laboratory of Inorganic Chemistry, Department of Chemistry, University of Athens, Panepistimiopolis, GR-15771 Athens, Greece ' Laboratory of Organic Chemistry, Department of Chemical Engineering, National Technical University of Athens, Zografou Campus, GR-15773 Athens, Greece
Abstract. Activated derivatives of N-protected a-amino acids are used as precursors for the synthesis of tetramic acids, nitrogen containing heterocycles that exhibit a vast range of biological activity. The complexation of tetramic acid enolates with various metal ions is studied and their coordinating features are investigated.
Introduction a-Amino acids have been extensively used for the synthesis of heterocycles for a number of reasons: the proteinogenic amino acids possess a limited but significant number of functional groups, which facilitates synthetic operations, their protection and deprotection chemistry is thoroughly documented and they are readily available commercially, usually in both enantiomeric forms. Moreover, amino acids are widely used as enantiomerically pure starting materials for the synthesis of optically active compounds [1].
J. Markopoulos et al. /Synthesis of Chiral Tetramic Acids
26
Our research group has been involved in the development of new synthetic routes of biologically active nitrogen containing heterocycles and in the study of their coordination chemistry for several years [2]. In this article, we would like to focus on the synthetic methodologies we have developed for the construction of tetramic acids, using W-protected-a-amino acids as starting materials as well as the synthesis of their complexes with metal ions. The basic heterocyclic nucleus, pyrrolidine-2,4-dione (tetramic acid, Scheme 1), is an integral part of a number of physiologically active natural products. The spectrum of biological activity displayed by these compounds is remarkable in its diversity; it includes potent antibiotic, antiviral and antifungal properties, cytotoxicity and mycotoxicity as well as the inhibition of tumors [3]. Scheme 1
For example, streptolydigin and tirandamycin, natural products isolated from fungi, contain the 3-acyl tetramic acid moiety and exhibit potent in vitro antibacterial activities and in vivo efficacy in mouse protection tests [3]. Xanthobacin A and discodermide, isolated from marine sponges, are macrocyclic lac tarns containing tetramic acid units and function as defensive chemicals on these sponges [4]. Aurantosides D, E and F, new polyene tetramic acids comprising an N-trisaccharide unit, isolated from marine sponges, exhibit cytotoxicity against P-388 murine leukemia cells and antifungal activity against a couple of fungi [5]. The most recently isolated naturally occurring tetramic acids are reutericyclin, a low molecular weight antibiotic from lactic acid bacteria [6], aflastatin A, a specific inhibitor of aflatoxin production [7] and cryptocin, a potent antimycotic agent [8]. Scheme 2. Representative examples of naturally occurring tetramic acids.
OH
O
O
Streptolydigin
iNHMe
Reutericyclin
The diversity of biological activity displayed by tetramic acids prompted us to develop a new synthetic methodology based on the following retrosynthetic scheme:
J. Markopoulos et al. /Synthesis of Chiral Tetramic Acids
27
(II)
Scheme 3. Retrosynthetic analysis of the new route to 3-substituted tetramic acids.
In order to achieve this synthesis a N-protected a-amino acid, with activated carboxylic function (I), and an active methylene compound (II) were required. The activation of the carboxyl group of the amino acid was succeeded using two activating agents well-known in peptide synthesis, namely N-hydroxysuccinimide and N-hydroxybenzotriazole whereas the appropriate active methylene compounds were chosen, in order to provide the desired substituents on the 3-position of the pyrrolidine-2,4-dione system. Synthesis of N-alkoxycarbonyl-3-substituted tetramic acids using N-hydroxysuccinimide esters of N-alkoxycarbonyl-a-aminoacids as precursors: The proposed methodology involves the C-acylation reaction of an active methylene compound 2 with the W-hydroxysuccinimide esters 1 of N-Boc and N-Z-amino acids (glycine, L-alanine, D,L-alanine) and /V-Boc-phenylalanine. O HO Y
OO,R"
HiC
002R"
HO
Y=CO2Me, CO2Et, COMe, GOPh
R'=H, Me Scheme 4. Synthesis of N-alkoxycarbonyl-3-substituted tetramic acids, 3. The C-acylation indermediates undergo an in situ cyclization reaction to afford N-alkoxy-carbonyl-3-substituted tetramic acids 3, via an intramolecular condensation mechanism.
28
J. Markopoulos et al. /Synthesis of Chiral Tetramic Acids
During the course of these reactions, N-hydroxysuccinimide esters of amino acids proved to be excellent acylating agents, in addition to their well-known advantages (i.e. stability and ease of preparation). Moreover, the N-hydroxysuccinimide formed during the acylation reactions is water-soluble, therefore easily removed from the reaction mixture. Application of the suggested methodology to the construction of chiral tetramic acids with a stereogenic center at C-5, led to the isolation of the corresponding optically active compounds [9]. 'One-pot' synthesis of optically active tetramic acids from N-acetyl a-amino acids mediated by 1-hydroxybenzotriazole: The 'one-pot' synthesis of N-acetyl-3ethoxycarbonyl tetramic acids 4 comprises of a C-acylation reaction between the 1hydroxybenzotriazole ester of the appropriate optically active amino acid and diethyl malonate (Scheme 5). COOC2H5 COOC2H5
R=CH2Ph, CH2CH(CH3)2, CH3
(a) NaH/diethyl malonate=l:2, DCC, anh. THF (b) NaH/diethylmalonate=2:2, DCC, anh. THF (c) EtONa, EtOH, rt Scheme 5. Synthesis of optically active 3-ethoxycarbonyl tetramic acids 4,6.
When the product was not the corresponding tetramic acid but the C-acylation compound 5, a cyclization reaction under basic conditions was performed to afford the corresponding tetramic acids 6. The proposed methodology succeeded to maintain the stereochemical integrity of the corresponding a-amino acids. Another advantage is that there is no need for isolating the intermediates 1-hydroxybenzotriazole esters of the chiral a-amino acids thus the time for the synthesis of the desired products is reduced and the overall yield is improved. Additionally, the reaction is simple, inexpensive, easily scaled up and
J. Markopoulos et al. / Synthesis of Chiral Tetramic Acids
29
proceeds with low racemization, providing a convenient way of synthesizing optically active tetramic acids [10]. Synthesis of 5-arylidene-3-acyl and 3-alkoxycarbonyl tetramic acids: 5-arylidene-3-acyl and 3-alkoxycarbonyl tetramic acids contain important structural adjuncts such as an enolic |3,(3'-tricarbonyl moiety, a lipophilic 3-acyl or 3-alkoxycarbonyl substituent at position 3 and a hydrophobic group at the 5-position which allow them to exhibit versatile biological activity. The double bonds between C-3 and C-4 as well as C-5 and C-6 of the 5-arylidene-3-substituted tetramic acid nucleus make these compounds very promising substrates for catalytic asymmetric hydrogenation, a method which has been recently used for the synthesis of statine analogues from tetramic acids [11]. Our goal was to develop a convenient route to 5-arylidene-3-substituted tetramic acids which, in turn, could be used as starting material for the synthesis of statine analogues.
o ff
H0v
^=<
R •CHO
CH3COC1 »• EtOH
R = Me, Pr, OEt X=NO2, Cl, OCH3, H Scheme 6. Synthesis of 5-arylidene-3-acyl and 3-alkoxycarbonyl tetramic acids.
The synthesis of the desired compounds (Scheme 6) involves the condensation of tetramic acids (lequiv.) with substituted-benzaldehydes (2 equiv.) in the presence of a solution of HC1 8% in ethanol (Scheme 6). These compounds were isolated in good yields and recrystallised from hot ethanol. Previous synthetic strategies to these compounds are less convenient and give lower yields. The Z configuration of the double bond was assigned according to the literature based on a comparison of the 1H NMR chemical shift data for the vinyl proton signals at the region of 6.42-6.65 ppm with those of similar tetramates [12]. Complexes of 3-acyl tetramic acids with metal ions: The (3,p'-tricarbonyl moiety present in the 3-acyl tetramic acids provides a suitable site for bidentate complexation to a metal. This is proved by several examples of naturally occurring tetramic acids: 'magnesidin', a natural antibiotic, was isolated as a 1:1 mixture of the covalent magnesium chelates of l-acetyl-3-hexanoyl- and 3-octanoyl-tetramic acid derivatives [13], 'geodin A', a new macrocyclic polyketide lactam tetramic acid which has recently been isolated as a magnesium salt [14] whereas 'oleficin' functions as an ionophore for Mg2+ and Ca2+ ions in isolated rat liver mitochondria [15]. The mechanisms responsible for the various biological properties of tetramic acids have been shown to include the complexation with various metal cations, which increases the liposolubility and permeability through cell walls thus enhancing their activity [16]. Recently, the synthesis of new platinum(II) complexes containing 3-alkanoyltetramic acids were synthesized and reported to exhibit a broad spectrum of biological properties, including antitumor activity [17]. Although the biological activity of tetramic acid derivatives is
30
J. Markopoulos el al. /Synthesis of Chiral Tetramic Acids
claimed to be related to their complexing ability, little is presently known about the structure of their metal complexes. o -i
20
Mg
Scheme 7. Structure of the naturally occurring tetramic acid Geodin A.
Having established novel, efficient routes for the synthesis of tetramic acids bearing various substituents, we set out to explore the coordination chemistry of these compounds. We firstly investigated the complexation of 3-butanoyl and 3-benzoyltetramic acids with metal ions [e.g. Co(II), Ni(II), Cu(II)]. Refluxing the tetramic acid with the appropriate metal halide or thiocyanate provided the Co, Ni, Cu halide and thiocyanate complexes of formula MX2L (M=metal ion, X=anion, L=tetramic acid). The magnetic and spectroscopic properties of these complexes indicated that they contain six-coordinated metals with both bridging anions and tetramic acid [18]. The synthesis and X-ray crystallographic studies of rhodium(I) and copper(II) complexes of N-acetyl-3-butanoyl tetramic acids enlightened the coordination pattern of the ligand [19–20]. In both cases, X-ray crystallography revealed that the metal adopts a slightly distorted geometry [square-planar for Rh(I) and octahedral for Cu(II)] with the tetramic acid enolate ligand adopting an O,O' mode of coordination via the functionalities associated with the hydroxyl group at C-4 and the acyl group at C-3 in the pyrrolidine ring. In Scheme 8 the crystal structure of the complex [Cu(abta)2(py)2].2H2O is presented.
Scheme 8. Crystal structure of the complex [Cu(abta)2(py)2].2H2O
J. Markopoulos et al /Synthesis of Chiral Tetramic Acids
Currently, we are involved in the synthesis and characterization of platinum(II) complexes of 3-acyl-tetramic acids, in order to examine their structure and get an insight on the structure activity relationships. The complexes of N-acetyl-3-acetyltetramic acid (ata) with the formula [Pt(N-N)(ata)]+[NO3]~ (where N-N = (NH3)2, ethylenediamine, 1,2-diamino-cyclohexane) have been synthesized and assigned the structure presented in Scheme 9 [21].
(N03)',e
Scheme 9.
Conclusions Activated derivatives of N-protected-a-amino acids have been successfully used as precursors for the synthesis of tetramic acids. The base-induced condensation of N-hydroxysuccinimide or N-hydroxy-benzotriazole esters of N-protected a -amino acids smoothly gives rise to 3-substituted tetramic acids, which are isolated in pure form and satisfactory yields. The main advantages of this methodology include mild conditions, short reaction times and applicability to the synthesis of tetramic acids of high enantiomeric purity. In addition, the coordination chemistry of tetramic acids has been investigated and useful information concerning the complexation ability of tetramic acid enolates was deduced by spectral and X-ray crystallographic studies. Further investigations are presently being conducted towards the biological evaluation of the synthesized tetramic acids and their complexes with metal ions as well as the application of the developed methodologies to the synthesis of naturally occurring tetramic acids. References [1] F. J. Sardina and H. Rapoport, Chem. Rev. 96 (1996) 1825. [2] a) O. Igglessi-Markopoulou, C. Sandris, J. Heterocyclic Chem. 22 (1985)1599-1606. b) A.Detsi, V.Bardakos, J.Markopoulos and O.Igglessi-Markopoulou, J. Chem. Soc., Perkin Trans. 1, (1996) 2909 c)V.Delieza, A.Detsi, V.Bardakos and O.Igglessi-Markopoulou, J. Chem. Soc., Perkin Trans. 7, (1997) 1487. [3] B. J.L. Royles, Chem. Rev. 95 (1996) 1981. [4] Y. Hashidoko, T. Nakayama, Y. Homma, S. Tahara, Tetrah. Lett. 40 (1999) 2957. [5] N.U. Sata, S. Matsunaga, N. Fusetani, R. W. M. Van Soest, J. Nat. Prod. 62 (1999) 969. [6] Holtzel, M.G. Ganzle, G.J. Nicholson, W.P. Hammes, G. Jung, Angew. Chem.,Int. Ed. 39 (2000)2766. [7] H. Ikeda, N. Matsumori, M. Ono, A. Suzuki, A. Isogai, H. Nagasawa, S. Sakuda, J. Org. Chem. 65 (2000) 438. |8] J.Y. Li, G. Strobei, J. Harper, E. Lobkovsky, J. Clardy, Org. Lett. 2 (2000) 767. [9] A.Detsi, J.Markopoulos, O.Igglessi-Markopoulou. Chem. Comm. (1996) 1323 [10] A.Detsi, M.Micha-Screttas, O.Igglessi-Markopoulou, J. Chem. Soc. Perkin Trans. I (1998) 2443.
31
32
J- Markopoulos et al. / Synthesis of Chiral Tetramic Acids
[11] Giorgos Athanasellis, Efstathios Gavrielatos, Olga Igglessi-Markopoulou, Synlett 10 (2001) 1653. [12]U. Schmidt, B. Riedl, G. Haas, H. Griesser, A, Vetter, S. Weinbrenner, Synthesis (1993) 216. [13]Giorgos Athanasellis, Efstathios Gavrielatos, Olga Igglessi-Markopoulou, J. Heterocycl. Chem. 38(5)(2001)1203. [14] H. Kohl, S.V. Bhat, J.R. Patell, N.M. Gandhi, J. Nazareth, P.V. Divekar, N.J. de Souza, Tetrahedron Lett. (1974) 12, 983. [15]R.J. Capon, C. Skene, E. Lacey, J.H. Gill, D. Wadsworth, T. Friedel, Geodia, J. Nat. Prod. 62 (1999)1256. [16] S.V. Bhat, H. Kohl, B.N. Ganguli, N.J. de Souza, Eur. J. Med. Chem.-Chim. Ther. 12 (1977) 53. [17]W.O. Foye, J. Pharm. Sci., 50(1961)93. [18]M. Keiichi, H. Sakie, M. Masato, H. Satoru, Jpn. Kokai Tokkyo Koho JP 01,313,488 [89,313,488] (CI. C07F15/00) 1989; b) H. Sakie, M. Keiichi, M. Masato, S. Tadashi, Jpn. Kokai Tokkyo Koho JP 02,48,59 [90,48,59] (CI. C07F15/00) 1990. [19]O. Markopoulou, J. Markopoulos, D. Nicholls, J. Inorg. Biochem. 39 (1990) 307. [20]B.T. Heaton, C. Jacob, J. Markopoulos, O. Markopoulou, J. Nahring, C.K. Skylaris, A.K. Smith, J. Chem. Soc., Dalton Trans. (1996) 1701. [21]E. Gavrielatos, C. Mitsos, G. Athanasselis, B.T. Heaton, A. Steiner, J.F. Bickley, O. IgglessiMarkopoulou , J. Markopoulos, J. Chem. Soc., Dalton Trans. (2001) 639. [22] E. Gavrielatos, G. Athanasselis, O. Igglessi-Markopoulou , J. Markopoulos, Anticancer Res. 21 (2001) 1589.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
33
L-Asparaginase: Structure, Properties and Anti-Tumor Activity A.A. Pritsa and D.A. Kyriakidis Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, Thessaloniki, Greece, 54006
Abstract. This is a brief overview on the recent developments on L-asparaginase, an enzyme with anti-tumor activity. The properties and different mechanisms of regulation of L-asparaginase were compared in different microorganisms in the light of their primary structure. The cloning of L-asparaginase genes from either microorganisms or plants and their expression of active and stable Lasparaginase is reviewed. The anti-tumor activity of L-asparaginase on a variety of cancer cell lines and its chemical modification or encapsulation in liposomes for better future clinical treatment of tumors is discussed as well.
Introduction L-Asparaginase (L-asparagine-amidohydrolase, EC 3.5.1.1) is the enzyme that converts L-asparagine to L-aspartic acid and ammonia. Interest in this enzyme arose in 1961 when it was discovered that the antilymphoma activity of whole guinea pig serum [1,2] was a result of the enzyme L-asparaginase [3]. The clinical action of Lasparaginase is attributed to reduction of L-asparagine, since sensitive neoplastic cells are selectively killed by L-asparagine deprivation. L-Asparaginase from various sources like Escherichia coli [4], Erwinia carotovora [5], Serratia marcescens [6] and Vibrio succinigenes [7] has been found to be pharmacologically active. L-Asparaginase therapy is complicated by immunological reactions and various side effects. One of the side effects was ascribed to the capability of most L-asparaginases to hydrolyse Lglutamine and to limit serum levels of this amino acid [8,9]. Therefore, for the clinical studies i) a glutaminase-free L-asparaginase and ii) an L-asparaginase with less side effects, will be more advantageous.
34
A.A. Pritsa and D.A. Kyriakidis / L- Asparaginase
Purification and properties of L-asparaginase The first attempt of L-asparaginase purification was from E. coli [4]. The enzyme was purified to homogeneity [10] in two isoenzymic forms: a low-affinity cytoplasmic enzyme, L-asparaginase I (EcAl) and a secreted high-affinity enzyme, Lasparaginase II (EcA2) and only this form possesses antitumor activity [11]. LAsparaginase was purified so far from Acinetobacter calcoaceticus [12], Erwinia carotovora [13], Tetrahymena pyriformis [14], Tetrahymena thermophila, [15], Thermus aquaticus [16] and Thermus thermophilus [17]. L-Asparaginase is a multienzyme, and consists in most cases of identical subunits [18,19] or in a few cases of non identical subunits [20] (Table 1). Many L-asparaginases possess L-glutaminase activity, a characteristic crucial for using the enzyme as anticancer drug. The hydrolysis of L-glutamine varies from 2%, for EcA2, [21] to 15%, for E. carotovora, [22]. DAsparagine is not usually hydrolyzed by L-asparaginase. EcA2 is located in periplasmic area of the cell [23]. L-Asparaginase from Chlamydomonas is membrane-bound and Lasparagine is hydrolyzed even from whole cells [24]. L-Asparaginase from T. pyriformis is a membrane-bound enzyme, associated mainly with rough endoplasmic reticulum [25]. Aproximately 75% of the enzymic activity from T. thermophila is located in microsomal fraction and the remaining 25% in the pellicles. The microsomal fraction was further analyzed and all activity was found to be associated with the rough endoplasmic reticulum [15]. Table 1: L-Asparaginases from various sources and their properties Subunit MW Km Anti-tumor activity (kDa) MW (M) (kDa) Escherichia coli 130 32,5 1,25 x 10-5 L5178Y leukemia Erwinia carotovora 135 35 Acute leukemia 105 180 Serratia marcescens 31,5 10-4 Gardner's lymphoma Source
Vibrio succinogenes
4,7 x 10-5
146
Chlamydomonas
275
Acinetobacter calcoaceticus
130
l,34x 10
Tetrahymena pyriformis
166
39
Tetrahymena thermophila
(I) (II) 1000 200
(I) 50
Thermus aquaticus
80
Pseudomonas strutzeri
34
Erwinia aroideae Thermus thermophilus
155 200
3 x l0-3
25
33 38 33
-3
2,2 x l0 (ID 93
-4
2,8x10
7
Gardner's lymphoma
24
6C3HED
12
MCF7, T47D, BT20, Walker256
14,73
15 16 Dalton's lymphoma
75
HeLa, SK-N-MC, OAW42, MCF7, HT29, BT20, Raji, K562
17,74
-3
2,8 x l0-3
4 5 6
6C3HED
(I), (II) 4,1 x l0-3 8,6 x l0-3 1,45 xl0-4
Ref
18
A.A. Pritsa and D.A. Kyriakidis / L- Asparaginase
35
Regulation of L-asparaginase activity Regulation of L-asparaginase activity in vivo by different factors influencing the growth of microorganisms has been investigated. L-Asparaginase activity is dependent upon the aeration of cell culture [26], the addition of certain amino acids and organic acids [27], the substrate concentration in the growth medium [28-30] and the concentration of nitrogen [31,32]. Another report suggests that L-asparaginase activity of Leptosphaeria michotii is regulated by a phosphorylation-dephosphorylation reaction with an endogenous kinase and phosphatase [33]. Dephosphorylation of L-asparaginase from T. pyriformis by an alkaline phosphatase results in an activation of its catalytic activity. This enzyme also exhibits an intrinsic tyrosine kinase activity [34]. L-Asparaginase from T. pyriformis is a lipoprotein since is totally inhibited by phospholipase C. This inhibition can be reversed by the addition of phospholipids [35] and evidence for the formation of an enzymephospholipid complex is presented. A working hypothesis was suggested that Lasparaginase from T. pyriformis exists in four inter-convertible forms: Form A, phosphorylated complexed with lipids (active), form HA, dephosphorylated (highly active), form I, free of lipids (inactive) and form B, free of lipids and phosphate (active) [25]. The lipid dependence was also observed for L-asparaginase I and II from T. thermophila [15]. Incubation of the L-asparaginase I and II with phospholipase C resulted in gradually loss of their activities. The addition of a variety of phospholipids can reconstitute enzyme activity [15].
Sructure of L-asparaginase Crystallographic studies on L-asparaginase have been performed [36–38]. The preliminary structure of a glutaminase-asparaginase from Acinetobacter glutaminosificans has been described. The subunit of the enzyme folds into two domains: the N-terminal domain contains a five-stranded p-sheet surrounded by five a helices, while the C-terminal domain contains three a helices and less regular structure [36]. The crystal structure of EcA2 has been determined and demonstrated that the enzyme belongs to the class of o/P proteins. The enzyme consists of four identical subunits and is active only as tetramer [37]. Each subunit consists of two o/P domains connected by the linking sequence 191-212. The N-terminal domain contains an eightstranded mixed p-sheet. There is a topological switchpoint between Npl and NP3 where the C-terminal ends of those adjacent p-strands continue in opposite directions, folding into a-helices on other side of the sheet. This directional change of exit from the Cterminal ends of the P-strands creates a cleft characteristic of all parallel o/p structures that serves as a pocket utilized in ligand binding.The crystal structure of L-asparaginase from Erwinia chrysanthemi in the presence and absence of L-aspartate was determined and it was similar to that of EcA2 [38]. So far, bacterial and mammalian asparaginases are known to function as tetramers and may thus be described as dimers of dimers. On the other hand, plant asparaginases are active as dimers [39], suggesting that a dimer is sufficient for activity.
36
A.A. Pritsa and DA. Kyriakidis / L- Asparaginase
Active site The active site of L-asparaginase or EcA2 asparaginase was studied. Evidence for the presence of functional tyrosine residues was provided by chemical modification of the enzyme with reagents specific for tyrosine and NMR studies. The importance of tyrosine residues was eximined by site-directed mutagenesis, chemical modification and thermodynamic studies of protein denaturation [40]. Site-specific mutagenesis was used to replace three histidine residues, in conserved regions of EcA2, and suggested that His-183 is important for the stabilization of the native tetramer L-asparaginase. It was also indicated that His-87 is located in the interior of the protein, possibly adjacent to the active site [41]. A catalytic role for threonine-12 [42] and threonine-89 [43] of EcA2 established by site-directed mutagenesis, indicating that one of them is the initial nucleophile. The catalytic triad that forms the surface of the active site of EcA2 consists of the amino acids residues Thr-89, Asp-90 and Lys-162, that are connected by strong hydrogen bonds [37], and respectively Thr-95, Asp-96 and Lys-168 for Envinia chrysanthemi [38]. Chemical modification experiments on L-asparaginase purified from T. thermophilus implied the existence of histidyl, arginyl and a carboxylic residues located at or near active site while serine and mainly cysteine seems to be nessesary for active form [17]. Catalytic mechanism of L-asparaginase Kinetic evidence indicated that catalysis by L-asparaginase proceeds via a covalent intermediate, probably a p-aspartyl enzyme. The deamidation reaction becomes in two steps: firstly a nucleophile attacks the Cy of the substrate forming an acyl-enzyme intermediate and releasing the amino group. Secondly, another nucleophile, a water molecule, attacks the tetrahedrical intermediate releasing the product. This mechanism was confirmed by NMR studies. L-aspartate with a protonated side chain binds to the enzyme, at low pH (<5), thus both L-asparagine and L-aspartate can function as substrates at low pH [43]. The nature and identity of the primary nucleophile of the enzyme participating in the formation of the acyl-enzyme intermediate have been major subjects in the investigation of the mechanism of asparaginases. The hydroxyl group of Thr-12 and Thr-89 of EcA2 are closest to the side chain carboxylate of an aspartate bound in the active site. These residues are the most likely candidates for the primary nucleophile. Bacterial L-asparaginases were the first threonine amidohydrolases described in the literature. Site-directed mutagenesis of Thr-12 of EcA2 [42] demonstrated that Thr-12 has no effect on structure of the enzyme or substrate binding but is involved in catalysis, participating in the covalent acyl-enzyme intermediate, like the active-site serine residues of the serine hydrolases, an uncommon role of threonine residue. Site-directed mutagenesis of Thr-89 of EcA2 and crystallographic studies of the inactive mutant [43] presented aspartate covalently bound to Thr-12 supporting that Thr-12 assumes the role of the primary nucleophile. Upon mutation of Thr-89, the hydrogen-bonding network of the asparaginase triad (Thr-89, Lys-162, Asp-90) is completely disrupted, thus the mutant is completely inactive.
A.A. Pritsa and D.A. Kyriakidis / L- Asparaginase
37
Genes of L-asparaginase The primary structure of L-asparaginase from E. coli [44], Erwinia chrysanthemi [45], Acinetobacter glutaminasificans [46] is already known. EcAl is encoded by the ansA gene, which identified it as a protein of 35,388 Da [47]. The deduced amino acid sequence of EcAl shows discernible similarity to EcA2 in a region immediately adjacent to the active site of the enzyme. An open reading frame (ORF1), encoding a protein of 23,336 Da, is found 10 bp downstream from ansA; the ribosomebinding site of ORF1 overlaps the stop codon of ansA. Deletions within the 5' region of ansA abolish expression of ansA and reduce expression of ORF1 suggesting that ansA and ORF1 constitute an operon. EcA2 is encoded by the ansB gene which has been cloned and sequenced [48]. The deduced amino acid sequence differs in eleven positions from direct amino acid sequencing. A cleavable secretory signal peptide precedes the N terminus of the mature protein. Cyclic AMP receptor protein and anaerobiosis confirmed regulation of ansB [47]. Saccharomyces cerevisiae produces two L-asparaginases (ASPs): intracellular ASP I and cell-wall ASP II. Purified preparations of ASP II exhibit two protein bands upon sodium dodecyl sulfate polyacrylamide electrophoresis. Cloning and sequencing of the ASP3 gene and partial amino acid sequencing as asparaginase II, implied that both bands are encoded by ASP3 but have different termini [49]. The ASP I is encoded by the ASPl gene that shows 46% overall sequence identity to ASP3. The deduced protein product has a subunit of 41,414 Da and shows substantial sequence homology to the bacterial amidohydrolase family and 46% overall sequence identity to ASP3 [50]. The cloning, nucleotide sequence and expression of the Bacillus subtilis ans operon, which codes for L-asparaginase and L-aspartase was also reported [51]. A genomic sequence encoding Lupinus angustifolius L-asparaginase was the first report of this gene from a plant source [52]. An L-asparaginase cDNA clone was also isolated from Lupinus arboreus [53]. It was suggested that seed L-asparaginase is a single-copy gene. The plant asparaginase amino acid sequence has no significant homology with rnicrobial asparaginases but was 23% identical and 66% similar to a human glycosylasparaginase. Molecular cloning of the gene encoding L-asparaginase from Arabidopsis thaliana was also achieved [54]. Anti-tumor activity of L-asparaginase L-Asparaginase is unique among cancer chemotherapeutic agents for at least two reasons. First, the enzyme can exert its cytotoxic activity at sites far removed from the tumor itself. Secondly, L-asparaginase is a catalytic protein and one molecule of the enzyme will degrade thousands molecules of L-asparagine per minute. Its cytotoxicity is based on a biochemical difference between sensitive and resistant cells. Unlike normal cells, the malignant cells can only synthesize L-asparagine slowly and are dependent on an exogenous supply [55]. This sensitivity of the cells is related to the deprivation or the presence of small amounts of L-asparagine synthetase in contrast to non-sensitive neoplastic cells and physiological tissues [56]. In order to increase the anti-tumor activity and the plasma half-life of Lasparaginase, the enzyme has been modified with many ways. L-Asparaginase was chemically modified with polyethylene glycol [57] and a comb-shaped copolymer of polyethylene glycol and maleic anydride [58]. Clinical studies to determine the
38
A.A. Pritsa and D.A. Kyriakidis /L- Asparaginase
cytotoxicity of polyethylene glycol- L-asparaginase show that the enzyme is well tolerated in patients with advanced solid tumors and possess good anti-tumor activity [59]. L-Asparaginase has been also modified with N,O-carboxymethyl chitosan and the modified enzyme retained high catalytic activity prolonged blood half-life, over 33 times longer than that of native enzyme, making it more useful for clinical treatments of tumors [60]. Immobilization of L-asparaginase is another way to improve its anti-tumor activity. L-Asparaginase was immobilized in the inner surface of the hollow fibers in a dialyzer [61] and on the surfaces of liposomes [62]. More recently the enzyme was immobilized into a biocompatible poly(ethyleneglycol)-albumin hydrogel retaining a higher activity and plasma half life than native enzyme providing a functional bioreactor for use in vivo [63]. A new derivative of L-asparaginase, palmitoyl-Lasparaginase, has been incorporated in liposomes. Liposomal encapsulation of palmitoyl-L-asparaginase resulted in a prolongation of the blood half-life, abrogation of acute toxicity and preservation of in vivo anti-tumor activity [64]. Encapsulation in liposomes of L-asparaginase prevents the induction of anti-asparaginase antibodies, mitigates the anaphylactic reaction and the survival was prolonged [65]. Since L-asparaginase monotherapy was associated with a high response rate but short remission duration, the enzyme was employed within the framework of combined chemotherapy schedules which achieve treatment, response and long-term remissions in the majority of patients [66]. L-Asparaginase has been used in combination with other anticancer drugs like vincristine [67], epirubicin [68], interferon [69], cytosines, arabinoside [70], methotrexate, prednisone [71], adriamycin, cyclophosphamide [72] etc. L-Asparaginase of T. pyriformis is a multi-subunit enzyme exhibiting protein kinase activity as well. L-Asparaginase activity is affected by its phosphorylation state. Both native and dephosphorylated L-asparaginase show antiproliferative activity on three breast cancer cell lines (T47D, MCF7 and BT20) and on Walker 256 cells [73]. These cells do not possess measurable L-asparaginase or L-asparagine synthetase activity. Addition of D-asparagine, a competitive inhibitor of T. pyriformis Lasparaginase, counteracts the antiproliferative activity of L-asparaginase, indicating that L-asparaginase and not the kinase activity is responsible for this effect [73]. The purified thermostable L-asparaginase of T. thermophilus acts as an hexamer, is glutaminase-free and presents anti-tumor activity against human cell lines: K-562, Raji, SK-N-MC, HeLa, BT20, MCF7, HT29 and OAW42 [74]. These results indicate that effort has to be made to investigate further other target cells and different combination schemes with other antiproliferative drugs should be tried. References [1] [2] [3] [4] [5] [6] [7] (8] [9] [ 10]
Kidd JG. J Exptl Med 98 (1953) 565. Kidd JG. J Exptl Med 98 (1953) 583. Broome JD. Nature 191 (1961) 1114. Mashburn LT and Wriston JC. Arch Biochem 105 (1964) 451. Wade HE, Elsworth R, Herbert D, Keppie J and Saryeant K. Lancet 2 (1968) 776. Rowley B and Wriston JCJr. Biochem Biophys Res Commun 28 (1967) 160. Distasio JA, Niederman RA, Kafkewitz D and Goodman D. J Biol Chem 251 (1976) 6929. Schwartz RS. Nature 224 (1969) 275. Miller HK and Balis ME. Biochem Pharmacol 18 (1969) 2225. Whelan HA and Wriston JC Jr. Biochemistry 8 (1969) 2386.
A.A. Pritsa and D.A. Kyriakidis /L- Asparaginase
39
[11] Cambell HA, Mashburn LT, Boyse EA et al . Biochemistry 6 (1967) 721. [12] Joner PE, Kristiansen T and Einarson M. Biochim Biophys Acta 327 (1973) 146. [13] Lee SM, Ross JT, Gustafson ME, Wrobe MH and Muschik GM. Appl Biochem Biotechnol 12 (1986)229. [14] Triantafillou DJ, Georgatsos JG and Kyriakidis DA. Mol Cell Biochem 81 (1988) 43. [15] Tsavdaridis IK, Triantafillidou DC and Kyriakidis DA. Biochem Mol Biol International 32 (1994) 67.
[16] [17] [ 18] [ 19] [20] [21]
[54] [55] [56] [57] [58] [59]
Curran MP, Daniel RM, Guy GR and Morgan HW. Archiv Biochem Biophys 241 (1985) 571. Pritsa AA and Kyriakidis DA. Mol Cell Biochem 216 (2001) 93. Tiwari N and Dua RD. Indian J Biochem Biophys 33 (1996) 371. Raha SK, Roy SK, Dey SK and Chakrabarty SL. Biochem International 21 (1990) 987. Rozalska M. Acta Microbiol Pol 38 (1989) 233. Ho PKP, Milikin BE, Bobbit LJ, Grinnan LE, Burck JP, Frank HB, Boech DLV and Squires RW. J Biol Chem 245 (1970) 3708. Cammack KA, Marlborough DJ and Miller S. Biochem J 126 (1972) 361. Cedar H and Schwartz JH. J Biol Chem 242 (1967) 3753. Paul JH and Cooksey KE. Can J Microbiol 25 (1979) 1443. Tsirka SAE and Kyriakidis DA. Biochem Intern 19 (1989) 9. Svobodova O and Strabanova-Necinova S. Biochim Biophys Acta 321 (1973) 643. Netrval J. Folia Microbiol 22 (1977) 106. Kamerud J and Roon R. J Bacteriol 165 (1986) 293. Spring K, Jerlstrom P, Burns D and Beacham I. J Bacteriol 166 (1986) 135. Maladkar NK, Singh VK and Naik SR. Hindustan Antibiot Bull 35 (1993) 77. Roon R, Murdoch M, Kunze B and Dunlop P. Archiv Biochem Biophys 219 (1982) 101. Paul J and Cooksey K. Plant Physiol 68 (1981) 1364. Jerebzoff-Quintin S and Jerebzoff S. Biochem Biophys Res Commun 140 (1986) 1135. Tsirka SAE and Kyriakidis DA. Mol Cell Biochem 95 (1990) 77. Tsirka SAE and Kyriakidis DA. Mol Cell Biochem 83 (1988) 147. Ammon HL, Weber IT, Wlodawer A et al. J Biol Chem 263 (1988) 150. Swain AL, Jaskolski M, Housset D and Mohana Rao JK. Proc Natl Acad Sci 90 (1993) 1474. Miller M, Rao JK, Wlodawer A and Gribskov MR. FEBS Lett 328 (1993) 275. Chang KS and Farnden KJ. Archiv Biochem Biophys 208 (1981) 49. Derst C, Wehner A, Specht V and Rohm KH. Eur J Biochem 224 (1994) 533. Wehner A, Harms E, Jennings MP, Beacham IR, Derst C, Bast P and Rohm KH. Eur J Biochem 208(1992)475. Harms E, Wehner A, Aung H-P and Rohm KH. FEBS Lett 285 (1991) 55. Palm GJ, Lubkowski J, Derst C, Schieper S, Rohm KH and Wlodawer A. FEBS Lett 390 (1996) 211. Maita T and Matsuda G. Hoppe Seylers Z Physiol Chem 361 (1980) 105. Minton NP, Bullman HM, Scawen MD, Atkinson T and Gilbert HJ. Gene 46 (1986) 25. Tanaka S, Robinson EA, Appella E et al. J Biol Chem 263 (1988) 8583. Jerlstrom PG, Bezjak DA, Jennings MP and Beacham IR. Gene 78 (1989) 37. Bonthron DT. Gene 91 (1990) 101. Kim KW, Kamerud JQ, Livingston DM and Roon RJ. J Biol Chem 263 (1988) 11948. Sinclair K, Warner JP and Bonthron DT. Gene 144 (1994) 37. Sun DX and Setlow P. J Bacteriol 173 (1991) 3831. Dickson JM, Vincze E, Grant MR et al. Plant Mol Biol 20 (1992) 333. Lough TJ, Reddington BD, Grant MR, Hill DF, Reynolds PH and Farnden KJ. Plant Mol Biol 19 (1992)391. Casado A, Caballero JL, Franco AR, Cardenas J, Grant MR and Munoz-Blanco J. Plant Physiol 108(1995)1321. Capizzi RL, Bertino GR and Handschumacher RE. Ann Rev Med 21 (1970) 433. Patterson MKJr and Orr G. Biochem Biophys Res Commun 26 (1967) 228. Kawamura K, Igarashi T, Fujii T, Kamisaki Y, Wada H and Kishimoto S. Int Arch Allergy Appl Immunol 76 (1985) 324. Saito T, Ueno T, Sekine T et al. Leukemia 11 (1997) 408. Taylor CW. Dorr RT, Fanta P, Hersh EM and Salmon SE. Cancer Chemother Pharmacol 47 (2001) 83.
40
[60] [61] [62] [63] [64] [65] [66] [67]
A.A. Pritsa and D.A. Kyriakidis / L- Asparaginase
Qian G, Zhou J, Ma J, Wang D and He B. Artif Cells Blood Substit Immobil Biotechnol 24 (1996) 567. Callegaro L, Assone F, Cecconato E, Malinverni A, Pasteris V and Rossi V. Int J Artif Organs 6 (1983)91. Claassen E and van Rooigen N. Prep Biochem 13 (1983) 167. Jean-Francois J and Fortier G. Biotechnol Appl Biochem 23 (1996) 221. Jorge JC, Perez-Soler R, Morais JC and Cruz ME. Cancer Chemother Pharmacol 34 (1994) 230. Caspar MM, Perez-Soler R and Cruz ME. Cancer Chemother Pharmacol 38 (1996) 373. Muller HJ and Boos J. Crit Rev Oncol Hematol 28 (1998) 97. Ek O, Gaynon P, Zeren T, Chelstrom LM, Myers DE and Uckun FM. Leuk Lymphoma 31 (1998) 143.
[68] [69] [70] [71 ] [72] [73]
Yang CH, Yang LJ, Jaing TH and Chan HL. Int J Dermatol 39 (2000) 621. Litvinova EA, Mentkevich GL and Chimishkian KL. Vestn Ross Akad Med Nauk 6 (2000) 16. al-Gamdi H, Sabbah R, Martin J and Patay Z. Pediatr Hematol Oncol 17 (2000) 341. Aguayo A, Cortes J, Thomas D, Pierce S, Keating M and Kantarjian H. Cancer 86 (1999) 1203. Nagura E, Minami S, Nagata K et al. Nippon Ronen Igakkai Zasshi 36 (1999) 52. Kyriakidis DA, Tsirka SAE, Tsavdaridis IK, Iliadis SN and Kortsaris AH. Mol Cell Biochem 96 (1990)137. [74] Pritsa AA, Papazisis KT, Kortsaris AH, Geromichalos GD and Kyriakidis DA. Anti-Cancer Drugs 12(2001) 137. [75] Manna S, Sinha A, Sadhukan R and Chakrabarty SL. Curr Microbiol 30 (1995) 291.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) 1OS Press, 2002
Growth Inhibitory and Apoptotic Effects of Gonadotropin-Releasing Hormone (GnRH) on Human Hepatocellular Carcinoma C.V. Andreu-Vieyra and H.R.Habibi Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada, T2N 1N4
Abstract. During the process of tumorigenesis, cells acquire new capabilities including higher sensitivity to growth signals, insensitivity to growth-inhibitory signals, and unlimited replicative potential. In addition, tumor cells acquire the abilities to metastasize and escape from apoptotic controls. Thus, deregulated cell proliferation and suppression of apoptosis are the basis for neoplastic transformation. Hormones play important roles in the control of such a delicate balance in reproductive and non-reproductive tissues. However, the mechanism by which this balance is kept is poorly understood. Gonadotropin releasing hormone (GnRH) has been shown to exert anti-proliferative activities in cancer cells derived form the reproductive tract. The molecular basis for the tumor suppressor activity of GnRH is still under investigation, but it is clear that induction of apoptosis may follow signalled cell growth inhibition. In this review, we will focus on the anti-proliferative and proapoptotic effects of GnRH and present evidence that this hormone acts as a tumor suppressor in hepatocarcinoma cells.
Introduction The process of tumor development can be seen as the final outcome of specific physiological capabilities acquired by the cells that are part of the tumor. These capabilities include physiological alterations such as hypersensitivity to growth signals, insensitivity to growth-inhibitory signals, and unlimited replicative potential. In addition, tumor cells acquire the abilities to metastasize, invade, promote angiogenesis and escape from apoptotic controls [1, 2]. Thus, deregulated cell proliferation and suppression of apoptosis are the basis for neoplastic transformation [3, 4]. Hormones play important roles in the control of such a delicate balance in reproductive and non-
4]
42
C. V. Andreu-Vieyra and H.R. Habibi/Growth Inhibitory and Apoptotic Effects of GnRH
reproductive tissues. However, the mechanism by which this balance is kept is poorly understood. The objective of this review is to examine the information concerning the anti-proliferative and pro-apoptotic effects of GnRH in cancer cells, with emphasis on the results obtained in our laboratory on anti-tumor effects of GnRH on human hepatocellular carcinoma. Apoptosis: Apoptosis was the name given by Kerr et al. [5] to specific morphologic changes leading to cell death by a process different from necrosis. Apoptosis is a type of programmed cell death (PCD) that exerts a relevant role in the destruction of nonfunctional, harmful or abnormal cells during embryo development and adult cell growth [6-9]. This programmed cell death can be triggered by cell type- and physiologicaldependent stimuli. These stimuli include physiological factors such as hormones, cytokines, growth factor withdrawal, reactive oxygen species (ROS) as well as nonphysiological factors such as UV and gamma irradiation, heat shock, osmotic stress and chemical drugs [9–13]. The appropriate number of cells that constitute an organ is determined by the accurate balance between cell proliferation and cell death, which is altered in tumors resulting in a higher rate of cell proliferation than death. This is caused, in part, as a consequence of acquired mechanisms that render tumor cells refractory to the induction apoptosis. This diminished ability to activate the apoptosis program in tumor cells is in part a consequence of defects in the components of the apoptotic machinery, but also in their ability to respond to normal controls that trigger the suicide program [4]. Tumor cells undergo apoptosis in the presence of appropiate stimulus, although the phsyiological threshold of sensitivity to apoptosis is higher in cancer cells compared to that of normal cells. Cancer therapies can take advantage of this property. Genetic and epigenetic (e.g. genomic imprinting) changes in tumor suppressors genes and protooncogenes contribute to deregulation of apoptosis in tumor cells, which alters the control of apoptosis and survival pathways. Hormones play a major role in the regulation of growth, development, homeostasis, and programmed cell death. Tumors vary in their response to radiation and cytotoxic treatment, both of which induce apoptosis [14]. Endogenous and exogenous hormones by driving cell proliferation increase the number of cell divisions and the opportunity for random genetic errors. DNA replication errors during cell division create random mutations, which may give rise to a malignant phenotype. Hormonal stimulus to cell division continues along the progression pathway. For example, estrogens have been shown to stimulate cancer cell proliferation and angiogenesis, as well as to inhibit metastasis, or migration of cancer cells, which are characteristics of invasive cancers [15, 16]. The genes involved in tumor cell progression are not known, although candidates include those encoding enzymes in the endocrine hormone biosynthesis or transport pathways, DNA repair genes, tumor-suppressor genes and proto-oncogenes [17]. Apoptosis vs. necrosis: Some morphological features of apoptosis include: perinuclear chromatin condensation, changes in cell shape and blebbing of the plasma membrane without loss of integrity. The majority of the cytoplasmic organelles remain intact within structures called apoptotic bodies, which are absorbed by neighbouring cells or phagocytosed by specialized cells such as macrophages in vivo [18–21]. Necrosis is an accidental type of cell death, usually triggered by injury. During necrosis, cells and
C. V. Andreu-Vieyra and H.R. Habibi /Growth Inhibitory and Apoptotic Effects of GnRH
4.
organelles undergo swelling, blebbing and an increase in membrane permeability. Finally, blebs may burst, causing an inflammatory response, due to the release of intracellular contents into the pericellular space. Therefore, in contrast to necrosis, apoptosis is a cell death that does not causes this type of inflammatory response [22]. In fact, mechanisms allowing phagocytes to recognize apoptotic cells for clearance are uncoupled from inflammatory responses. For example, transforming growth factor (TGF) (3 1, an anti-inflammatory molecule is released from macrophages engulfing apoptotic cells [22]. The biochemical characteristics of apoptosis have been reviewed elsewhere [21, 23-25] and will be discussed in the context of hepatocellular carcinoma in the following section. Hepatocellular carcinoma and apoptosis: Hepatocellular carcinoma is among the 10 most common human cancers, with a worldwide incidence of 1 million cases per year and a mortality index of 0.94 [26]. There is a pronounced geographic variation with higher incidence in Africa and Asia [27]. In North America, the incidence of this disease increased 2.4/100,000 from the 1991 to the year 1995 (28). Hepatocellular carcinoma is associated with cirrhosis in the majority of the cases, and there is evidence for higher risk of developing cancer with infection with hepatitis B and C virus, as well as, exposure to agents that cause chronic liver injury [27, 28]. The available therapy for treatment of liver cancer is the complete surgical resection, while systemic or local chemotherapy and radiation therapy are used primarily as palliative treatments [27, 28]. Recently, a combination of cisplatin, doxorubicin, interferon alpha and 5-fluorouracil in systemic treatment showed good response in patients with hepatitis B-related hepatocellular carcinoma, suggesting a breakthrough in the treatment of this disease [28]. However, advanced liver cancer is often inoperable and/or resistant to radiotherapy and the majority of patients present extensive liver tumors and/or metastases. The overall 5-year survival expectancy is at present less than 5 percent [27]. Thus, liver cancer is considered to be one of the most lethal forms of human malignancies. Studies showing a relationship between estrogenic and androgenic hormones and liver cancer incidence suggest that this may be a hormone-dependent neoplasia [29]. During carcinogenesis in rat and human liver, DNA synthesis increases concomitant with apoptosis; however, proliferation predominates over apoptosis in hepatocellular carcinoma progression [30, 31]. Pre-neoplastic cells were found to be more susceptible to undergo apoptosis by food restriction and TGFpVl treatment than normal cells, while TGFa was important for proliferation [30]. Alterations in TGFJ3-1 signaling pathway observed in hepatocarcinoma cells include the inactivation of the TGF3-1 receptor type 1 and 2 and post-receptor mechanisms, and contribute to the decreased rate of apoptosis observed in these cells [31]. Human hepatocellular carcinomas were found to be resistant to various apoptotic stimuli, including serum starvation, and treatment with the protein kinase C inhibitor staurosporine [32]. Recently, it has been shown that primary human hepatocellular carcinoma and several liver derived cell lines, including HepG2, HuH-7 and Hep3B, express high levels of the anti-apoptotic protein Bcl-xL, which belongs to the Bcl-2 family of proteins [33,35]. Downregulation of this Bcl-xL was shown to render cancer cells susceptible to apoptosis, indicating that its overexpression in liver cancer cells may play a role in cell survival [33, 34].
44
C. V. Andrea- Vieyra and H. R. Habibi / Growth Inhibitory and Apoptotic Effects of GnRH
The activation of specialized pro-proteases, named procaspases, is at least partially responsible for the observed apoptotic phenotype in certain cell types [23]. Fujikawa et al. [64], have shown that the expression of caspases 1 and 3 was downregulated in primary hepatocellular carcinoma compared with normal liver cells, and suggested that this may contribute to the resistance to apoptosis observed in these cancer cells. Apoptosis may involve the release of mitochondrial apoptogenic factors to the cytosol with or without activation of procaspases [34]. Alternatively, apoptosis may be triggered by a mitochondrion-independent mechanism. The release of apoptogenic factors from the mitochondrion may occur upon activation of death receptors, such as Fas, or by increase in ROS. These factors include cytochrome c, the flavoprotein AIF (Apoptosis Inducing Factor), a mitochondrial DNAse named endonuclease G, Smac (Second Mitochondria-derived Activator of Caspases) and some procaspases (particularly procaspase 9) [36–38]. Thus, in hepatocarcinoma cells, apoptosis can still occur by activation of caspase 8 through the Fas/Fas ligand system or via release of mitochondrial AIF without caspase activation. Sphingolipid metabolites, such as sphingosine, have been shown to induce apoptosis in human hepatoma cells via inhibition of the AKT kinase survival pathway [39]. The Mitogen Activated Protein Kinase (MAPK) and the phosphatidylinositol-3kinase (PI-3K) signaling pathways seem to be important for the survival of HepG2 cells [40]. Thus, inhibition of these pathways, which may be exacerbated in cancer cells, is a possible target for drug design. Liver cells have been shown to be highly sensitive to Fas-induced apoptosis. Indeed, HepG2 cells expressing Fas ligand exerted cytotoxic effects in cells expressing Fas, indicating that cell death mediated apoptosis in liver tumor cells can occur by a paracrine or autocrine mechanism [41]. The levels of negative regulators of Fas signal transduction pathway such as FAP, a phosphotyrosine phosphatase that interacts with Fas, may be increased in tumor cells [42]. Interestingly, it has been shown that Nuclear Factor kappa B (NFkB), a transcriptional activator and pro-survival signal, is concomitantly activated with Fas signaling stimulation in HepG2 and HuH-7 cells [43]. This may explain discrepancies observed in Fas ligand stimulated apoptosis reported by certain investigators [41, 44]. Induction of apoptosis by Activin has been recently found to be mediated by the IB and IIB receptors and Smad 2 and 4 proteins in HepG2 cells [45]. Deregulation of cyclin A, which is required for the activation of cyclin A dependent kinase and the Gl/S transition in the cell cycle, has also been implicated in the pathogenesis of hepatocarcinoma cells [46]. Interestingly, cyclic AMP was found to induce inhibition of cyclin A expression and cyclin A dependent kinase activity, leading to growth arrest in HepG2 cells [46]. It is therefore apparent that hepatocarcinoma cells display alterations in certain apoptotic signaling pathways, and their response to a variety of apoptotic stimuli is dependent on the extension of these alterations. Identification of the defective signal transducers will help the design of anti-tumor therapies. GnRH functional diversity: GnRH is a decapeptide, first described for its stimulatory action on the release of gonadotropin hormones by the pituitary. There is now clear evidence demonstrating the presence of GnRH and GnRH receptors in various extrapituitary tissues, including ovary and testis of a number of vertebrates [47, 48]. More than fourteen variants of GnRH with largely conserved molecular structure have been characterized in vertebrates and protochordates [48, 49]. There is evidence suggesting
C. V. Andreu- Vieyra and H. R. Habibi / Growth Inhibitory and Apoptotic Effects of GnRH
45
that the majority of vertebrate species, including humans, express at least two GnRH variants [50]. In addition to mammalian GnRH (GnRH-I), a second GnRH variant (cGnRH-II/GnRH-II) has also been demonstrated in human endometrium, mouse and human neuronal cell lines and primate hypothalamus [51–53]. Several studies in mammals and non-mammalian vertebrates indicate that GnRH may play a paracrine/autocrine role in the control of follicular development, ovulation as well as induction of apoptosis leading to follicular atresia [54,103]. In addition, the growth inhibitory actions of GnRH observed in tumor cells of mammary gland, prostate and pancreatic origin appear to be independent of GnRH-induced pituitary gonadotropin release [55]. Inhibition of estrogen-induced cell proliferation in endometrial cancer cells was observed following treatment with GnRH agonists [56]. GnRH was also shown to inhibit proliferation of ovarian cancer cell lines as well as epidermal growth factorstimulated proliferation of ovarian, breast and endometrium derived cell lines [57, 58]. Treatment with GnRH has also been shown to increase the rate of apoptosis in endometrial tissue obtained from patients with endometriosis [59]. In addition, GnRH has been used as a carrier for cytotoxic agents to target GnRH-receptor bearing tumor cells [60]. In this context, new peptide conjugates of lamprey GnRH (IGnRH), with low activity at the pituitary level have been used in experimental trials in rat to treat breast tumors in vivo [61]. These peptides did not disrupt the ovarian cycle of the rats while caused the remission of the tumors [61]. GnRH signalling in cancer cells: GnRH receptors have been characterized in different tissues including pituitary, brain, ovary, breast, prostate, liver and pancreas [50]. Rat and human tumor cell lines derived from these tissues have also been shown to express GnRH receptors [29, 62–65]. There is now evidence for the presence of three receptor subtypes in vertebrates, although only two receptor subtypes have been so far found in humans [66–73]. GnRH receptor mechanism of action involves G1l and/or Gq guanosine nucleotide binding regulatory proteins, although there is evidence for the coupling of this receptor to other G-protein types, such as Gcc [74]. Multiple second messenger systems including phospholipid turnover, calcium mobilization, protein kinase C, phospholipase D, arachidonic acid metabolism, and mitogen protein kinase have been shown to be activated in response to GnRH [75–78]. Differences in the postreceptor mechanisms have been observed in pituitary cells from non-mammalian vertebrates upon treatment with native GnRH forms [79–81]. Second messengers coupled to GnRH receptors present in tumor cells lines derived from the reproductive tract appear to be different to other cell types [68, 82]. For example in ovarian carcinoma cells, there is evidence for protein tyrosine dephosphorylation upon GnRH treatment [82, 83], while in endometrial, prostate and ovarian carcinoma cells, inhibitory G-protein activation has been observed [82–84]. In addition, GnRh treatment activates the Fas/Fas ligand, a system that belongs to the tumor necrosis factor (TNF) a family, in tumor cells [85]. It should be noted that Fas/CD95 is characterized by the presence of a death domain that is able to recruit signalling proteins such as Fas associated death domain protein (FADD) and procaspase 8 [86, 87]. GnRH treatment of endometrial and ovarian carcinoma cells expressing GnRH receptors was shown to increase Fas ligand protein [88, 89, 93]. The activated caspase 8 could in turn cleave and activate other substrates including procaspase 3 or Bid, a pro-apoptotic Bcl-2 family protein, leading to apoptosis [90]. It has been suggested that downregulation of Fas and upregulation of Fas ligand could be the way this tumors escape from the surveillance of
46
C. V. Andreu-Vieyra and H.R. Habibi / Growth Inhibitory and Apoptotic Effects of GnRH
the immune system [91]. The Fas system may also be involved in sphingomyelin hydrolysis to ceramides via the activation of sphingomyelinases in ovarian cancer cells [92]. In turn, ceramides inhibit cell growth by activation of phosphatases and serine/threonine protein kinases [92]. More recently, it was shown that treatment of leiomyomas with GnRH increased the expression of anexin V, a natural inhibitor of protein kinase C [94]. Concomitant to a decrease in cell proliferation, an increase in anexin V mediated by PKC was observed [94]. Thus, it is apparent that more than one signalling pathway may mediate the antiproliferative actions of GnRH in cancer cells from different origin. Growth inhibitory and apoptotic effects of GnRH on hepatocarcinoma cell lines: The presence of GnRH binding sites in hepatocarcinoma cells (HepG2) was first described by Pati & Habibi [29]. Subsequent studies demonstrated the expression of GnRH and GnRH receptors (GnRH-R) in primary hepatocarcinoma cells [94]. The expression of GnRH and GnRH-R was found to be correlated with the degree of tumor cell differentiation characterized by higher expression of both molecules at early stages of carcinogenesis [94]. Pati and Habibi [29] demonstrated that [Dlys-6]GnRH, a potent mammalian GnRH agonist, significantly suppressed growth of HepG2 and HUH-7 in a time and dose related manner. This suppressive effect was specific, since it was inhibited by a GnRH antagonist and was not observed with other pituitary peptides. Treatment with different GnRH forms, including [Dlys-6]-GnRH (Figure 1), mammalian GnRH (mGnRH), salmon GnRH (sGnRH), chicken-GnRH-II (cGnRH-H), catfish GnRH (cfGnRH) and lamprey GnRH (IGnRH) were found to significantly decrease HepG2 proliferation [29]. Interestingly, [Dlys-6]-GnRH, IGnRH and cfGnRH were equally potent in suppressing tumor cell growth. More recently, Palyi et al [96] demonstrated that IGnRH and other GnRH synthetic analogs, conjugated with a copolymer stimulated apoptosis in breast, prostate and endometrium derived cancer cells, exert a potent antitumor property. Hepatocarcinoma cells were found to have higher binding affinities for non-mammalian GnRH peptides compared to non-cancerous human and rat liver cells [29]. The observed higher antitumor potency of IGnRH compared to mammalian GnRH suggests the presence of different GnRH receptor types in the HepG2 cells [29]. No differences have been reported between GnRH receptors expressed in normal and reproductive tract-derived tumor cell lines [92]. However, it has recently been shown that a second form of GnRH receptor, different from the originally characterized pituitary GnRH receptor is expressed ubiquitously in peripheral human tissues [97]. This new receptor has 55% identity with the type II receptor of amphibians and fish and contains a carboxy-terminal tail, which is absent in the mammalian type I receptor [97]. Differential expression of this novel type II GnRH receptor in tumor cells may explain the different binding affinities for GnRH observed in normal and tumor liver cells. Alternatively, GnRH receptors in cancer cells may be coupled to a different signaling pathway. Because of their low luteinizing hormone release potency in humans, IGnRH analogs may be used as specific tumor suppressor agents. Agonists derived from this GnRH form would have the advantage of reduced complications that could arise from high dose treatments with GnRH [29].
C.V. Andreu-Vieyra and H.R. Habibi / Growth Inhibitory and Apoptotic Effects of GnRH
700000 -
47
T
Dlys-6-GnRH (M)
Figure 1. Quantitation of the number of HepG2 cells by trypan blue exclusion after GnRH treatment. Cells were cultured during 24h or 48h in the presence or absence of the mammalian GnRH agonist Dlys6-GnRH. Cells were harvested, stained with trypan blue and counted in a hemocytometer chamber. Values represent the average of four separate experiments ±standard error. *: p<0.05' **: p<0.01. Note the decrease in the number of viable cells with increasing concentrations of the hormone.
Further studies demonstrated that GnRH inhibited zinc-induced expression of metallothionein, which is a zinc binding protein [98]. Metallothionein is also important for cell proliferation since it regulates the availability of zinc for the activity of RNA and DNA polymerases. Over-expression of metallothionein was found in cancer cells of different origins, including breast [98] suggesting that this protein may be responsible for the resistance to cisplatin observed in these cancer cells. However, it has been shown that metallothionein is downregulated in primary hepatocellular carcinoma samples from Japan, China and America [99, 100]. The mechanism of downregulation for this protein is not yet understood [101]. A relationship between induction of apoptosis and levels of metallothionein has been suggested by Lazo et al [102], since this protein acts as an antioxidant, preventing DNA oxidative damage. Mice nil for the metallothionein gene were shown to be more sensitive to radiation-induced apoptosis in the thymus than wild-type animals [101]. In addition, different regulators of apoptosis, such as cadmium, zinc, glucocorticoids, tumor necrosis factor alpha and lipopolysaccharides, increased metallothionein levels. Indeed, a negative correlation between metallothionein levels and incidence of cells undergoing apoptosis has been shown in primary hepatocellular carcinoma [99, 101]. Thus, downregulation of metallothionein could render the cancer cells more susceptible to apoptosis, a property that could be exploited in the design of cancer treatments.
48
C V. Andreu- Vieyra and H.R. Habibi / Growth Inhibitory and Apoptotic Effects of GnRH
Recently, we have found that [Dlys-6] GnRH, IGnRH, sGnRH and cGnRH-II were able to induce low molecular weight DNA fragmentation in HepG2 cells. [Dlys6] GnRH significantly increased DNA fragmentation after 24 and 48h (Figure 2) of treatment in a dose related manner. Induction of apoptosis was found to be correlated with a significant decrease in cell proliferation (Figure 1), in agreement with previous reports [29]. Morphological assessment of apoptosis was carried out in GnRH-treated HepG2 cells (stained with a mixture 1:1 ethidium bhromide: orange acridine, a technique that allows for discrimination between necrotic and apoptotic cells). Apoptotic and healthy cell nuclei stain green, with apoptotic nuclei being smaller and displaying condensed chromatin compared to healthy nuclei. No necrotic cells, which stain orange, were observed after treatment with GnRH (Figure 3). The number of cells per colony and the number of tumor cell colonies were reduced following GnRH treatment (Figure 3). Apoptotic nuclei were found in the GnRH-treated cells but not in the controls. In addition, GnRH-treated cells appeared larger in size compared with controls (Figure 3).
|Dlys-6|GiiRH(M)
Figure 2. Effect of the mammalian GnRH agonist Dlys6GnRH. 5xl05HepG2 cells/well were cultured in 1 ml MEM medium supplemented with 10%FBS, l00mML-glutamine and sodium bicarbonate at 37°C in a 95%air:5%CO2 atmosphere. GnRH was added 24h after plating. Cells were harvested at after 24h and 48h and analyzed for apoptosis using an ELISA for oligonucleotide detection. Values represent the average of 4 experiments±standard error.*=p<0.05;**=p<0.01.Note that the different GnRH forms were able to increase DNA fragmentation (a hallmark for apoptosis).
C.V. Andreu-Vieyra and H.R. Habibi/Growth Inhibitory and Apoptotic Effects of GnRH
49
Figure 3. Chromatin condensation in HepG2 cells treated with a mammalian GnRH agonist. Cells were cultured during 24h in the presence (B) or absence (A) of 10-8 M Dlys-6GnRH agonist. The cells were stained while alive with ethidium Bromide and orange acridine 1:1 dye mixture. Viable cells and apoptotic cells stain green; necrotic cells stain orange. Apoptotic cells show condensed chromatin aggregates while viable cells chromatin is disperse.Note that the agonist prevented the formation of colonies and caused chromatin condensation and aggregation (arrowheads). 400X.
The overall results indicate that the anti-tumor effect of GnRH is in part due to the induction of apoptosis in HepG2 cells. Our finding suggest that activation of Fas/Fas ligand system may mediate GnRH-induction of apoptosis in HepG2 cells as suggested for ovary
50
C. V. Andreu- Vieyra and H.R. Habibi I Growth Inhibitory and Apoptotic Effects of GnRH
and uterus derived tumor cell lines [92]. An interesting possibility is that GnRH binding to its receptor may lead to activation of INK (Jun-N-terminal Kinase) pathway in a PKC dependent or independent manner, which may in turn activate the apoptotic machinery [77].
Summary There is clear evidence that GnRH exerts anti-tumor effects on hepatocarcinoma cells, in part, through induction of apoptosis. This opens the possibility of using GnRH as a tool for treatment of liver cancer, since GnRH induction of apoptosis is tissue specific. In particular, lamprey GnRH, which has little gonadotropin-release activity in mammals, displays a potent anti-tumor property. Thus, the use of lamprey GnRH analogs could be exploited to treat liver cancer without side effects on the cycling control of the reproductive tract. The understanding of the mechanisms underlying the growth inhibitory and apoptotic effects of GnRH will lead to the development of improved potent synthetic peptides to treat hepatocellular carcinoma.
References: [1] Hanahan, and R. Weinberg, Cell 100 (2000) 57. [2] L. Liotta, and E. Kohn, Nature 411 (2001) 375. [3] P. Meier, A. Finch, and G. Evan, Nature 407 (2000) 796. [4] G. Evan, and K. Vousden, Nature 411 (2001) 342. [5] J. Kerr, A. Wyllie, and A. Currie, Br J Cancer 26 (1972) 239. [6] Wyllie, J. Pathol. 153 (1987) 313. [7] H. Billig, I. Funita, C. Rivier, J. Tapanainen, M. Parvinen et al., Endocrinology 136 (1995) 5. [8] G. Gosden, and N. Spears, Brit. Med. Bull. 53 (1997) 644. [9] M. Stellar, Science 267 (1995) 1445. [10]J. Kerr and B. Harmon. Definition and incidence of apoptosis: an historical perspective. In: Apoptosis: The molecular basis of cell death. In L. Tomei, and F. Cope, New York Cold Spring Harbor Laboratory Press, 1991,Vol 2. [11]G. Majno and I. Joris, Am. J. Pathol. 146 (1995) 3. [12] Wyllie, J. Kerr, and A. Currie, Int. Rev. Cytol. 68 (1980) 251. [13]G. Chen, X. Sun, R. Snowden, D. Dinsdaie and D. Skilleter, Biochem. J. 286 (1992) 331. [14] W. Kiess, and B. Gallaher, Eur J Endocrinol 138 (1998) 482. [15] Losordo, and J. Isner, Arterioscler Thromb Vase Biol 21 (2001) 6. [16] H. Rochefort, D. Chalbos, S. Cunat, A. Lucas, N. Platet, and M. Garcia, J Steroid Biochem Mol Biol 76(2001)119. [17] H. Henderson and H. Feigelson, Carcinogenesis 21 (2000) 427. [18]J. Kerr, and B. Harmon. Definition and incidence of apoptosis: an historical perspective. In: L. Tomei, and F. Cope (eds.). Apoptosis: The molecular basis of cell death. Cold Spring Harbor Laboratory Press, New York, 1991, Vol. 2. [19]G. Majno, and I. Joris, Am. J. Pathol. 146 (1995) 3. [20] Wyllie, J. Kerr, and A. Currie, Int. Rev. Cytol. 68 (1980) 251. [21]G. Hacker, Cell Tissue Res 301 (2000) 5. [22] J. Savill, and V. Fadok, Nature 407 (2000) 784. [23] W. Earnshaw, L. Martins, S. Kaufmann, Annu Rev Biochem 68 (1999) 383. [24] M. Hengartner, Nature 407 (2000) 770. [25] H. Stennicke and G. Salvesen, Biochim Biopliys Acta 1477 (2000) 299. [26] V Rustgi. Gastroenterol Clin North Am 16 (1987) 545. [27] T. Kevin. S. Watkins and A. Curley. Liver and Bile Ducts. In: Abeloff: Clinical Oncology, Churchill Livingstone Inc. 2000, Chapter64 pp. 1681. [28] E. Bergsland and A. Venook, Citrr Opin Oncol 12 (2000) 357. [29]D. Pati and H. Habibi, Endocrinology 136 (1995) 75. [30] R. Schulte-Hermann, K. Hufnagl, A. Low-Baselli, W. Rossmanith, A. Wagner, B Ruttkay-Nedecky, W. Bursch. L. Mullauer, W. Parzefall. and B. Grasl-Kraupp, Digestion 59 Suppl 2 (1998) 64.
C. V. Andreu- Vieyra and H.R. Habibi / Growth Inhibitory and Apoptotic Effects of CnRH
51
[31 ] S. Thorgeirsson, T. Teramoto and V. Factor, Semin Liver Dis 18 (1998) 115. [32]T. Takehara, X. Liu, J. Fujimoto, S. Friedman, H. Takahashi, Hepatology 34 (2001) 55. [33] D. Luo, S. Cheng, H. Xie, Y. Xie, Biochem Cell Biol 78 (2000) 119. [34] E. Bossy-Wetzel, D. Green, Mutat Res 434 (1999) 243. [35] M. Konopleva, S. Zhao, Z. Xie, H. Segal], A. Younes, DF. Claxton, Z. Estrov, SM. Kornblau M. Andreeff, Adv Exp Med Biol 457 (1999) 217. [36] Yuan J, Yankner B A, Nature 407 (2000) 802. [37] E. Daugas, D. Nochy, L. Ravagnan, M. Loeffler, S. Susin, N. Zamzami, and G. Kroemer, FEBS Lett 476 (2000) 118. [38] L. Li, X. Luo, and X. Wang, Nature 412 (2001) 95. [39] H. Chang, L. Tsai, L. Chuang, and W. Hung, J Cell Physiol 188 (2001) 188. [40]H. Mitsui, N. Takuwa, T. Maruyama, H. Maekawa, M. Hirayama, T. Sawatari, N. Hashimoto, Y. Takuwa, and S. Kimura, Int J Cancer 92 (2001) 55. [41] Y. Zhu, K. Luo and J. Wang, Zhonghua Gan Zang Bing Za Zhi 7 (1999) 69. [42JT. Sato, S. Irie, S. Kitada, and J. Reed, Science 268 (1995) 411. [43] H. Marusawa, M. Hijikata, K. Watashi, T. Chiba, and K. Shimotohno, Microbiol Immunol 45 (2001) 483. [44] K. Kubo, Y. Matsuzaki, M. Okazaki, A. Kato, N. Kobayashi, and K. Okita, Liver 18 (1998) 117. [45] Chen, D. Yahalom, O. Laskar-Levy, S,. Rahimipour, N. Ben-Aroya, and Y. Koch, Endocrinology 142 (2001) 830. [46] J. Lee, Y. Choi, P. Nguyen, J. Kim, S. Lee, and J. Trepel, Biochim Biophys Acta 1449 (1999) 261. [47] H. Habibi. Gonadotropin-releasing hormone as a paracrine regulator of ovarian function. In: P. Rao, and R. Peter (eds.) Neural regulation in the vertebrate endocrine system, Kluver Academy, Plenum publishers, New York, 1999, pp. 101–110. [48] H. Habibi, C. Andreu-Vieyra and E. Mirhadi. Functional significance of gonadal GonadotropinReleasing Hormone. In: H. Goos, R. Rastogi, H. Vaudry and R. Pierantoni (eds) Perspective in Comparative Endocrinology: Unity and diversity. ISBN: 88 323 1526 2, Monduzzi Editore, Italy, 2001, pp. 959-968. [49]Bauer-Dantoin, and J. Jameson, Endocrinology 136 (1995) 4432. [50]R. Millar, B. Troskie, Y. Sun, T. Ott, I. Wakefield, D. Myburch, A. Pawson, J. Davidson, C. Flanagan, A. Katz, J. Hapgood, S. Sealfon, R. Peter, E. Terasawa, and J. King. Plasticity in the structural and functional evolution of GnRH: a peptide for all seasons. In: S. Kawashima, and S. Kikuyama, (eds.). Proceedings of XIII Congress of Comparative Endocrinology, Yokohama, Japan, Monduzzi Editore, Bologna, Italy. 1997, pp. 15-28. [51] K. Cheon H. Lee, I. Parhar and I. Kang, Mol Hum Reprod 7 (2001) 447. [52] V. Latimer, S. Rodrigues, V. Garyfallou, S. Kohama, R. White, R. Fernald, and H. Urbanski, Brain Res Mol Brain Res 75 (2000) 287. [53] V. Latimer, S. Kohama, V. Garyfallou and H. Urbanski, J Clin Endocrinol Metab 86 (2001) 324. [54] J. Tilly, Rev. Reprod. 1 (1996) 162. [55] M. Marelli, R. Moretti, D. Dondi, M. Motta, and P. Limonta, Endocrinology 140(1999) 329. [56] G. Sica, G. Schinzari, C. Angelucci, G. Lama, and F. Iacopino,Mol Cell Endocrinol 176 (2001) 121. [57] Grundker, P. Volker, K. Schulz, and G. Emons, Gynecol Oncol 78 (2000) 194. [58] G. Emons, S. Weiss, O. Ortmann, C. Grundker and K. Schulz, Eur J Endocrinol 142 (2000) 665. [59] A Imai, A Takagi, and T Tamaya, Am J Obstet Gynecol 182 (2000) 1142. [60] S Rahimipour, I Bilkis, V Peron, G Gescheidt, F Barbosa, Y Mazur, Y Koch, L Weiner, and M Fridkin, Photochem Photobiol 74 (2001) 226. [61] I Teplan, Acta Biol Hung 51 (2000) 1. [62] A Tieva, P Stattin, P Wikstrom, A Bergh, J. Damber, Prostate 47 (2001) 276. [63] A Tieva, Wilkstrom P, Olofsson JI, Bergh A, Damber JE, Prostate 39 (1999) 101. [64] A Paradiso, A Pezzetta, G Cellamare, F Schittulli, F Marzullo, and S Reshkin, J Endocrinol Invest 23 (2000) 90. [65] K Fujikawa, K Shiraki, K Sugimoto, T Ito, T Yamanaka, K Takase, and T Nakano, Anticancer Res 20 (2000) 1927. [66]L Wang, L Xie, and R Zhang, Endocrine 14 (2001) 325. [67] R Millar, and J. King. Plasticity and conservation in gonadotropin-releasing hormone structure and function. In: K. Davey, R. Peter and S Tobe (eds), Perspectives in comparartive endocrinology, National Research Council of Canada, Otawa, 1994, pp. 129-136 [68|K Cheng, and P. Leung, Can J Physiol Pharmacol 78 (2000) 1029. [69]R Millar, D Conklin. C Lofton-Day, E Hutchinson, B Troskie. N Illing, S Sealfon, and J Hapgood, J Endocrinol 162 (1999) 117.
52
C. V. Andreu-Vieyra and H.R. Habibi/Growth Inhibitory and Apoptotic Effects of GnRH
[70] R Millar, S Lowe, D Conklin, A Pawson, S Maudsley, B Troskie, T Ott, M Millar, G Lincoln, R Sellar, B Faurholm, G Scobie, R Kuestner, E Terasawa, and A Katz, Proc Natl Acad Sci U S A 98 (2001) 9636. [71] S Sealfon, H Weinstein and R Millar, Endocr Rev 18 (1997) 180. [72] N Illing, B Troskie, C Nahorniak, J Hapgood, R Peter, R Millar, Proc Natl Acad Sci U S A 96( 1999) 2526. [73] B Troskie, N Illing, E Rumbak, Y Sun, J Hapgood, S Sealfon, D Conklin, and R Millar, Gen Comp Endocrinol 112 (1998) 296. [74] Z Naor, D Harris, and S. Shacham, Front Neuroendocrinol 19 (1998) 1. [75] P. Leung, and G. Steele, Endocr Rev 13 (1992) 476. [76] G. Steele and P. Leung. Signal transduction mechanisms in ovarian cells. In: E. Adashi and P. Leung (eds), The ovary, Raven Press, New York, 1993, pp.113-117 [77] Z Naor, O Benard, and R Seger, Endocrinol Metab 11 (2000) 91. [78] S Kang, C Tai, P Nathwani, K Choi, and P Leung, Endocrinology 142 (2001) 671. [79] S Shacham, D Harris, H Ben-Shlomo, I Cohen, D Bonfil, F Przedecki, H Lewy, I Ashkenazi. R Seger, and Z. Naor, Vitam Horm 63 (2001) 63. [80] J. Chang and R. Jobin. Regulation of gonadotropin release in vertebrates: a comparison of GnRH mechanism of action. In: K. Davey, R. Peter and S. Tobe (eds) Perspectives in comparative endocrinology. National Research Council of Canada, Ottawa. 1994, pp. 41–45. [81]J. Chang, F. Van Goor, A. Lo, J. Johnson R. Jobin and J. Goldberg. Signal transduction in gonadotropin (GTH-II) secreation in goldfish pituitary cells. In: S. Kawashima and S. Kikuyama (eds.) Proceedings of the XIII Congress in Comparative Endocrinology, Yokohama, Japan. Monduzzi Editore, Bologna (Italy), 1997, pp. 29-33. [82]Imai, H. Takagi, T. Furui, S. Horibe,, T. Fuseya, and T. Tamaya, Cancer 77 (1996) 132. [83]Imai, S. Horibe, H.Takagi, T. Ohno, and T. Tamaya, Eur. J. Obstet. Gynecol. Reprod. Biol. 74 (1997)73. [84] P. Limonta, R Moretti, M Marelli, D. Dondi, M. Parenti, and M. Motta, Endocrinology 140 (1999) 5250. [85] S. Nagata, and P. Goldstein, Science 267 (1995) 1449. [86] P. Krammer, Nature 407 (2000) 789. [87] H. Hug, Biol. Chem. 378 (1997) 1405. [88] Imai, A. Takagi, S. Horibe, H. Takagi and T. Tamaya, Int. J. Oncol.13 (1998) 97. [89] S. Quirk, R. Cowan, S. Joshi, and K. Henrikson, Biol. Reprod. 52 (1995) 279. [90]Strasser, and D. Vaux, Biochim Biophys Ada 1470 (2000) R1-R11. [91] Imai, A. Takagi, S. Horibe, H. Takagi, and T. Tamaya, J. Clin. Endocrinol. Metab. 83 (1998) 427. [92] Imai and T. Tamaya, Vitam Horm 59 (2000) 1. [93] Imai, A. Takagi, and T. Tamaya, Am J Obstet Gynecol 182 (2000) 1142. [94]H. Yamamoto, H. Sato, S. Shibata, M. Murata, J. Fukuda and T. Tanaka, Mol Hum Reprod 7 (2001) 169. [95] J. Zhang, G. Huang, and W. Huang, Zhonghua Yi Xue Za Zhi 78 (1998) 343. [96]Palyi, B. Vincze, S. Lovas, I. Mezo, J. Pato, A. Kalnay, G. Turi, D. Gaal, R. Mihalik, I. Peter, I. Teplan, and R. Murphy, Proc Natl Acad Sci U S A96 ( 1999) 2361. [97] J. Neill, L. Duck, J. Sellers and L. Musgrove, Biochem Biophys Res Commun 282 (2001) 1012. [98] D, Pati, H. Habibi, and L.Gedamu, Eur J Biochem 242 (1996) 36. [99] M. Waalkes, B. Diwan, S. Rehm, J. Ward, M. Moussa, M. Cherian, and R. Goyer, J. Pharmacol Exp Ther 277 (1996) 1026 [100] Deng, S. Chakrabarti, M. Waalkes, and M. Cherian, Histopathology 32 (1998) 340. [101] L. Cai , G. Wang, Z. Xu, D. Deng, S. Chakrabarti, and M. Cherian, Anticancer Res 18 (998) 4667. [102] J. Lazo,and B. Pitt, Annu Rev Pharmacol Toxicol 35 (1995) 635. [103] C Andreu-Vieyra, and H Habibi, Can J Physiol Pharmacol 72 (2000) 1003.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
53
Recent Applications of the Amide Approach to the Synthesis of Medicinally Interesting Polyamine Analogues N. Tsiakopoulos, S. Vassis, M. Militsopoulou, C. Damianakos, P. Gatos, K. Voyiatzi, E. Pantazaka and D. Papaioannou Department of Chemistry, University of Patras, GR-265 00 Patras, Greece Abstract. Acylation of a variety of mono- or diamines with the isolable succinimidyl esters TrtNH(CH2)2CO2Su and TrtNH(CH2)2CONH(CH2)3CO2Su, followed by LiAlH4 reduction, provided a variety of linear, cyclic, conformationally restricted, crowned and cross-conjugated polyamines.
Introduction Linear polyamines (PAs), like spermidine (SPD, 1) and spermine (SPM, 2) and conjugates (PACs) are widely distributed in the living organisms and are associated with interesting biological functions. A variety of linear, branched and cyclic PA analogues and PACs have been synthesized in order to determine structure-activity relationships and identify lead compounds for the development of PA-based Pharmaceuticals [1]. The currently used syntheses, both in solution and on solid supports, involve either the selective functionalization of the amino groups of commercially available linear PAs or the assembly of the PA skeleton at the desired length and number of differently modified amino groups by coupling suitable amino components (fragment syntheses). The amide approach involves the acylation of suitable amino components by activated carboxylic acids followed by reduction of the thus obtained amides, usually with LiAlH 4 (LAH) or B2H6. Several examples of this synthetic protocol have been already reported and reviewed [l d,e]. We have recently shown that the PA skeleton of N-alkylated PAs and PACs of the alkaloid kukoamine A (KukA, 3) type can be simply assembled using the 'active' esters 4–6 (Figure 1) with N-hydroxysuccinimide (HOSu) or 1-hydroxybenzotriazole (HOBt) of the Ntriphenyl-methyl (trityl, Tit)- or polymeric o-Cl-Trt (PCTr)-protected amino acids P-alanine (pAla) and y-aminobutyric acid (yAba) to acylate suitable amino components [2]. In the present work, we provide further examples of our methodology which uses isolable 'active'
54
N. Tsiakopoulos et al. / Recent Applications of the Amide Approach
esters of amino acids, such as 4, and of peptides, such as 7, for the formation of the amide bond and describe applications of the amide approach reported in the literature since the publication of our latest review article [Id].
(X WA.. ,.n H
; G=H, X=OH, n=1 yAba;G=H,X=OH,n=2 4 ; G=Trt, X=OSu, n=1 5; G=Trt, X=OBt, n=2 6; G=PCTr, X=OBt, n=1
u
Trt.^k x^-x^OSu N ^^^^ n H O 7
3
Figure 1. Structures of compounds encountered in this work.
Linear polyamines: Indeed, condensation of 'active' ester 4, used as an N-C3 synthon, with putrescine (PUT, 8), used as an N-C4-N synthon, followed by LAH reduction, provided the key-intermediate N1,Nl2-Trt2-SPM (9, Scheme 1) in 61% overall yield. This compound was successfully used in the synthesis of a variety of N-alkylated or acylated SPM derivatives [2]. The same protocol can be applied to any other linear a,o)-diamine [H2N-(CH2)n-NH2], to provide SPM analogues of the general type [3.n.3]-TEA (TEA=tetraamine). For example, bisacylation of 1,8-diaminooctane with 4, also followed by LAH reduction, gave the corresponding N1,Nl6-Trt2-[3.8.3]-TEA 10 in 48% overall yield [3,4]. Recently, we extended this methodology to accommodate the corresponding SPD derivatives. Thus, selective N-monotritylation of PUT, followed by acylation of the other nitrogen function with 4 gave the amide 11 in 73% yield. LAH reduction of amide 11 provided the corresponding N1,N8-Trt2-SPD (12) in 75% yield [5]. It should be noted that the intermediates 9 and 12 have been also prepared by other research groups through direct, selective, tritylation of the primary amino functions of the corresponding, commercially available, polyamines SPD and SPM [6]. Furthermore, acylation of the in situ generated ester H2N(CH2)3CO2SiMe3 with 4, followed by sequential methanolysis and activation with HOSu and N,N'dicyclohexylcarbodiimide (DCC) gave the also isolable 'active' ester 7 in 69% yield. Acylation of a variety of primary or secondary amines with 7, followed by LAH reduction, provided a series of selectively N-alkylated SPD derivatives 13 in 60-80% yields [7]. In particular, derivatives 13a and 13c are key-intermediates in the synthesis of a variety of other N-alkylated or acylated and cyclic SPD analogues [7,8].
N. Tsiakopoulos et al. /Recent Applications of the Amide Approach
55
13a; R1=R2=H R1
N' R2
13b;R 1 =H,R 2 =R 130; R1=R2=Bn
Scheme 1. Synthesis of linear polyamines and derivatives. Reagents: (i) 4/Et3N, DCM or DMF; (ii) LiAlH4, THF ; (iii) Trt-Cl ; (iv) Me3SiCl, DCM ; (v) MeOH ; (vi) HOSu/DCC, THF/DMF (3:1) ; (vii) R'R2NH/Et3N, DMF ; (viii) 7/Et3N, DMF ; (ix) Boc2O/Et3N/DMAP(cat.), CHC13.
'Active' ester 7 was also used, as an N-C3-N-C4 synthon, to bisacylate PUT. LAH reduction of the thus obtained tetra-amide, followed by per-tert-butoxycarbonylation of the crude product and purification by flash column chromatography (FCC), gave the fully protected [3.4.4.4.3J-HXA (HXA=hexaamine) derivative 14a in 25% overall yield. From this, the [3.4.4.4.3]-HXA 14b was readily obtained in 75% yield upon complete deprotection with 50% trifluoroacetic acid (TFA) in dichloromethane (DCM) [3]. The amide approach has been recently employed also by other research groups for the assembly of linear or macrocyclic polyamines in solution or on solid phase [9]. Diborane was invariably used in these syntheses for the reduction of the amide bonds and various protocols were reported to decompose the borane-amine intermediates thus formed [9]. Cyclic polyamines: For the preparation of cyclic PAs we envisaged succinic anhydride (SA) as a C4 synthon. Indeed, reaction of 9 with an equimolar amount of SA provided the anticipated monoacylated derivative 15 (Scheme 2) along with the corresponding diacylated product and unreacted starting material [8]. Monoamide 15 precipitated from the reaction medium and thus obtained pure in 55% yield. The cyclization of 15, to afford the bislactam 16 in 57% yield, was effectively mediated by the condensing agent bromotripyrrolidinophosphonium hexafluorophosphate (PyBrOP) in the presence of i Pr2NEt. LAH reduction of bislactam 16 gave the fully protected cyclic SPM analog 17 ([4,9]-cSPM) in 78% yield. Routine detritylation with 20% TFA in DCM, followed by acylation with the isolable 'active' ester 20 and finally catalytic hydrogenation/hydrogenolysis, provided the novel cyclic KukA analog 21 ([4,9]-cKukA) in
56
N. Tsiakopoulos et al. /Recent Applications of the Amide Approach
42% overall yield [8]. Ester 20 was readily available in 72% yield upon activation of acid 19 [2a] with DCC and HOSu. This cyclization protocol was readily extented to include the synthesis of cyclic SPD analogues, as exemplified herein with the synthesis of the globally restricted SPD analogue 27 ([l,8]-cSPD) from N4-Trt-SPD (22). The latter was readily obtained from SPD in 40% yield through a three-steps sequence, involving selective trifluoroacetylation of both primary amino functions [10], tritylation of the remaining N4-function and finally alkaline hydrolysis to remove the trifluoroacetyl protecting groups. Acylation of 22 with an equimolar amount of SA gave a mixture of the two possible monoacylation products (23 and 24), along with the diacylated product and unreacted SPD. From this reaction mixture, the monoacylated products were readily separated by FCC in 38% yield and, when subjected to identical ring-closure conditions as for 16, gave a 56% yield of the bislactam 25. LAH reduction of 25 provided the N-tritylated heterocycle 26 in 44% yield, from which [l,8]-cSPD was obtained upon routine detritylation [8]. Syntheses of cyclic SPD and SPM analogues, using however different methodology, have been already described by other research groups [11].
HO
GD
VIII, IX,
Trt H2N^^N^
R1 Trt HNU/~^(V,
~NH,
NH r»2
22 1
23 ; R =CO(CH2)2CXD2H, R2=H
Trt.
24 ; R1=H, R2=CO(CH2)2CO2H
25
26 ; R=Trt 27 ; R=H
IV
Scheme 2. Synthesis of cyclic SPD, SPM and KukA analogues. Reagents: (i) SA, DCM; (ii) PyBrOP/'Pr2NEt, CHCI3 ; (iii) LiAIR,, THF ; (iv) TFA/DCM (1:4) ; (v) 20/EtjN, DMF ; (vi) H2 (1 atm)/IO^ Pd-C, AcOH/MeOH/H2O (5:4:0.2) ; (vii) HOSu/DCC, THF/DMF (3:1) ; (viii) CF,CO2Et/H2O, MeCN ; (ix) TrtCI/*Pr2NEt, DMF ; (x) K2COV H2O/MeOH.
N. Tsiakopoulos et al. / Recent Applications of the Amide Approach
57
Conformationally restricted polyamines: 'Active' ester 4 can be also applied to the preparation of PA molecules bearing conformation restrictions, as exemplified herein with the incorporation of aromatic or heterocyclic rings into the PA backbone. However, ester 4 reacts rather sluggishly with aromatic diamines such as the 4,4'-diaminodiphenylmethane and the 2,8-diaminofluorene to provide the corresponding bisamides 29 and 33 (Scheme 3) in low yields (25-45%), along with the monoacylated products (25-30% yields). Much cleaner reactions however take place by using instead the corresponding acid TrtNH(CH2)2CO2H [2b] and PyBrOP as the coupling agent in the presence of 'Pr2NEt and a catalytic quantity of 4-dimethylaminopyridine (DMAP). The amides 29 and 33 are then obtained in 68% yield. LAH reduction of these amides provided unexceptionally the corresponding TEA derivatives 30 and 34 in 57-77% yields, from which the unprotected polyamines [3.9.3]-TEAs 31 and 35 are easily obtained upon, routine detritylation [3]. Aromatic rings, as well as double and triple bonds and saturated or unsaturated carbocyclic rings of various sizes have been incorporated by other research groups into PA molecules as conformational constrains [12]. Polyamine analogues incorporating the tetrahydropyranyl ring were synthesized from the trans-acid 36 and the cis-methyl ester 43, whose preparation was recently described by our group.[13] Thus, activation of acid 36 with DCC and HOSu, followed by in situ ammonolysis with 30% aqueous (aq) NH3 and finally LAH reduction, provided the conformationally restricted PUT analogue 37 in 53% overall yield.[14] Coupling of 37 with ester 4 gave the amide 38 in 85% yield, which upon LAH reduction provided the protected triamine ([3.4]-TRA) 39 in 55% yield. Finally, detritylation of 39 gave the SPD analogue 40. On the other hand, detritylation of 37, followed by coupling also with ester 4 and LAH reduction provided the protected TEA 41 in 43% overall yield. Finally, detritylation of 41 gave the SPM analogue 42. Detritylation of ester 43 and coupling with ester 4 gave the amide 44 in 75% yield. Ammonolysis of this compound followed by LAH reduction gave the cis-TRA 45 in 18% overall yield [15]. Routine detritylation of 45 provided the cis-SPD analogue 46 [14]. Other amino acids, such as L-proline [9e,f] and trans-4-hydroxy-L-proline [16] have been recently incorporated into cyclic and linear or branched, respectively, PA skeleta.
58
N. Tsiakopoulos et al. /Recent Applications of the Amide Approach
H,N
31 ; R=H
H
41 ; R=Trt 42 ; R=H
''"
OMe
H 39 ; R=Trt 40 ; R=H
VII, II
Scheme 3. Synthesis of conformationally restricted polyamine analogues. Reagents: (i) Trt-pAlaOIWyBrOP/'PrzNEt/CDMAPCcat.), CHCI3 ; (ii) LiAlH,, THF ; (iii) TFA/DCM (1:3) ; (iv) DCOHOSu, DMF ; (v) conc, aq NH3 ; (vi) 4/iPr2NEt, DMF ; (vii) g. NH3, MeOH.
Crowned polyamines: The 'active' esters 4 and 7 can be also employed for the synthesis of crowned polyamines (CRPAs), such as 50, 54 and 59, from the commercially available azacrown ethers 47 and 51 and the aminomethyl crown ether 55. Indeed, bisacylation of 47 with ester 4, provided the bisamide 48 in 65% yield. LAH reduction of 48 gave unexceptionally the protected tetraamine 49, which upon detritylation gave the crowned TEA 50 in 56% overall yield [17]. This compound can be considered as a conformationally restricted [3.5.3]-oxa-SPM analogue [18]. Similarly, bisacylation of aza-crown 51 with ester 7 provided the tetra-amide 52 in 92% yield. LAH reduction and detritylation provided the oxa-HXA 54 ([3.4.5.4.3]-oxa-HXA) in 58% overall yield. A combination of the above mentioned two 'active' esters was used to build-up the PA skeleton of the CRPA 59 incorporating a guanidyl function. Thus, acylation of 55 with ester 4 gave the expected monoamide in 93% yield, which upon sequential detritylation and coupling with ester 7 gave the triamide 56 in 75% yield. This was then reduced with LAH, treated with benzyloxycarbonyl chloride (ZCI) and finally purified by FCC to give the corresponding Z3-CRPA derivative 57 in 60% yield. From the thus obtained 57, the trityl group was
N. Tsiakopoulos et al. / Recent Applications of the Amide Approach
59
selectively removed with TFA/DCM and the resulting intermediate was functionalized at the primary amino function with the reagent MeS-C(=NZ)-NHZ to give the fully protected CRPA derivative 58 in 70% yield. Finally, catalytic hydrogenolysis of 58 resulted in the removal-of all five Z protecting groups giving a 70% yield of the CRPA 59, isolated as the corresponding tetrahydrochloride [17]. Polyamine cross-conjugates and fully reduced analogues: PA cross-conjugates (e.g. SPDSPM conjugates) of the type of the alkaloid tenuilobine (TEN, 65, Scheme 5) [19] and fully reduced analogues can be readily obtained using selectively N-trityl protected PA derivatives and commercially available linear a,to-dicarboxylic acids as bridging elements. Examples of the former building blocks are the already described SPD derivative 12 and the SPM derivative 60, readily obtained in 55% yield through the selective monotritylation [20] of the ditritylated SPM derivative 9. For the introduction of a 4-C bridge, succinic anhydride was employed whereas for the 16-C bridge required in the synthesis of TEN, the monophenacyl ester 61 of 1,16-hexadecanedioic acid (Hda) was used. Ester 61 was obtained in 65% yield through selective monoesterification of Hda with phenacyl bromide, followed by purification with FCC. Thus, PyBrOP-mediated condensation of 12 with ester 61 provided the amide 62 in 70% yield. The phenacyl protecting group was then removed with PhSNa and the thus obtained acid (57% yield) was coupled to the SPM derivative 60 to give the fully protected TEN 64 in 68% yield. Routine detritylation of 64 completed the formal synthesis of TEN (65) in 98% yield [20]. On the other hand, LAH reduction of 64 followed by detritylation gave the fully reduced TEN analogue 66 in 84% overall yield.
\
/
47
* --
Kl
\ i
. .'
48
Kl
'NTrt
H R'N-
r° °^N-^-~
•v^-N
"^o^J
H
~%
49 ; R=Trt 50 ; R=H ^_
[ Hi
60
N. Tsiakopoulos et al. / Recent Applications of the Amide Approach
9
9
H
H
II, V
NH
Scheme 4. Synthesis of crowned polyamines. Reagents: (i) 4/Et3N, DMF-CHCIj (1:1) ; (ii) LiAlH,, THF ; (iii) TFA/DCM (1:3) ; (iv) 7/Et,N, DMF-CHCIj (3:1); (v) PhCH2OCOCI (ZCl)/Et3N, CHCI3; (vi) Z-NHC(=N-Z)-SMe/iPr2NEt, CHCI 3 ; (vii) H2 (1 atm)/10% Pd-C, AcOH/MeOH/H2O (4:1:0.1).
When the SPD derivative 12 was initially treated with one equivalent of SA and the resulting acid was sequentially coupled, in an one-pot reaction, to the SPM derivative 60, the cross-conjugate 67 was obtained in 60% yield. Detritylation of 67 produced the TEN analogue 68 in 95% yield, bearing a 4-C bridge. Finally, sequential LAH reduction and detritylation of 68 gave the fully reduced TEN analogue 69 in 48% overall yield. Symmetric polyamine conjugates (e.g. SPD-SPD conjugates) and reduced analogues have been already described by other research groups [21, 6a,b].
N. Tsiakopoulos et al. / Recent Applications of the Amide Approach
61
CD >=o
R
HN^^Nx_x^^^N"^ R 67; R=Trt V 'C— *. 68; R=H
^NH R
NH-
69
Trt-NH
Trt-NH
viii, vi
O 'OH
62 ^ HN-Trt
HN-Trt HN-R
NH H5N
63
R-NH
NH
N-R
64 ; R=
VII, VI
66
O HN R
•«••* . . KN. " H R-NH 2 Scheme 5. Synthesis of cross-conjugated polyamines. Reagents : (i) TrtCl/Et3N, DCM ; (ii) i i PhCOCH2Br/ Pr2NEt, DMF ; (iii) 61/PyBrOP/ Pr2NEt, DMF ; (iv) PhSH/NaH(55%)/imidazole (cat.), DMF ; (v) 60/PyBrOP/iPr2NEt, DMF or DCM; (vi) TFA/DCM (1:2) ; (vii) LiAlH4, THF ; (viii) SA/iPr2NEt, DCM.
Conclusions Linear, conformationally constrained and crowned PAs of variable length and number of nitrogen functions can be readily assembled using the amide approach and isolable succinimidyl esters of N-tritylated linear amino acids and peptides to couple to commercially available or synthetic a,0)-diaminoalkanes, aryldiamines, heterocyclic diamines, aza-crown ethers and aminomethyl crown ethers. The amide approach can be also used to prepare cyclic SPD and SPM analogues, cross-conjugates and their fully reduced analogues using suitably N-tritylated PAs and the commercially available anhydrides or the readily prepared monophenacyl esters of linear a,w-dicarboxylic acids as the bridging elements. Further applications of this general approach to the synthesis of other novel PA analogues and conjugates, the examination of the metal ion complexing properties of the crowned PAs and the biological evaluation of novel PAs described herein are currently under investigation. Acknowledgements The European Commission and the Greek Ministry of Education are gratefully acknowledged for financial support in the form of MSc fellowships (N.T., S.V., M. M.,
62
N. Tsiakopoulos et al. / Recent Applications of the Amide Approach
C.D. and P.G.) and consumables. We also thank the Research Committee of the University of Patras for funding part of this work in the form of a PhD fellowship (N.T.) and consumables. References [1] (a) I. S. Blagbrough and E. Moya. Animal venoms and insect toxins as lead compounds in the design of agrochemicals-especially insecticides. In : L.G. Copping (Ed.). ISBN 0 85404 414 0. The Royal Society of Chemistry, Cambridge, 1996, pp. 329-359; (b) I. S. Blagbrough, S. Carrington and A. J. Geall. Pharm. Sci. 3 (1997) 223; (c) S. Schulz., Angew. Chem. Int. Ed. Engl. 36 (1997) 314; (d) G. Karigiannis and D. Papaioannou., Eur J. Org. Chem. (2000) 1841; (e) V. Kuksa, R. Buchan, and P. KongThoo Lin., Synthesis (2000) 1189; (f) G. Theodorides., Tetrahedron 56 (2000) 2339 and references cited within the above mentioned review articles. [2] (a) G. Karigiannis, P. Mamos, G. Balayiannis, I. Katsoulis and D. Papaioannou. Tetrahedron Lett. (1998) 5117; (b) P. Mamos, G. Karigiannis, C. Athanassopoulos, S. Bichta, D. Kalpaxis, D. Papaioannou and G. Sindona., Tetrahedron Lett. (1995) 5187; (c) C. Athanassopoulos, G. Balayiannis, G. Karigiannis and D. Papaioannou., N-Tritylamino acids in the synthesis of analogs of bioactive compounds for structureactivity relationship studies. In : J. Matsoukas and T. Mavromoustakos (Eds.). ISBN 90 5199 425 7. IOS Press, Amsterdam, 1999, pp. 137-151. [3] N. Tsiakopoulos, E. Pantazaka and D. Papaioannou, unpublished results. [4] The structures of new compounds were determined by IR, ESI-MS, NMR and microanalysis Yields of the reactions described in this work are not optimized. All polyamines obtained as final products were actually isolated as their corresponding polytrifluoroacetate salts. [5] K. Voyiatzi, E. Pantazaka and D. Papaioannou, unpublished results [6] The direct selective tritylation of primary amino functions of commercially available linear triamines (TRAs), such as nor-SPD, SPD and SPM have been described by the following research groups : (a) E. Zang and P.J. Sadler. Synthesis of hexamine ligands by using trityl as an N-blocking group. Synth, Commun. 27 (1997) 3145; (b) L. Covassin, M. Desjardins, R. Charest-Gaudreault, M. Audette, M.-J. Bonneau and R. Poulin. Synthesis of spermidine and norspermidine dimers as high affinity polyamine transport inhibitors. Bioorg. Med. Chem. Lett. 9 (1999) 1709; (c) K.E. Krakowiak and J.S. Bradshaw. Selective protection of the primary amino functions of linear tetraamines using the trityl group. Synth. Commun. 28 (1998) 3451. [7] S. Vassis, G. Karigiannis, G. Balayiannis, M. Militsopoulou, P. Mamos, G. W. Francis, and D. Papaioannou., Tetrahedron Lett. (2001) 1579. [8] M. Militsopoulou, N. Tsiakopoulos, C. Chochos, G. Magoulas and D. Papaioannou., Tetrahedron Lett. (2002), in press. [9] (a) S. Manku, C. Laplante, D. Kopac, T. Chan and D.G. Hall., J. Org. Chem, 66 (2001) 874; (b) A.N. Acharya, J.M. Ostresh and R.A. Houghten., Tetrahedron 57 (2001) 9911; (c) F. Wang, S. Manku and D.G. Hall., Org. Lett. 2 (2000) 1581; (d) M. Couturier, J.L. Tucker, B.M. Andresen, P. Dube and J.T. Negri., Org. Lett. 3 (2001) 465; (e) M. Achmatowicz and J. Jurczak., Tetrahedron : Asymmetry 12 (2001) 111; (f) M. Achmatowicz and J. Jurczak., Tetrahedron : Asymmetry 12 (2001) 487. [101 (a) M.C. O'Sullivan and D.M. Dalrympe., Tetrahedron Lett. (1995) 3451; (b) D. Xu, K. Prased, O. Repic and T.J. Blacklock., Tetrahedron Lett. (1995) 7357. [11] (a) E. Kimura and T. Yatsunami., Chem. Pharm. Bull. jpn 28 (1980) 994; (b) G. Brand, M.W. Hosseini and R. Ruppert., Tetrahedron Lett. (1994) 8609. [12] For some, very recent, examples see : (a) R.J. Bergeron, J. Wiegand, J.S. McManis, W.R. Weimar, R.E. Smith, S.E. Algee, T.L. Fannin, M.A. Slusher and P.S. Snyder. J. Med. Chem. 44 (2001) 232; (b) V.K. Reddy, A. Sarkar, A. Valasinas, L.J. Marton, H.S. Basu and B. Frydman. J. Med. Chem. 44 (2001) 404; (c) A. Valasinas, A. Sarkar, V.K. Reddy, L.J. Marton,'H.S. Basu and B. Frydman. J. Med. Chem., 44 (2001) 390; (d) Y. Zou, Z. Wu, N. Sirisoma, P.M. Woster, R.A. Casero,Jr., L.M. Weiss, D. Rattendi, S. Lane and C.J. Bacchi. Bioorg. Med. Chem. Lett. 11 (2001) 1613. [13] G. Balayiannis, G. Karigiannis, P. Gatos, D. Papaioannou and E. De Clercq. Tetrahedron Lett. (2000) 6191. [ 14] P. Gatos and D. Papaioannou. unpublished results. [15] The ammonolysis of ester 44 produced, in addition to the expected amide (30% yield after FCC purification), a comparable amount of Trt-pAla-NH? and a small quantity of Trt-j}Ala-OMe. [16] D. Nagamani and K.N. Ganesh. Org. Lett. 3 (2001) 103. [17] C. Damianakos, N. Tsiakopoulos and D. Papaioannou, unpublished results.
N. Tsiakopoulos et al. / Recent Applications of the Amide Approach
63
[18] For a comprehensive review on oxapolyamines see ref. le (pp. 1203–1206) in the present article. Polyamines bearing linear poly(ethylene glycol) residues have been recently reported. For examples see : (a) S.W. Garrett, O.R. Davies, D.A. Milroy, P.J. Wood, C.W. Pouton and M.D. Threadgill. Bioorg. Med. Chem. 8 (2000) 1779-1797; (b) E.K. Fan, Z.S. Zhang, W.E. Minke, Z. Hou, C.M.L.J. Verlinde and W.G.J. Hol. J. Amer. Chem. Soc. 122 (2000) 2663. [19] The isolation, identification and the formal syntheses of TEN and of some recently described analogues see : (a) M.K.-H. Doll, A. Guggisberg and M. Hesse., Heterocycles 42 (1996) 319; (b) M.K.-H. Doll, A. Guggisberg and M. Hesse. Helv. Chim. Acta 79 (1996) 541; (c) K. Popaj, A. Guggisberg and M. Hesse. Helv. Chim. Acta 83 (2000) 3021. [20] S. Vassis, I. Govaris, K. Voyiatzi, P. Mamos and D. Papaioannou., Tetrahedron Lett. (2002), in press [21] G. Sosnovsky and J. Lukszo. Z. Naturforsch. 41b (1986) 122.
64
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Design and Synthesis of Fluorescent Steroidal Lipopolyamine Conjugates for Monitoring Gene Delivery I. S. Blagbrough and A. P. Neal Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, U.K. Abstract The rational design and practical preparation of unsymmetrical polyamine carbamates and amides, based on steroid templates of cholesterol and the bile acid lithocholic acid as the lipid moiety, provides fluorescent molecular probes that condense DNA. These novel lipopolyamine conjugates mimic the positive charge distribution found in the triamine spermidine and the tetra-amine spermine alkaloids. These probes are useful in monitoring gene delivery for non-viral gene therapy.
Introduction and Background Gene delivery for gene therapy of a variety of human and also for animal diseases e.g. cystic fibrosis [1], hepatitis [2], haemophilia [3], cancer [4-6], and more speculatively mitochondrial gene therapy for respiratory chain defects, i.e. defects that involve the final common pathway of oxidative metabolism [7], can be performed with either a viral [8-10] or a non-viral vector [11–14]. Lipopolyamines (cationic lipids) recently developed for nonviral gene therapy show significantly lower toxicity than viral vectors, although they are currently less efficient transfection vectors than viruses [11–14]. The poor efficiency of DNA delivery to the nucleus, especially using non-viral vectors, is a major limitation of the gene therapy approach. A poor understanding of the molecular mechanisms of action in non-viral gene delivery [11, 15], and a lack of correlation between in vivo and in vitro biological activity [16-18] are unresolved issues and constitute the main current challenges. Whilst true gene therapy, i.e. the expression in cells of genetic material that has therapeutic activity, holds promise for the treatment of the significant human diseases listed above, the non-viral gene delivery vehicle, or vector, carrying the genetic material into the cell, must be optimised to increase the efficiency of gene delivery to eukaryotic cells. Uptake of condensed DNA by target cells proceeds via endocytosis, but poor nuclear localisation is another major barrier to efficient gene expression (measured using reporter genes e.g. Pgalactosidase expression). Thus, a greater knowledge of the molecular mechanisms of
I.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
65
transfection w i l l provide a basis for rational non-viral vector design [11–15, 19–24]. The possibility of also enhancing the efficiency of non-viral gene delivery to the nucleus by incorporating specific nuclear targeting (localisation) sequences in vectors is another area of interest [23, 24]. Therefore, our aims in this research programme include the design and synthesis of useful molecular probes, initially as model compounds and then for the study of DNA-lipopolyamine vector interactions at the molecular and intracellular levels. H H
H,N
Figure 1
Polyamines are polycationic at physiological pH and play key roles in biological systems. In chromatin, polyamines such as the triamine spermidine 1 and tetra-amine spermine 2 (Fig. 1) help to package DNA into nuclei through neutralisation of the poly-anionic phosphate backbone charges, bringing about DNA condensation and assisting in the control of DNA solution conformations. Lipopolyamines condense DNA more efficiently than simple polyamines [25] - this is a key first step in non-viral gene delivery. Employing a high molecular weight cationic polymer for non-viral gene delivery to transfect target cells, is often associated with unacceptably high toxicity [26]. Small molecule lipopolyamines are therefore of considerable interest due to their advantages of low toxicity, low immunogenicity, controllable syntheses, and defined molecular structure for pharmaceutical characterisation. We are studying the synthesis of steroidal polyamine conjugates, secondary alcohol functionalised cholesteryl carbamate 3 and lithocholic acid amide 4 (Fig. 1). Lipopolyamines reported in this research area (Fig. 2) include inter alia: Transfectam 5 [27] and RPR-120535 6 [28] have branched or linear spermine moieties linked to two C18-lipid chains, whereas Genzyme's Lipid GL #67 7 possesses a cholesteryl lipid moiety [17]. Vitamin D2 (ergocalciferol) polyamine conjugate 8 is a steroid with an open B-ring, designed to probe the structure-activity effects on transfection of modifying the geometry within the hydrophobic steroid motif [29]. Buckminster Fullerene tetraammonium ion conjugate 9 is typical of a new class of cationic lipid incorporating a C60 hydrophobic core, where the amine moiety was conjugated to fullerene by a cycloaddition reaction [30]. Gemini surfactant GS4 10 is a tetra-lysine based cationic lipid, carrying a maximum of 6 positive charges and conjugated with two C|2-lipid chains [31, 32]. Novel spermine-based cationic gemini surfactants symmetrically acylated with two C18-lipid and two tri-lysine chains, with up to 8 positive charges, have also recently been reported [33]. Non-viral transfection efficiency cannot rationally be improved without a greater knowledge of the detail of intracellular events. In order to address the rational design of small molecule vectors, the molecular mechanisms of these poorly understood steps must be delineated. Fluorescent microscopy is useful for the study of intracellular events. We are therefore synthesising the functionalised steroid polyamine conjugates cholesteryl carbamale 3 and lithocholic acid amide 4 (Fig. 1) as DNA condensing agents which we
66
I.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
have designed in order that a variety of fluorophores can be introduced allowing the rapid synthesis of a required library of steroidal lipopolyamine fluorescent probes [34–38]. The polyamine conjugates are designed to mimic the charge distribution found in the naturally occurring triamine spermidine 1, and are prepared by regio-controlled conjugation of a suitably protected derivative 11 of the symmetrical polyamine spermine 2 (Fig. 3) [39,40].
I.S. Blagbrough and A.P. Neal /Fluorescent Steroidal Lipopolyamine Conjugates
67
H,N
H,N
H,N
O
OH
Figure 2
Results and Discussion Design and Synthesis of Functionalised Polyamine-Steroid Conjugates: The synthesis of the small molecule ligands 3 and 4 is described in outline. Spermine 2 was unsymmetrically tri-tert-Boc protected in a one-pot reaction (Fig. 3) [39, 40]. Ethyl trifluoroacetate reacts rapidly and cleanly with primary amines, allowing poly-Boc protection of all the remaining amine functional groups. Trifluoroacetamides are easily cleaved, in the presence of Boc groups, at pH 11 and treatment with conc, aqueous NH3 gave one free primary amine functional group in 50% overall yield. Acylation with cholesteryl chloroformate gave the protected cholesteryl carbamate 12 in 95% yield [40].
68
I.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
CF-CCLEt, MeOH
Selective protection of | primary amine breaks 3.4.3 symmetry
i. Boc20,MeOH ii. Conc. aq. NH3
One pot multi-gram synthesis and purification of protected polyamines Cholesteryl chloroformate Et3N, CH2CI2
Boc
12
Figure 3
For fluorescent microscopy to study the intracellular phenomena of non-viral gene delivery, Byk et al. reported a fluorescent derivative of a polyamine conjugated to two C18lipid chains and they have synthesised fluorescent plasmid DNA using rhodamine-type fluorophores [28, 41]. Polyethylenimine (PEI) is a cationic aziridine polymer with known transfection activity [42]. Mikos and co-workers used Oregon Green labelled PEI in multicolour fluorescence studies and provided initial evidence that internalised DNA-PEI complex can be tracked to the nucleus [43]. We have set ourselves the goal of lighting-up the inside of the cell with wavelength-specific small molecule fluorescent probes and monitoring the processes in, and elucidating the barriers to efficient gene delivery. For the introduction of a fluorescent label to cholesteryl, we decided to hydroborate the cholesterol A5-alkene functional group [44]. Tri-Boc protected cholesteryl carbamate 12 was reacted with borane-dimethyl sulfide (BH3.dms) for 4 h in anhydrous CH2Cl2- The organoborane was oxidised for 30 min with basic (NaHCO3) H2O2. The use of BH3.dms complex provides mild conditions for the selective reaction of an alkene in the presence of carbamate functional groups, despite the poor reactivity of the cholesteryl alkene due to a significant steric constraint. The methyl substituents on the top face (p*-face) of cholesteryl direct the syn-addition of borane to the a-face in the major product. The desired 6ochydroxy steroid 13, with a trans-fused AB ring junction, is obtained on oxidative work-up and after chromatography in 70% yield. In addition, a small fraction of the BH3.dms adds to the P-face affording the cis-fused AB, 6f3-OH stereoisomer 14 (5%) which is more polar
l.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
69
and these products were separated by column chromatography over silica gel. Given that hydroboration is a syn-addition, in the less-polar major product H-6ax displays two J ax-ax and one Jax-eq coupling at 5 3.35 ppm, dt (J = 5 and 11 Hz) (Fig. 4), Rf 0.5, EtOAc-hexane 1:1 v/v, 13 C-NMR: 5 52.3 (C-5), 68.4 (C-6), 73.9 (C-3) ppm, HR FAB-MS found 933.7321 (M+ + 1), C53H97N4O9 requires 933.7256, was the desired trans-fused AB system. On further elution, a minor product was obtained, Rf 0.1, EtOAc-hexane 1:1 v/v, HR FAB-MS found 933.7274 (M+ + 1), this was assigned to the p-hydroxy isomer from a nonstereospecific reaction [45]. The cis-fused AB ring junction, in the minor product 14, mimics the shape found in the naturally occurring steroid bile acids (Fig.5), e.g. lithocholic acid, polyamine amides of which, e.g. 4 we have shown to condense duplex DNA [46, 47]. Boc
I A^L^S
>^z^T^^ I
Boc
0
xk
i. BH3.dms, CH2C12, 4 h ii. H2O2/NaHCO3, 30 min Boc Boc
Major product, H-6 has two Jax-ax and one Jax-eq coupling (3.35 ppm, dt, J= 5, 11 Hz) Me T I Max
>--^-
~1
I
3.35 Figure 4
T
r*—T\—7
^/-4*-^/^H eq
70
I.S. Blagbrough and A.P. Neat / Fluorescent Steroidal Upopolyaminc Conjugates
Boc
Boc
Figure 5
Similarly, we prepared spermidine mimic target molecule 4 from TFA deprotection of the three Boc protecting groups in 15, itself obtained by acylation of amine 11 with lithocholic acid (DCC, HOBt, CH2Cl2). In order to mimic the positive charge distribution of naturally occurring spermine 2 in the target molecules, we have investigated a reductive alkylation approach using N1,N2,N3-tri-Boc spermine 11. N-Cbz protected 6-aminohexan-l-ol was prepared using benzyl chloroformate. The alcohol was oxidised under Swern conditions (anhydrous DMSO-oxalyl chloride) to the corresponding aldehyde followed by reductive amination with N1,N2,N3-tri-Boc spermine 11 [39, 40]. The resulting secondary amine was Boc protected to form tetra-Boc-mono-Cbz pentamine with methylene spacing 3.4.3.6 16. The Z group, orthogonal to Boc, was then cleaved by palladium catalysed hydrogenolysis or by transfer hydrogenation using cyclohexene as a H2-donor. These steps achieved an unsymmetrical extension of N1,N2,N3-tri-Boc spermine 11 with assured regiochemistry in the product 17 after acylation with cholesteryl chloroformate. The resulting tetra-Boc protected 3.4.3.6-cholesteryl carbamate 17 was hydroborated using the procedure outlined above and the major product was isolated, purified, and identified as secondary alcohol 18 FAB-MS found 1154 (M + Na+), C64H117N5O11 requires 1131.
I.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
71
Fluorescent labels were then introduced to these lipopolyamine conjugates to generate our designed fluorescent target molecules. Fmoc, as either a fluorescent tag or as a primary amine protecting group, was first introduced by esterification (DCC, DMAP) with the short alkyl chain 5-Fmoc-aminopentanoic acid (6APA), chosen as an appropriate and pragmatic spacer. The secondary alcohols of cholestane B-ring 13 affording ester 19, and of lithocholic acid A-ring 15 affording 20 were reacted in this manner (Fig. 6). Further manipulation of these esters 19 and 20 by regioselective deprotection of one primary amine functional group by fluoride facilitated Fmoc removal with TBAF [48], and then reaction with fluorescein isothiocyanate afforded, after TFA removal of the Boc protecting groups, thiourea target molecules 21 and 22 respectively. It is significant that the secondary alkyl cholesteryl carbamate functional group remains intact during this poly-Boc deprotection. Final purification of these water soluble poly-trifluoroacetic acid salts afforded the desired fluorescent steroidal lipopolyamine conjugates which can react with DNA. 13
Fmoc-SAPA-OH DCC, DMAP CH2CI2
Fmoc-5APA-OH 15
DCC, DMAP CH2CI2
Fmoc,K
i. TBAF, THF 19
ii. Fluorescein isothiocyanate B3N, CH2CI2 iii. TFA, CH2CI2 20
i. TBAF, THF ii. Fluorescein isothiocyanate Et3N, CH2CI2 iii. TFA, CH2CI2
Figure 6
21
72
I.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
Monitoring Lipopolyamine Interactions with DNA: Spectroscopic assays allow us to monitor DNA condensation, the first stage in lipopolyamine mediated gene delivery, using our designed probes 3, 4, 21, and 22 (Fig. 7). These DNA assays include both ethidium bromide fluorescence quenching [49] and light scattering (A = 320 nm) [50] as, at this stage, the DNA is compacted into toroidal particles whose formation can be monitored by light scattering [50]. Using these techniques, we have shown that our synthetic steroidal lipopolyamines e.g. 4 and cholesteryl carbamate 23 from Boc-deprotection of 12 [40], an N1-acylated regioisomer of GL #67 7 [17], condense DNA more efficiently than the natural polyamines spermidine 1 and spermine 2. The synthetic lithocholic acid amide of spermine 2, converted into an ester and then a thiourea with fluorescein isothiocyanate, spermidine equivalent 22, condensed both calf thymus and synthetic DNA with the same efficiency as spermidine 1 in the ethidium bromide assay. Conclusions and Future Studies Using our designed steroidal lipopolyamine probes (Fig. 7), we are studying DNAlipopolyamine complexes with respect to their formation by DNA condensation. The design and synthesis of fluorescent lipopolyamines allows us to study the intracellular events during transfection. Comparable DNA-binding efficiency to that of the unlabelled lipopolyamines and robust fluorescent spectral properties across the varying cellular pH range are desirable properties in these ligands. We have achieved a controlled chain extension of suitably protected polyamines using reductive alkylation [39] to mimic spermine 2 rather than spermidine 1 in the target molecules. A practical method for the efficient hydroboration of cholesteryl carbamates has allowed us to prepare our designed trans-AB steroidal lipopolyamines, together with the corresponding cis-AB ring junction as the minor product of the hydroboration reaction or from naturally occurring bile acids. We are able to introduce fluorophores of choice by our Fmoc-chemistry. In this way, we have synthesised the beginnings of a library of fluorescent lipopolyamine probes based on steroid moieties as the lipophilic unit. The design in our versatile synthetic route to Fmoc protected aminoesters of cholesteryl carbamate 19 and lithocholic acid polyamine amide 20 allows a range of selected fluorophores to be readily incorporated e.g. fluorescein isothiocyanate which is a widely used fluorescent probe in confocal microscopy (cf 3 and 21, and 4 and 22). The availability of a wide range of fluorophores is important in order that a library of fluorescent derivatives may be accessed via this Fmoc deprotection route. N-Hydroxysuccinimide activated reagents to introduce these fluorophores include Oregon Green 488 24 (Fig. 7), a difluorinated fluorescein analogue which offers improved spectral properties over the physiological pH range involved in transfection processes and Alexa Fluor 350 25. Together with fluorescein, fluorophores 24 and 25 are negatively charged. Lissamine Rhodamine B 26 and other rhodamine-type fluorophores incorporate positive charges; Fmoc 27 and BODIPY-FL 28 fluorophores are lipophilic. Steroid lipopolyamine conjugates of these fluorophores will have different efficiencies in the modes of conjugate binding to DNA. As well as our studies on lipopolyamine-mediated DNA condensation, we are optimising these probes to use them in monitoring the steps of gene delivery within cells. At present, a poor understanding of the mechanisms of action of non-viral vectors remains an important unresolved issue. With a greater knowledge of these mechanisms [51, 52], new non-viral vectors with improved transfection efficiency can be rationally designed. In conclusion, these results help us to design more efficient molecular probes for DNA-binding in this area of non-viral gene delivery using lipopolyamine moieties.
I.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
HO"
28 Figure 7
73
74
I.S.
Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
Acknowledgements We acknowledge financial support from the EPSRC (studentship to A.P.N.). The results in these studies in synthetic and analytical medicinal chemistry, related to "Nucleic Acids Ligands for Gene Therapy", are in part based upon our recent oral and poster presentations, published in various abstracts [34–38].
References [1] J. C. Davies, D. M. Geddes and E. W. F. W. Alton, J. Gene Med. 3 (2001) 409. [2] H. C. Chiou, M. A. Lucas, C. C. Coffin, M. G. Banaszczyk, C. R. Ill and C. P. Lollo, Expert Opin. Bioi Therapy 1 (2001) 629. [3] M. K. L. Chuah, D. Collen and T. van den Driessche, J. Gene Med. 3 (2001) 3. [4] M. N. Barnes and T. B. Pustilnik, Curr. Opin. Obstet. Gynecol. 13 (2001) 47. [5] L. Xu, K. F. Pirollo and E. H. Chang, J. Cont. Rel. 74 (2001) 115. [6] T. Bettinger and M. L. Read, Curr. Opin. Molec. Therapeut. 3 (2001) 116. [7] V. Weissig and V. P. Torchilin, Adv. Drug Del. Rev. 49 (2001) 127. [8] N. J. Silman and A. R. Fooks, Curr. Opin. Molec. Therapeut. 2 (2000) 524. [9] L. J. Chang and J. He, Curr. Opin. Molec. Therapeut. 3 (2001) 468. [10] Mergia, S. Chari, D. L. Kolson, M. M. Goodenow and T. Ciccarone, Virology 280 (2001) 243. [11]S. Blagbrough, S. Carrington and A. J. Geall, Pharmaceutical Sciences 3 (1997) 223. [12]G. Schatzlein, Anti-Cancer Drugs 12 (2001) 275. [13] W. Poulon and L. W. Seymour, Adv. Drug Del. Rev. 46 (2001) 187. [14]M. D. Brown, A. G. Schatzlein and I. F. Uchegbu, Int. J. Pharm. 229 (2001) 1. [15]H. Lee, S. K. R. Williams, S. D. Allison and T. J. Anchordoquy, Anal. Chem. 73 (2001) 837. [16]S. Walker, M. J. Sofia, R. Kakarla, N. A. Kogan, L. Wierichs, C. B. Longley, K. Bruker, H. R. Axelrod, S. Midha, S. Babu and D. Kahne, Proc. Natl. Acad. Sci. USA 93 (1996) 1585. [17]E. R. Lee, J. Marshall, C. S. Siegel, C. W. Jiang, N. S. Yew, M. R. Nichols, J. B. Nietupski, R. J. Ziegler, M. B. Lane, K. X. Wang, N. C. Wan, R. K. Scheule, D. J. Harris, A. E. Smith and S. H. Cheng, Human Gene Therapy, 7 (1996) 1701. [18] Y. Kwok, Y. S. Yang and K. G. Rice, Curr. Opin. Molec. Therapeut. 3 (2001) 142. [19]G. Byk, B. Wetzer, M. Frederic, C. Dubertret, B. Pitard, G. Jaslin and D. Scherman, J. Med. Chem. 43 (2000) 4377. [20] Wetzer, G. Byk, M. Frederic, M. Airiau, F. Blanche, B. Pitard and D. Scherman, Biochem. J. 356 (2001) 747. [21]Dauty, J. S. Remy, T. Blessing and J.-P. Behr, J. Am. Chem. Soc. 123 (2001) 9227. [22] M. A. llies and A. T. Balaban, Expert Opin. Therap. Pat. 11 (2001) 1729. [23] C. W. Poulon, Adv. Drug Del. Rev. 34 (1998) 51. [24] M. Johnson-Saliba and D. A. Jans, Curr. Drug Targets 2 (2001) 371. [25] S. Remy, C. Sirlin, P. Vierling and J.-P. Behr, Bioconjugate Chem. 5 (1994) 647. [26] R. I. Mahato, A. Rolland and E. Tomlinson, Pharm. Res. 14 (1997) 853. [27] J.-P. Behr, B. Demeneix, J.-P. Loeffler and J. Perez-Mutul, Proc. Natl. Acad. Sci. USA 86 (1989) 6982. [28] G. Byk, C. Dubertret, V. Escriou, M. Frederic, G. Jaslin, R. Rangara, B. Pitard, J. Crouzet, P. Wils, B. Schwartz and D. Scherman, J. Med. Chem. 41 (1998) 224. [29] T. Ren, G. S. Zhang, F. Liu and D. X. Liu, Bioorg. Med. Chem. Lett. 10 (2000) 891. [30] Nakamura, H. Isobe, N. Tomila, M. Sawamura, S. Jinno and H. Okayama, Angewandte Chemie 39 (2000) 4254. [31] P. Camilleri, A. Kremer, A. J. Edwards. K. H. Jennings, O. Jenkins, I. Marshall, C. McGregor, W. Neville, S. Q. Rice, R. J. Smith, M. J. Wilkinson and A. J. Kirby, Chem. Commiin. (2000) 1253 and 1553. [32]C. McGregor, C. Perrin, M. Monck, P. Camilleri and A. J. Kirby, J. Am. Chem. Soc. 123 (2001) 6215. [33]Ronsin, C. Perrin, P. Guedat, A. Kremer, P. Camilleri and A. J. Kirby, Chem. Commun. (2001) 2234. [34] P. Neal and 1. S. Blagbrough, J. Pharm. Pharmacol. 52 (2000) S116. [35] P. Neal and I. S. Blagbrough, Abs. Pap. Am. Client. Soc. 221 (2001) 332-MEDI. Part 2. [36] P. Neal and 1. S. Blagbrough, Abs. Pap. Am. Chem. Soc. 221 (2001) 352-MED1. Part 2. [37] P. Neal and I. S. Blagbrough, J. Pharm. Phamiacol. 53 (2001) S1.
I.S. Blagbrough and A.P. Neal / Fluorescent Steroidal Lipopolyamine Conjugates
[38] P. Neal and I. S. Blagbrough, J. Pharm. Pharmacol. 53 (2001) S245. [39] J. Geall and I. S. Blagbrough, Tetrahedron 56 (2000) 2449. [40] J. Geall, R. J. Taylor, M. E. Earll, M. A. W. Eaton and I. S. Blagbrough, Bioconjugate Chem. 11 (2000) 314. [41] Neves, G. Byk, V. Escriou, F. Bussone, D. Scherman and P. Wils, Bioconjugate Chem. 11 (2000) 51. [42] O. Boussif, F. Lezoualch, M. A. Zanta, M. D. Mergny, D. Scherman, B. Demeneix and J.-P. Behr, Proc. Natl. Acad. Sci. USA 92 (1995) 7297. [43] W. T. Godbey, K. K. Wu and A. G. Mikos, Proc. Nad. Acad. Sci. USA 96 (1999) 5177. [44] R. Tavares, T. Randoux, J.-C. Braekman and D. Daloze, Tetrahedron 49 (1993) 5079. [45] E. Jung and T. W. Johnson, Tetrahedron 57 (2001) 1449. [46] J. Geall, D. Al-Hadithi and I. S. Blagbrough, Chem. Commun. (1998) 2035. [47] S. Blagbrough, D. Al-Hadithi and A. J. Geall, Tetrahedron 56 (2000) 3439. [48]Ueki and M. Amemiya, Tet. Lett. 28 (1987) 6617. [49] J. Geall and I. S. Blagbrough, J. Pharm. Biomed. Anal. 22 (2000) 849. [50] J. Geall, M. A. W. Eaton, T. Baker, C. Catterall and I. S. Blagbrough, FEBS Lett. 459 (1999) 337. [51 ] Labat-Moleur, A. M. Steffan, C. Brisson, H. Perron, O. Feugeas, P. Furstenberger, F. Oberling, E. Brambilla and J.-P. Behr, Gene Therapy 3 (1996) 1010. [52]M. Gorman, M. Aikawa, B. Fox, E. Fox, C. Lapuz, B. Michaud, H. Nguyen, E. Roche, T. Sawa and J. P. WienerKronish, Gene Therapy 4 (1997) 983.
76
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Synthesis of (S)-2-Amino-oleic Acid and Other Bioactive Molecules through Glutamate Semialdehydes V. Constantinou-Kokotoua, V. Magriotia, T. Makridisb and G. Kokotosb a
Chemical Laboratories, Agricultural University of Athens, Greece >ry of Organic Chemistry, Department of Chemistry, University of Athens, Greece Laboratory
b
Abstract. 2-Amino-oleic acid is of great interest, because it is used as a tissue treatment for advanced generation bioprostheses. Efficient methodology for the synthesis of enantiopure 2-amino-oleic acid was developed using either tert-butyl (2S)-2[bis(fert-butoxycarbonyl)amino]-5-oxopentanoate or methyl (2S)-2-[bis(tertbutoxycarbonyl)amino]-5-oxopentanoate as key-intermediates. Furthermore, a route to enantiopure (0-functionalized a-amino acids was developed.
Introduction In recent years, the field of amino acids and peptides has gained enormous popularity and relevance, particularly with the emergence of unnatural analogues as components of compounds with therapeutic potential [1]. The need to replace natural amino acids in peptides with non-proteinogenic counterparts in order to obtain new medicinal agents, exhibiting better binding to specific receptors and more potent inhibition of target enzymes, has stimulated a great deal of innovation on synthetic methods. Lipidic a-amino acids (LAAs) is a class of unnatural amino acids with long aliphatic side chains. LAAs and lipidic peptides combine the structural features of lipids with those of amino acids and peptides [2]. These compounds are lipophilic due to the long carbon chains, yet show polar, chemical and conformational behaviour characteristics of amino acids and peptides. Amides and esters of saturated LAAs as well as lipopeptides present interesting biological properties [3–5]. Racemic LAAs and their oligomers have been used as drug delivery systems [6,7] and for the construction of a lipid-core-peptide system for the development of adjuvant semisynthetic vaccines [8]. Long chain 2-amino alcohols and 1,2-diamines obtained from LAAs exhibit interesting cytotoxic [9–11] and anti-inflammatory activity [12]. The enantiomeric synthesis of saturated LAAs, based on the oxidative cleavage of amino diols, obtained by the regioselective opening of enantiomerically enriched 2,3-epoxy alcohols, has been presented [13]. Also, the synthesis of optically active 8,E-unsaturated LAAs, including a-amino arachidonic acid, has been achieved through a Wittig reaction on the suitable glutamate semialdehyde [ 14].
V. Constantinou-Kokotou et al. / Synthesis of (S)-2-Amino-oleic Acid
77
(S)-2-Amino-oleic Acid Efficient methodology for the synthesis of optically pure 2-amino-oleic acid (1) (Figure 1) has been developed by our group using either tert-butyl (2S)-2-[bis(tertbutoxycarbonyl)amino]-5-oxopentanoate or methyl (2S)-2-[bis(tertbutoxycarbonyl)amino]-5-oxopentanoate as key-intermediates.
o OH NH2
J
/
Figure 1. (S)-2-Amino-oleic acid
Applications of 2-Amino-oleic Acid: Racemic 2-amino-oleic acid is used as a tissue treatment for advanced generation bioprostheses [15]. Valvular defects are among the most frequent causes of circulatory insufficiency. Surgical treatment, which usually consists in the implantation of prosthetic valve, improves haemodynamic conditions through anatomic correction. Heart valve prosthesis implanted for that purpose are divided into mechanical and biological valves, ( valve leaflets derived from human or animal tissues-bioprosthesis) [16]. The introduction of tissues crossed-linked with formaldehyde or glutaraldehyde into heart valve production ensured valve sterility. Calcification is a frequent cause of the clinical failures of glutaraldehyde-pretreated bioprosthetic heart valves fabricated from glutaraldehyde-cross-linked porcine aortic valves. Racemic 2-amino-oleic acid has been shown in in vivo studies to be a promising anti-calcification agent. In fact, the incubation of pericardial valve cross-linked with glutaraldehyde with the admixture of 2-amino-oleic acid revealed reduced calcium ions diffusion through such tissue and a nearly 20-fold decrease in calcium deposition [17,18]. The presence of the amino group is very important, because it seems that it binds with aldehyde residues of glutaraldehyde [19]. The existing synthetic method [15,20] starts from oleic acid and through a number of modifications at the exposition leads to the racemic 2-amino-oleic acid. However, this sequence of reactions may be applied only for the synthesis of racemic product. On the contrary, the proposed, by our group, methodology permits the synthesis of either (S)- or (R)- enantiomer depending on the chirality of the starting material, prepared from (S)- or (R)-glutamic acid. (S)-2-Aminooleic acid is expected to find a lot of applications for the construction of either peptide or lipid mimetics. Synthesis of (S)-2-Amino-oleic Acid Through tert-Butyl (2S)-2-[bis(tertButoxycarbonyl)amino]-5-oxopentanoate: The natural amino acid L-glutamic acid was used as inexpensive starting material. A strategy combining N-Boc group and a-tert-butyl ester group was chosen so that both groups may be removed simultaneously at the last step of the synthesis. Thus, (S)-Boc-Glu(OMe)-OBu1 (2) was treated with Boc2O in the presence of DMAP to afford derivative 3 (Figure 2). Selective reduction of the Y-methyl ester of compound 3 using DIBALH afforded aldehyde 4 [21].
78
V. Constantinou-Kokotou et al. / Synthesis of(S)-2-Amino-oleic Acid
u
u o
Jl
MeO.A^^AoBu' NHBoc
DIBALH Et
BOC2
°-DMAP
' *°
-78°C
i u N(Boc)2
N(Boc)2
Figure 2. Synthesis of re/t-butyl (2S)-2-[bis(ferr-butoxycarbonyl)amino]-5-oxopentanoate
Treatment of the key intermediate aldehyde 4 with the ylide, generated from EtOOC(CH2)3P*Ph3 Br and KHMDS yielded the Z-olefin 5 (Figure 3). The corresponding saturated compound 6, obtained from 5 after catalytic hydrogenation, was selectively reduced under controlled conditions by DBBALH to afford aldehyde 7. Wittig reaction of this aldehyde with the ylide generated from CrtyCHWsP^PhB Br", followed by subsequent removal of Boc and Bu' protecting groups, gave (5)-2-amino-oleic acid [21]. EtOOC(CH2)3PPh3Br. ^ KHMDS. THF, -78 °C
N(Boc)2 •OBu1
EtO
N(Boc)2
N(Boc)2
EtO
H2.10%Pd/C
OBu1
DIBALH, Et2O
r o
KHMDS, toluene, -78 C
-78 °C
N(Boc)2
1.TFA/CH2CI250%, 2 - neutralization
8
1
Figure 3. Synthesis of (5)-2-amino-oleic acid using aldehyde 4
Synthesis of (S)-2-Amino-oleic Acid Through Methyl (2S)-2-[bis(tertButoxycarbonyl)amino]-5-oxopentanoate: An improved route to (5)-2-amino-oleic acid used methyl (25)-2-[bis(/e/t-butoxycarbonyl)amino]-5-oxopentanoate (11) as key intermediate. The synthesis of this aldehyde [14] is depicted in Figure 4. Suitably protected glutamic acid semialdehyde 11 and related compounds have been proven useful synthons for the synthesis of biologically interesting compounds [22-25]. L-Glu
2. BoczO, Et3N, MeOH
MeO
-
O
0 U
.. T
OMe
N(Boc)2 10
g
DIBALH. Et2O .78oc
O
H^kx-XjAoMe NfBocfe 11
Figure 4. Synthesis of methyl (2S)-2-[bis(fe/-r-butoxycarbonyl)amino]-5-oxopentanoate
79
V. Constantinou-Kokotou et al. /Synthesis of(S)-2-Amino-oleic Acid
The Horner-Wadsworth-Emmons olefination reaction of aldehyde 11 with the phosphonate anion generated from triethyl-4-phosphonocrotonate by treatment with LiOH afforded compound 12 (Figure 5). After hydrogenation of 12, compound 13 was selectively reduced by DIBALH to afford aldehyde 14. Wittig reaction of 14 with the ylide generated by treatment of CH3(CH2)8P+Ph3 Bf with KHMDS produced compound 15. This fully protected a-amino acid was identified as Z-olefin after !H NMR analysis. o II
H2, 10%Pd/C
EtOOCCH=CHCH2P(OEt)2
11
OMe N(Boc)2
EtO
LiOH, THF
12
DIBALH, Et2O EtO
;5
OMe N(Boc)2
-78°C
j OMe N(Boc)2
C9H19PPhfB® KHMDS, toluene -78°C
14
13
s
Y OMe N(Boc)2
15
Figure 5. Synthesis of protected (S)-2-amino-oleic acid
Free 2-amino-oleic acid and derivatives were prepared as depicted in Figure 6. Both Boc groups of 15 were removed by treatment with HC1 in Et2O to produce compound 17. The saponification of 15 by treatment with 1 N NaOH was proved a very slow reaction. One out of the two Boc groups of 15 was removed selectively by treatment with Mg(ClO4)2 [26]. In the case of Af-monoprotected compound 16 the saponification was not complete after 24 h. Since racemization may occur after prolonged treatment under alkaline conditions, to prepare free 2-amino-oleic acid 1, it is recommended first to remove both Boc groups and then the methyl ester group by treatment with NaOH. Mg(CIO4)2, MeCN
is
.
°
^^r^fV™8 NHBoc 16
4N HCI/Et2O
OMe NH2.HCI
INNaOH, MeOH
5
I
OH
NH2
17
Figure 6. Free (S)-2-amino-oleic acid and derivatives
Both proposed procedures to 2-amino-oleic acid are free of racemization. To verify that point, (5)-2-amino-oleic acid obtained as depicted in Figure 3 was converted into its methyl ester. This compound as well as compound 17 were converted into their corresponding amides with (+)- and (-)-Mosher acid [27]. !H and I9F NMR analysis of all Mosher amides indicated absence of any diastereomeric signal. Both methods for the synthesis of (5)-2-amino-oleic acid, are simple and efficient. However, the latter presents higher overall yield.
80
V. Constantinou-Kokotou et al. / Synthesis of (S)-2-Amino-oleic Acid
Synthesis of Enantiopure -Functionalized a-Amino Acids The stereoselective synthesis of a-amino acids and the construction of bioactive chiral molecules using amino acids has been achieved through a variety of methods [28]. Approaches using regioselective functionalization of readily available chiral building blocks, e.g. aldehydes obtained from amino acids, are especially attractive for the synthesis of unnatural amino acids. An efficient method to prepare enantiopure co-functionalized ctamino acids using the protected glutamic acid y-aldehyde 11 as key-intermediate was developed. Our attention was focused on long chain derivatives, because LAAs are of great importance for the synthesis of drug delivery systems and bioactive lipid mimetics. The Wittig olefination of aldehyde 11 with oxido, carboxy or amino ylides may directly lead to a-amino acids containing a free functional group in the side chain. Maryanoff [29] has studied the reaction of hexanal and benzaldehyde with triphenylphosphonium ylides containing anionic nucleophilic groups in their side chain. Following Maryanoff's procedure the reactions of 11 with the ylides generated from phosphonium salts of 10-bromo-decan-l-ol and 11-bromo-undecanoic acid were tested. In each case, the desired product was not obtained. Thus, we decided to use an ylide containing an co-protected hydroxy group. To prepare such ylides, a,to-alkane diols 20a,b were treated with TrtCl and DMAP in pyridine to produce mono-protected diols 21a,b (Figure 7). The free hydroxy group of 21a,b was activated by conversion to mesylate and the methanesulphonyloxy group was replaced by iodide ion. Iodides 22a,b were treated with PPh3 in MeCN under reflux to produce triphenylphosphonium salts 23a,b. HCL.OH TrtCl. DMAP TrtOv^OH Tin pyridine "" ^ 20a,b 21a,b
1 MsCI. Et3N
2Nal
TrtCL^I ™1 22a,b
PPh3. MeCN
TrtCX.PPhs*! *'n 23a,b
b 10 Figure 7. Synthesis of triphenylphosphonium salts 23a,b
Aldehyde 11 reacted with the ylides generated by treatment of 23a,b with KHMDS in toluene at 0 °C (Figure 8). The reactions were carried out at -78 °C and the protected amino acids 24a,b were produced in high yields. Both compounds were identified as Zolefins, by 'H NMR analysis. Methyl ester of amino acid 25 was obtained almost quantitatively by treatment of 24b with HC1 in THF. To check the enantiomeric purity, compound 25 was converted into the corresponding Mosher amides [27] and studied by I9F NMR analysis. An enantiomeric excess > 95% was indicated for 25 by the absence of any diastereomeric signal in the 19F NMR spectrum of each MTP amide.
81
V. Constantinou-Kokotou et al. / Synthesis of (S)-2-Amino-oleic Acid
OMe 23aor23b. . KHMDS, toluene N(Boc)2
OMe
4N HCI/THF
N(Boc)2 24a, n=4 24b, n=10
11
"OMe
H2, 10%Pd/C
NH2.HCI 25
"OMe N(Boc)2 26
Figure 8. Synthesis of o>hydroxy a-amino acid derivatives
Catalytic hydrogenation of 24b produced the saturated oo-hydroxy a-amino acid 26 (Figure 8). This derivative was chosen as a model compound and was converted to co-carboxy, a>oxo and co-amino a-amino acids as depicted in Figure 9. Oxidation of 26 using 2.5 equivalents of NaOCl in the presence of 4-acetamido-2,2,6,6-tetramethyl-l-piperidinyloxy free radical (AcNH-TEMPO) and Aliquat 336 [30] produced the to-carboxy amino acid 27. In the absence of the phase transfer catalyst and using 1.1 equivalent of NaOCl [31], the 00oxo derivative 28 was isolated. To prepare diamino acids, the hydroxy group of 26 was activated as its methanesulphonate and was converted directly into azide 29 by treatment with sodium azide in DMF at 60 °C in high yield. Catalytic hydrogenation of 29 produced the free o>-amino functionalized derivative 30, whereas in the presence of N-(9fluorenylmethoxycarbonyloxy)succinimide the selectively protected derivative 31 was isolated. NaOCl, AcNH-TEMPO, Aliquat, KBr, NaHCO3, CHjClj, H2O
NaOCl, AcNH-TEMPO, NaBr, NaHCO3, EtOAc, toluene, HSO
1 MsCI, Et3N 2 NaN3, DMF
Figure 9. Synthesis of protected co-functionalized a-amino acids
82
V. Constantinou-Kokotou et al. /Synthesis of(S)-2-Amino-oleic Acid
In conclusion, an efficient route for the synthesis of enantiopure o>-hydroxy, co-carboxy and (D-amino a-amino acids was developed. The length of the side chain depends on the starting alkanediol used to prepare the phosphonium salts. Both enantiomers of CDfunctionalized a-amino acids may be prepared, because the chirality of the final product depends on the chirality of the key-intermediate glutamic acid y-aldehyde 11 obtained from the available in both enantiomeric forms Glu. References [I] For reviews see: (a) J. Game, Angew. Chem., Int. Ed. Engl. 33 (1994) 1699. (b) A. E. P. Adang et al., Rec. Trav. Chim. Pays Bas 113 (1994) 63-78. (c) G. L. Olson et al., J. Med. Chem. 36 (1993) 3039. (d) A. Giannis and T. Roller, Angew. Chem., Int. Ed. Engl. 32 (1993) 1244. [2] W.A.Gibbons et al., Liebigs Ann. Chem. (1990) 1175. [3] G. Kokotos et al.. Int. J. Peptide Protein Res. 48 (1996) 160. [4] G. Kokotos et al., Amino Acids 11 (1996) 329. [5] A. Nicolaou et al., J. Pep. Sci. 3 (1997) 291. [6] I. Toth, J. Drug Targeting 2 (1994) 217. [7] I. Toth et al., J. Med. Chem. 42 (1999) 4010. [8] I. Toth et al., Tetrahedron Lett. 34 (1993) 3925. [9] G. Kokotos et al., Bioorg. Med. Chem. Lett. 8 (1998) 1525. [10] V. Constantinou-Kokotou et al., Anticancer Res. 18 (1998) 3439. [II] T. Markidis et al., Anticancer Res. 21 (2001)2835. [ 12] G. Kokotos et al, Lipids 34 (1999) 307. [ 13] G. Kokotos et al., Tetrahedron: Asymmetry 7 (1996) 857. [14] G. Kokotos et al., J. Org. Chem. 63 (1998) 3741. [15] J. M. Girardot, Prevention of Prosthesis Calcification, US Patent No. 4,976,733, 1990. [16] J. K. Nozynski et al., Med. Sci. Monit. 7 (2001) 550. [17] S. K. Khanna et al.. Thorax 36 (1981) 330. [18] J. P. Gott et al., Ann. Thorac. Surg. 53 (1992) 207. [19] W. Chen et al.. Circulation, 90 (1994) 323. [20] F. Amat Guerri, Grasas Aceites (Seville) 26 (1975) 90. [21] V. Constantinou-Kokotou et al., J. Peptide Res. 58 (2001) 325. [22] N. Hernandez and V. Martin, J. Org. Chem. 66 (2001) 4934. [23] T. Markidis and G. Kokotos, J. Org. Chem. 66 (2001) 1919. [24] A. Sutherland et al., Chem. Commun. 6 (1999) 555. [25] J. M. Padron et al.. Tetrahedron: Asymmetry 9 (1998) 3381. [26] J.A. Stafford et al.. Tetrahedron Lett. 34 (1993) 7873. [27] J. A. Dale et al., J. Org. Chem. 34 (1969) 2543. [28] (a) R. H. Williams, The Synthesis of Optically Active a-Amino Acids, Pergamon, New York, 1989. (b) G. M. Coppola and H. F. Schuster, Asymmetric Synthesis: Construction of Chiral Molecules Using Amino Acids, Wiley, New York, 1987. [29] B. E. Maryanoff et al, J. Am. Chem. Soc. 107 (1985) 217. [30] P. L. Anelli et al., J. Org. Chem. 52 (1987) 2559. [31 ] M. R. Leanna et al.. Tetrahedron Lett. 33 (1992) 5029.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
83
Development of New Methds for
Fluorometric Assays of Enzymes
C. Tzougraki Department of Chemistry, University of Athens, Panepistimiopolis, Zografou, Athens 15771, Greece
Abstract. This article describes the development of new fluorescent markers and their use in the solution or solid phase synthesis of fluorogenic and intramolecularly quenched fluorogenic substrates (IQFS) for proteases or Upases. The determination and study of several proteases, such as chymotrypsin, oxytokinase, prolylendopeptidase, aminopeptidase M, elastase and trypsin are reviewed. The results of an extensive study on the donoracceptor combination and their application in the synthesis of very sensitive IQFS, as well as in the conversion of p-nitroanilide substrates into IQFS, are given. A new general and convenient method for the direct synthesis of any fluorogenic substrate or IQFS by using exclusively solid phase peptide synthesis is also described.
Introduction Proteases play essential roles in the regulation of important biological processes and serve as possible indicators for the evaluation of various diseases [1]. Synthetic peptide substrates are widely used for studies of proteolytic enzymes and determination of their activities. These substrates must have the appropriate amino acid sequence for interaction with the active sites of enzymes. A number of convenient methods exist for studying the hydrolysis of these substrates, such as potentiometry, spectroscopy, etc. [2–5]. Spectroscopic methods offer the advantage of the small quantity of the sample required for the measurement. In particular, synthetic fluorogenic substrates have found a wide application in basic and clinical research for the determination of proteases, because the assays based on them are convenient and more sensitive than those based on chromogenic substrates. The fluorogenic substrates are of the general form P-A n -A n .| ...A2-A|-F, while the estimation of the enzyme activity is based on the increase of fluorescence observed upon enzymatic liberation of the fluorophore HF.
84
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
Several fluorescent compounds have been used as fluorophores HF, mostly coumarin derivatives, such as 7-amino-4-methylcoumarin (AMec) (1) [6], 7-amino-4(trifluoromethyl)coumarin (AFMec) (2) [7], 7-amino-3-phenylcoumarin [8], 7-amino3,4-benzocoumarin [8], 7-aminocoumarin-4-methanesulphonic acid [9], etc. (Scheme 1). AMec is the most common and widely used leaving group in fluorometric assays. A considerable number of proteases, such as chymotrypsin [6, 10], trypsin [10, 11], elastase [10], ami nopeptidase [12], thrombin [13, 14], renin [15], yglutamyltransferase [16] and oxytokinase [17], have been studied by using fluorogenic substrates, N-acyl-derivatives of AMec, and many of the latter are commercially available. However, the tremendous number and diversity of enzymes in nature, their appearance in impure form in coexistence with others and their low concentrations are the main reasons that demand more specific and sensitive analytical methods. Some years ago, while working mainly in the field of peptide chemistry, following the tradition of our laboratory, we were also engaged in the synthesis of heterocyclic compounds, and more specifically in the synthesis and study of coumarin and quinolinone derivatives [18]. Having found that some of them had very good fluorescence properties, we decided to examine them as potential markers in synthetic peptide substrates. Thus, we became involved in the field of enzymatic analysis, hoping to develop more sensitive and specific methods of enzymatic assays. In this review, some results from our efforts in the above field will be summarized. Coumarin and quinolinone derivatives as fluorescent markers: In order to evaluate new fluorescent markers, several substrates for chymotrypsin, Glt-Phe-NHRx, were prepared. [19], where RXNH2 are the compounds shown in Scheme 1, 7-amino-4methyl-2-quinolinone (3, AMeq), 7-amino-4-trifluoromethyl-2-quinolinone (4, AFMeq), 3-aminocoumarin (5, 3-Ac), 6-aminocoumarin (6, 6-Ac), 3-acetamido-6aminocoumarin (7, 3-AcNH-6-Ac) and 3-acetamido-8-aminocoumarin (8, 3-AcNH-8Ac). The synthesis of compounds 5, 8 and 9 has been described in Ref. 18 and that of 4 in Ref. 20. AMeq was prepared according to the literature [21]. The fluorescence properties of the free amines and those of the corresponding substrates were examined [19] and compared with those of others known from the literature.
H,N
AMec AFMec AMeq AFMeq
X = NH X = NH
= CH3 . = CF3 = CH3 = CF3
5 6 7 8
3-Ac 6-Ac 3-AcNH-6-Ac 3-AcNH-8-Ac
R, R, R, R,
= NH2 =H = NHCOCH, = NHCOCH,
Scheme 1: Coumarin and quinolinone derivatives used as fluorescent markers.
R2 = R} = H R2 = NH2, R, = H R2 = NH2. R, = H R2 = H. R, = NH2
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
85
Table 1. Fluorescence properties of substituted coumarins and quinolinonesa Rel. Fluor.b
Maxima
Compound
1
0.98 345 445 AMec 365 0.23 490C AFMec 1.00 344 418 AMeq 3 0.31 360 470 AFMeq 4 0.14 370 333 9 Glt-Phe-NH-Meq 0.07 340 Glt-Phe-NH-FMeq 390 10 1.0 uM solution in 0.05 M TES, pH 8, containing 1% DMSO. D The fluorescence intensities are relative to the intensity of AMeq on an equimolar basis. c 495 in Ref.[7a]. 2
i (b) lOr
s
350
40O
450
500
Wavelength (nm)
400
450
500
*
Figure 1. Fluorescence spectra of AMeq ( ), a: X^x = 344 nm; P: XeX = 360 nm, and Glt-Phe-NH-Meq (—-), a: Xex = 333 nm; (3: A^ = 360 nm. 1.0 uM solution in 0.05 M TES, pH 8, containing 1% DMSO.
Among the compounds examined, AMeq and AFMeq had the best fluorescence properties. Their excitation and emission maxima and their relative fluorescence intensities are distinctly different than those of their N-acyl derivatives, as shown in Table 1. Compounds AMec and AFMec are also listed for comparison. As expected, the replacement of the methyl group of AMeq by the electron withdrawing trifluoromethyl group in AFMeq produced a red shift of 52 nm and a decrease in fluorescence intensity [22]. Both 2-quinolinone derivatives AMeq and AFMeq exhibited a blue shift of approximately 25 nm and greater fluorescence intensity than their coumarin analogs AMec and AFMec respectively [20]. At maximum wavelengths the relative fluorescence intensities of AMeq and AFMeq are only 7- and 4.5-fold greater than those of their corresponding N-acyl derivatives respectively (Figure la). However, at XeX = 360 nm and A«m = 435 nm AMeq has a relative fluorescence approximately 1000-fold greater than that of an equimolar solution of the substrate, retaining 28% of its maximum fluorescence (Figure Ib).
86
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
Similarly, the relative intensity of AFMeq at A«x = 380 nm and Xem = 480 nm is 850fold greater than that of its N-acyl derivative, retaining 41% of its maximum. At the mentioned wavelengths the presence of the substrate in each case does not interfere, and enzymatic hydrolyses can be followed by measuring the development of fluorescence. As has been reported by Zimmerman et al. [6], AMec possessed a relative fluorescence approximately 700-fold greater than that of an equi molar amount of the substrate, retaining 22% of the maximum fluorescence. The results of the performed study showed that the 2-quinolinone derivatives AMeq and AFMeq would be exceptionally useful fluorophores in synthetic peptide substrates for proteases, because they are highly fluorescent, easily prepared in one step synthesis and they can be acylated by amino acids in high yield. Having proved the utility of the proposed fluorescent markers, we further proceeded to their application for the determination and study of several proteases. Studies of several proteases Determination of chymotrypsin: Chymotrypsin (CT) and CT-like enzymes play an important role in metabolism by participating in many processes, such as digestion, fertilization of ova, inhibition of thrombin-induced platelet aggregation, peptide hormone generation, and catabolism, etc. Table 2. Kinetic parameters of several fluorogenic substrates' Compound
1, V -1 Detection limit Km Kcat f^-m [mM] [M-'s1] [ng/ml] 47 9 Glt-Phe-NH-Meq 0.50 10 10 Glt-Phe-NH-FMeq 99 0.28 35 11 0.67 Glt-Phe-NH-Mec 78 500 12 Glt-Leu-Phe-NH-Meq 0.62 0.7 710 13 Glt-Leu-Phe-NH-FMeq 0.33 367 10 14 Glt-Tyr-NH-Meq 0.13 630 Glt-Leu-Tyr-NH-Meq 0.22 15 2100 16 TFA-H-Ala-Ala-Phe-NH-Mec 0.50 1660 25 a 1.0 uM solution in 0.05 M TES, pH 8, containing 1% DMSO.
Ref. [19] [20] [6, 10] [20] [25] [25] [3, 10]
From the above mentioned study, conducted on the model compounds, it was already concluded that the compound Glt-Phe-NH-Meq (9) was a suitable substrate for chymotrypsin. Its detection limit was 10 ng/ml, 50 times less than that obtained by using the corresponding substrate of AMec, Glt-Phe-NH-Mec (11) (Table 2). A concentration of 10 ng/ml of the enzyme chymotrypsin has been found in the adults' serum by radioimmunoassay [23]. This finding requires a higher sensitivity assay. Therefore, we tried further to increase the sensitivity of the assay by extending the peptide backbone, in order to take advantage of the role of the amino acid at the P2 position on the binding affinity between substrate and enzyme. It is known that chymotrypsin hydrolyses long peptide amides more efficiently than short ones and that bulky non-polar residues at the P2 position interact with the side chain of isoleucine-99 of chymotrypsin [24]. Thus the substrates Glt-Leu-Phe-NH-Meq (12) and Glt-Leu-PheNH-FMeq (13) with leucine at P2 position were prepared and their kinetic parameters
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
87
were measured. The results of their hydrolysis catalyzed by chymotrypsin are given in Table 2, along with others for comparison. All four substrates 9, 10, 12 and 13 were specific for chymotrypsin showing no detectable hydrolysis when incubated with porcine pancreatic or leukocyte elastase. As expected, the presence of the amino acid leucine at the P2 position increased the kcat/Km value, while it had no considerable effect on the Km value. It also increased the sensitivity of the assays. As little as 0.7 ng/ml of enzyme can be detected using the substrate Glt-Leu-Phe-NH-Meq, which was the most sensitive fluorogenic substrate reported. The assays using the substrates containing the fluorescent marker AMeq were more sensitive than those using the corresponding substrates containing AFMeq. However, as the assays using the latter substrates were conducted at longer wavelengths, the background fluorescence exhibited by most biological matter, when excited in the spectral region 330–360 nm, could be avoided. Later, the substrates Glt-Tyr-NH-Meq (14) and Glt-Leu-Tyr-NH-Meq (15) with tyrosine at P1 position were synthesized [25]. The improved kinetic parameters of 14 and 15 compared to those of 9 and 12 proved that the presence of tyrosine instead of phenylalanine increases the affinity to the enzyme. The substrate 15 had the highest kcat/K.m value among the fluorogenic ones reported. All six substrates 9, 10, 12–15, are suitable for a sensitive and convenient determination of chymotrypsin. Determination of Oxytokinase: The determination of the activity of serum oxytocinasecystine aminopeptidase (CAP - EC 3.4.11.3) - which progressively increases during the course of normal pregnancy [26, 27], is very useful in the evaluation of the placental insufficiency [28–30]. Increased CAP activity has been found in the serum of patients with serious liver diseases and in seminal plasma [31]. CAP activity has been detected in the brain synaptic membranes [32] and in a haemolysate of human erythrocytes [33]. Recently, it has been suggested that placental oxytocinase may have an intriguing possibility in a variety of events not restricted to the regulation of pregnancy induced phenomena [34]. Several colorimetric assays using cystyl or cysteinyl p-naphthylamides or pnitroanilides have been developed for the determination of CAP activity in human serum. However, the utilization of these substrates is limited by their low solubility in water, and an affinity to oxytocinase (0.5 - 2.0- 10-3 mol 1-1) by 2-3 orders lower than the affinity of the natural substrate - oxytocin. The required amount of organic solvent for their solubilization influences adversely the course of the enzymic reaction. These drawbacks led to the application of a fluorogenic substrate H-Cys(Bzl)-NH-Mec [35] for ultramicrodetermination CAP in clinical practice. Since we had shown that in case of chymotrypsin substrates containing AMeq permit a more sensitive enzyme determination than the corresponding ones containing AMec, we prepared the fluorogenic substrate H-Cys(Bzl)-NH-Meq (17) and we used it for the determination of oxytocinase isolated from human retroplacental serum [36]. The enzymatic release of Ameq was followed at XeX = 340 nm and Xem = 430 nm, where its relative intensity is 100-fold greater than that of an equimolar solution of the substrate. Michael is constants (Km) of CAP for H-Cys(Bzl)-NH-Meq were determined at various pH values. The Km value at optimum pH (7.0-7.05) was 2.3 umol 1-1. The affinity of this substrate to oxytocinase was by two orders higher than that of chromogenic ones and greater than the affinity of the aminocoumarin substrate, as was evident from the comparison of the Km values: H-Cys(Bzl)-NH-N p , Km = 360 umol 1-1
88
c. Tzougraki / New Methods for Fluorometric Assays of Enzymes
[37], H-Cys(Me)-NH-Np, Km = 440 umol 1-1, H-Cys(Bzl)-NH-Mec, Km = 5.4 umol 1-1 [35]. The measurement of the relative activity of serum oxytocinase with several concentrations of DMSO at various pH values, showed that already 5% of DMSO in the reaction mixture caused an approximately 50% decrease in activity. However, due to the high affinity of the substrate 17, in the range of its concentrations used for the kinetic studies, the concentration of DMSO (<0.4%) does not influence adversely the course of the enzyme reaction, as in the case of chromogenic substrates, where the concentrations of the organic solvent exceeded 3%. Inhibition of Human Serum Oxytocinase by GnRH Peptides: The inhibition of human serum oxytocinase by GnRH peptides was studied [38] by using the substrate HCys(Bzl)-NH-Meq (17). The hydrolysis of 17 was inhibited by all peptides containing the pyroglutamic acid. Graphical analysis of kinetic data (Lineweaver-Burk, EadieHofstee, Dixon and Hanes plots) revealed a non-competitive type of inhibition and TLC proved that the peptides were not degraded by the enzyme. The inhibition constants determined according to Dixon are listed in Table 3, indicating that the chicken GnRH II was the most potent peptide in inhibiting "oxytokinase" activity. The observed differences also showed a remarkable fact, that mammalian GnRH was the least potent among the natural GnRH peptides under study as far as the inhibition of human serum oxytocinase is concerned. Both the pyroglutamyl peptides not belonging to the family of GnRH peptides were able to inhibit oxytocinase activity, neurotensin being several times more efficient than TRH. Table 3. Inhibitory potencies of GnRH peptides
Peptides1
'
Human GnRH pGlu-His-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH2 Chicken GnRH I pGlu-His-Trp-Ser-Tyr-Gly-Leu-Gln-Pro-Gly-NH2 Chicken GnRH D pGlu-His-Trp-Ser-His-Gly-Trp-Tyr-Pro-Gly-NH2 Salmon GnRH pGlu-His-Trp-Ser-Tyr-Gly-Trp-Leu-Pro-Gly-NH2 [D-Ala6, ProNHEt9]human GnRH [D-Ile6, ProNHMe9]human GnRH p-Glu-His-Trp-OMe p-Glu-His-Pro-NH2 (TRF) Neurotensin pGlu-Leu-Tyr-Glu-Asn-Lys-Pro-Arg-Arg-Pro-Tyr-Ile-Leu-OH
Ki, lO^mol- 1 i 60 40 6 12 40 40 65 180 25
Oxytocine is hydrolyzed by "serum oxytocinase" with Km of 0.2-0.1 x 10 mol 1
Studies of secondary specificity of prolyl oligopeptidase: Prolyl oligopeptidase, a serine protease (EC 3.4.21.26, POP), preferentially hydrolyzes proline-containing oligopeptides at the carboxyl side of proline residues. POP is widely distributed in animals, plants, mushrooms, but in few species of bacteria [39–42]. The enzyme, isolated from various tissues, is active in vitro against a range of natural peptides [40]
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
89
and may play an important role in maturation and inactivation of peptide hormones and neuropeptides. Its involvement in amnesia, depression and Alzheimer's disease has been suggested [43] and some inhibitors have been examined for treatment of effects related to these diseases [44]. Its active center contains five subsites for binding with substrate and inhibitor, extending from S'2 to S3. However, the physiological role of the enzyme remains unclear, and its reaction mechanism needs further investigation. Assays of POP have been performed utilizing mostly chromogenic and few fluorogenic or intramolecularly quenched fluorogenic substrates [41, 45, 46]. The majority of these substrates contain glycine at P2 position, with little contribution to the secondary interaction with the enzyme. Because of their low affinity, they had to be employed in such concentrations that the organic solvent used for solubilization adversely affected the enzyme activity. In order to study the 82 binding site of POP and to obtain substrates of higher affinity to the enzyme, we synthesized a number of fluorogenic substrates, N-acyl derivatives of AMeq, listed in Table 4 [47]. The substrates Z-X-Pro-NH-Meq contain at P2 position amino acid residues bearing acidic, basic, hydrophilic and bulky hydrophobic side chains. The residues Cys, Lys, His and Ser are preceding Pro in the sequence of several natural active peptides. The tripeptide Z-Asn-Cys(Bzl)-Pro-NH-Meq with Asn, which is preceding Cys residue in the sequence of vasopressin and oxytocin, was also synthesized in order to examine the effect of the elongation of the sequence to P3 position. The kinetic parameters for the hydrolysis of the substrates catalyzed by porcine kidney POP were determined at A«x = 360 nm and Xex = 430 nm and are listed in Table 4. Table 4. Kinetic parameters for the hydrolysis of the substrates by prolyl oligopeptidase
No 18 19 20 21 22 23 24 25 26 27
Substrate P4Pr
ZZZ-
Pr
MetLys(Boc)LyszHiszSerZLeuzGlu(OMe)ZGluZAsnCys(Bzl)ZCys(Bzl)'' VIM* in nmol of produced AMeq per
Pi-
PI'
Km (MM)
Vmax
Vma./K.n
(nmol miiV' mg"') a
(nmol min"1 mg"1 uM"1)
NH-Meq 5.2 6.1 ProNH-Meq 2.9 1.4 Pro2.1 4.4 ProNH-Meq 9.1 ProNH-Meq 3.5 NH-Meq Pro6.8 6.2 ProNH-Meq 5.5 5.3 ProNH-Meq 5.8 6.8 NH-Meq Pro5.0 1.3 8.7 ProNH-Meq 2.1 ProNH-Meq 1.4 2.1 min and per mg of enzyme preparation.
1.2 0.5 2.1 0.4 0.9 1.0 1.2 0.3 0.2 1.5
The comparison of the kinetic parameters shows that all the substrates showed a high affinity to the enzyme and can be used for its fluorometric determination. The obtained data suggest that the S2 binding subsite of prolyl oligopeptidase consists of a cavity with a shape and size that can accommodate bulky side chains with variable functional groups [Lys, Cys(Bzl), Glu(OMe), Leu], with a clear preference to a positively charged group. Fluorogenic Substrates for Aminopeptidase M, Elastase and Trypsin: Four substrates containing AMeq with the appropriate amino acids with enzyme specificity were synthesized and evaluated [25]. Compounds HC1- H-Ala-NH-Meq (28) and HC1- H-LeuNH-Meq (29) are substrates for aminopeptidase M, Boc-(Ala)4-NH-Meq (32) for elastase and Z-Arg-NH-Meq- H2CO3 (36) for trypsin.
90
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
Table 5. Kinetic parameters of several fluorogenic substrates for various proteases
Ref. Km kcaT KIB V™ 1 [mmol [mmor f umol prod. "\ dm'3] dmV] \ min- mgenz. } Aminopeptidase M 28 HCI H-AIa-NH-Meq 0.22 1.17" 0.251 0.18 29 HCI H-Leu-NH-Meq 0.16 30 HCI H-Leu-NH-Mec 112) 0.12 90.5 b 31 HBr H-Leu-NH-FMec [7a] C Elastase 13.40 0.22 32 Boc-(A!a)4-NH-Meq 3.40 [49] 0.50 33 Suc-(Ala)3-NH-Q e 2.08d 1.10 34 MeO-Suc-(Ala)2-Pro-Val-NH-Mec 0.81 [50] 15 0.50 35 Ac-(Ala)2-Pro-Ala-NH-Mec [10] 0.14 9.53 Trypsin 36 Z-Arg-NH-Meq 5.20 0.25 37 Z-Arg-NH-Mec [10] f 0.90 0.12 38 Z-Arg-NH-Mecs [9] a c Specific activity (s.a.) 1.2 units/mg of commercial enzymes (c.e.). - s.a. 15 units/mg. - s.a. 5.1 d e f units/mg of c.e. - Value referred to mg of active enzyme. - AQ = 6-aminoquinolinone. - AMecs = 7Enzyme
No
Substrate
aminocoumarin-4-methanesuIfonic acid.
The Km, kcat/Km or V^* values of the substrates 28, 29, 32 and 36 and of several other selected fluorogenic substrates are listed in Table 5. Because the purity of aminopeptidase M and elastase used in our assays was not known, Vmax values were reported for these enzymes. Comparison of the kinetic parameters showed that the substrates 32 and 36 have a higher affinity to the corresponding enzymes, elastase and trypsin, than the other listed substrates. Since the substrates of trypsin differ only in the fluorophores used, the higher affinity of substrate 36 is due to the fluorophore AMeq. In case of aminopeptidase M, the Km values of substrates 28 and 29 are comparable to those of 30 and 31. All substrates are suitable for the fluorometric determination of the corresponding enzymes.
Intramolecularly Quenched Fluorogenic Substrates Studies of the quencher - fluorophore combination: Synthetic peptide substrates containing a fluorophore at the bond-breaking position, as the above discussed, cannot be used either for the direct determination of enzymes hydrolysing specifically an internal peptide bond or for specificity studies of the PI', ..., Pn' positions, since the PI' position is occupied by the fluorophore. For this purpose intramolecularly quenched fluorogenic substrates (IQFS), i.e. a peptide chain bearing a fluorophore on the one end and a quencher on the other, are needed. Enzymatic cleavage of the peptide chain at any point between the interacting groups destroys the intramolecular quenching, thus resulting in an increase of fluorescence. IQFS have been used to determine various proteases such as collagenase, retroviral proteases, kallikreins and human cathepsins [50, 51]. A wide variety of fluorophores and quenchers have been proposed for IQFS, as recently reviewed [50, 52]. In a systematic investigation of the quencher-fluorophore combination we have synthesized and studied the fluorescence properties of twelve intramolecularly quenched fluorogenic substrates of the general form Q-Gly-Phe-F [53]. As fluorophores (H-F) we have chosen the widely used amines AMec (1), AMeq (3) and Ac (5) (see
91
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
Scheme 1), which in the previous studies showed excellent fluorescent properties. The free amine Ac does not fluoresce in aqueous solution, but its N-acyl derivatives do. As quenchers Q, the aromatic systems 2,4-dinitrophenyl (Dnp), 2,4,6-trinitrophenyl (Tnp), 2-nitrophenylsulfenyl (Nps) and 3,5-dinitrobenzoyl (DnBz) were used. Table 6. Fluorescence properties of substrates containing Amec, Ameq and Ac
39 42 43 44 45 40 46 47 48 49 41 50 51 52 53 * RFU:
Compound H-Phe-NH-Mec Dnp-Gly-Phe-NH-Mec Nps-Gly-Phe-NH-Mec Tnp-Gly-Phe-NH-Mec DnBz-Gly-Phe-NH-Mec H-Phe-NH-Meq Dnp-Gly-Phe-NH-Meq Nps-Gly-Phe-NH-Meq Tnp-Gly-Phe-NH-Meq DnBz-Gly-Phe-NH-Meq H-Phe-NH-c Dnp-Gly-Phe-NH-c Nps-Gly-Phe-NH-c Tnp-Gly-Phe-NH-c DnBz-Gly-Phe-NH-c Relative Fluorescence Units.
F(RFU)* 105.02 2.89 7.33 3.47 21.43 29.22 0.56 1.04 1.47 3.44 12.06 0.12 0.45 0.86 4.71
q.e. % 97.3 93.0 96.7 79.6 98.1 96.4 95.0 88.2 99.0 96.3 92.9 61.0
The comparison of the fluorescence intensities of substrates Q-Gly-Phe-F (42-53) with those of the compounds H-Phe-F (39-41) at the maximum wavelengths of the latter showed that an efficient quenching of fluorescence is caused by all groups Q (Table 6). The quenching efficiency is expressed by the quotient Fo-F/Fo, where Fo and F are the fluorescence intensities of the donor in the absence and presence of the acceptor (Q). Less efficient quenching is observed at the maximum wavelengths of the free amines HF. The high quenching efficiency of the Dnp, Tnp and Nps groups is probably a result of a nonradiative long-range resonance energy-transfer mechanism [54], since there is a spectrum overlap between the absorption bands of Q-Gly-OH and the emission spectra of H-Phe-F (39–41) as shown in Fig. 2. Combination of Dnp group with the fluorophore Ac gave the best result.
1.0:
It 00
(T
0.5
-50
Figure 2. Fluorescence spectra of (a) H-Phe-NH-c (41); (b) H-Phe-NH-Mec (39) and (c) H-Phe-NH-Meq (40). 10 umol dm" 3 solutions in 0.05 mol dm 3 Tris-HCl, pH 7.4, containing 10% DMSO. UV-Vis absorption spectra of (d) DnBz-Gly-OH; (e) Nps-Gly-OH; (f) Dnp-Gly-OH and (g) Tnp-Gly-OH. 50 umol dm"3 solutions in 0.05 mol dm Tris-HCl, pH 7.4, containing 10% DMSO.
92
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
Substrates of this type can be used as IQFS for endopeptidases (path A), which cleave the Gly-Phe bond or as usual fluorogenic substrates, if the bond Phe-F undergoes the hydrolysis (path B):
A
Path A
Q-Gly-Phe-F —
t B
Q-Gly-OH + H-Phe-F
PathB
Q-Gly-Phe-OH + H-F
Intramolecularly Quenched Fluorogenic p-Nitroanilides: Chromogenic substrates containing a p-nitrophenyl group are widely used for the colorimetric determination of proteases. In order to convert such substrates into IQFS, ten model p-nitroanilides of the general structure F-Phe-NH-Np were synthesized and their fluorescence properties were studied [51]. As fluorophores F-OH were used the coumarin and quinolinone derivatives shown in Scheme 2. The fluorescence properties of the fluorescent compounds 54–56, 63-68, as well as those of the intramolecularly quenched fluorogenic substrates F-Phe-NH-Np (69-78) together with the quenching efficiencies, q.e., measured at the maximum excitation and emission wavelengths of each corresponding fluorophore are summarized in Table 7. The fluorescence properties of each substrate F-Phe-NH-Np were compared with those of the corresponding F-OH or F-Phe-OH. In the case of substrates (74, 75, 77, 78) their properties have to be compared with those of compounds F-Phe-OH (64, 65, 66, 68). The fluorescence spectra of compounds 63, 66, 67 and those of their corresponding pnitroanilide substrates are illustrated in Figure 3.
CH
54
c-NH-Glt-OH
55 56
Mec-NH-Glt-OH X=O Meq-NH-Glt-OH X=NH CH,
OH
57
c-COOH
58 59
Mec-COOH X=O Meq-COOH X=NH
93
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes O
NHBoc
H,CO' 60 61 62
Hca X=OH Mca X=CH3O Aca X=NH 2
63
Boc-NH-mcp
Scheme 2: Structures of fluorophores F-OH. 1600
355 375 395 415 435 455 475 495 515 535 555
Wavelength (nm) Figure 3. Fluorescence spectra of Boc-NH-mcp (63), Glt-NH-mcp-Phe-NH-Np (73), Xex = 340 nm; HcaPhe-OH (66), Hca-Phe-NH-Np (76), ^x = 380 nm; Mca-Phe-OH (67), Mca-Phe-NH-Np (77), ^ = 334 nm; 1 uM solutions in 0.05 M Tris-HCl, pH 7.8, containing 1% DMSO. Table 7. Fluorescent properties of the substrates 69-78 and fluorophores 54–56, 63–68
54 69 55 70 71 56 72 63 73 64 74 65 75 66 76 67 77 68 78
Compound3 c-NH-Glt-OH c-NH-Glt-Phe-NH-Np Mec-NH-Glt-OH Mec-NH-Glt-Phe-NH-Np Mec-NH-Glt-Gly-Phe-Np Meq-NH-Glt-OH Meq-NH-Glt-Phe-NH-Np Boc-NH-mcp Glt-NH-mcp-Phe-NH-Np c-CO-Phe-OH c-CO-Phe-NH-Np Meq-CO-Phe-OH Meq-CO-Phe-NH-Np Hca-Phe-OH Hca-Phe-NH-Np Mca-Phe-OH Mca-Phe-NH-Np Aca-Phe-OH Aca-Phe-NH-Np
Xex/nm 325 325 331 331 331 336 336 340 340 327 327 338 338 380 380 334 334 364 364
Xem/nm 382 382 394 394 394 365 365 398 398 410 410 384 384 467 467 393 393 458 458
F" 115 8 1620 45 78 420 23 1038 56 4.6 0.5 32.5 0.2 1596 80 1207 15 1674 186
q.e.c% 93.0 97.2 95.2 94.5 94.6 89.1 99.4 95.0 98.8
88.9 1 uM solution in 0.05 M Tris-HCl pH 7.8, containing 1% DMSO. b. Arbitrary units, c. q.e.=(F0-F)/F0; F0 and F are the fluorescence intensities of the donor in the absence and presence of the acceptor. a
94
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
The comparison of the fluorescent intensities of the substrates 69–78 with those of the corresponding fluorophores at the maximum wavelengths of the latter showed that an efficient quenching of fluorescence is caused in all cases. With the exception of compound 66, the quenching effect may be attributed to a nonradiative long-range resonance energy-transfer mechanism, because there is a spectrum overlap between the absorption band of p-nitroanilide derivative and the emission spectra of the fluorescent parts [54, 55]. In the case of the compound 66, the quenching effect could be explained by a collisional quenching mechanism. Fluorophores 54–57, 59-63 can be used for the conversion of peptide p-nitroanilides into IQFS, which retaining their chromogenic properties may be applied in both fluorometric and colorimetric assays. Solid Phase Synthesis of Fluorogenic or Intramolecularly Quenched Fluorogenic Substrates: Although solid phase synthesis (SPS) has predominated over the synthesis in solution, its application in the synthesis of FS or IQFS is limited. A fluorescent marker appropriate for use in solid phase synthesis of such substrates, must bear two functional groups, one for attachment to the solid support and one for the development of the peptide chain. In order to develop a facile and fast method for direct preparation of FS or IQFS by SPS, we examined the bifunctional coumarin derivatives 7-hydroxy4-coumarinyl acetic acid (Hca) (60) and 7-amino-4-coumarinyl acetic acid (Aca) (62) as fluorescent linkers (Scheme 2). They were chosen because of their excellent fluorescence properties. Both have been successfully linked through their carboxyl group with 2-chloro-chlorotrityl-resin [56] or (R,S) 2-chloro-4'-carboxy triphenylmethanol resin [57] to produce 79a,b and 80a,b respectively. Acylation of 79a with oleic or palmitic acid to produce 81 and 82 derivatives and then cleavage from the resin afforded fluorescent compounds Ole-Oca (83) and Pal-Oca (84) (Scheme 3), suitable for the enzymatic study of lipases [58].
OOH
79a,b 80a,b 81 82
R = OH, a: Y= -, b: Y=-CONHCH2R = NH2, a: Y=-, b: Y=-CONHCH2R = OCO(CH2)7CH=CH(CH2)7CH3, Y= R = OCO(CH2)14CH3, Y= -
83 84
R = CO(CH2)7CH=CH(CH2)7CHj R = CO(CH2)MCH3
Scheme 3: Resin-bound coumarin derivatives (79-82) and Aca-derivatives (83,84).
For preparation of FS or IQFS for proteases, the amino group of 80a or 80b is acylated with the desirable C-terminal amino acid by the DIG method. The development of the rest of the peptide chain is carried out utilizing standard Fmoc-derived SPPS methodology. The FS are converted into IQFS, containing Dnp group as quencher, by
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
95
the direct dinitrophenylation of the C-terminal amino acid residue of the resin-bound peptide. The usefulness of this method was proved by the successful synthesis of the compounds 85-89 (starting from 80b), potential substrates for cathepsin D. The substrates 86-89 (Scheme 4) containing various amino acid residues at P$ position can be used for secondary specificity studies of cathepsin D. The compounds 90–94 (Scheme 4) were also synthesized and their spectral properties were compared. An efficient quenching (94%) of the fluorescence of the compound 93 was observed at the excitation and emission maxima (Xex = 334 nm and Xem = 391 nm) of compound 92 [59]. Dnp-Lys-Pro-Ile-Cys-Phe-Ile-Lys-Leu-Aca-OH(85) Dnp-X-Lys-Pro-Ile-Cys-Phe-Ile-Lys-Leu-Aca-OH X Val Ser Lys Glu IQFS 86 87 88 89
H-Lys-Pro-Ile-Cys-Phe-Ile-Lys-Leu-Aca-OH(90) H-Ile-Lys-Leu-Aca-OH (91) H-Leu-Aca-OH (92) Dnp-Leu-Aca-OH*(93) Dnp-Leu-OH (94) Scheme 4: Aca and Dnp derivatives synthesized by SPPS.
The method described offers the advantage of synthesizing any fluorogenic substrate or intramolecularly quenched one by using exclusively solid phase peptide synthesis. The initial conjugation of the fluorescent marker Aca on the resin permits easy arjd fast development of any fluorogenic peptide substrate. Furthermore, the direct introduction of the Dnp group to the peptide chain provides a convenient preparation of IQFS. References [1] [2] [3] [4] [5] [6] [7] [8]
[121 [13] [ 14|
A. J. Barrett (Ed.), Proteinases in Mammalian Cells and Tissues, Elsevier/North-Holland Biomedical Press, Amsterdam, 1977. R. Lottenberg, U. Christensen, C.M. Jackson and P.L. Coleman, Methods Enzymol. 80 (1981) 341. L. Fareed, H.L. Messmore, J.M. Walenga and E.W. Bermes Jr., CRC Crit. Rev. din. Lab. Sci. 19(1983)71. A.A. Gershkovich and V.K. Kibirev, Bioorg. Khim. 14 (1988) 1461. G. Sarath, R.S. de la Motte, F.W. Wagner in: R.J. Beynon and J.C. Bond (Eds.), Proteolytic Enzymes, a Practical Approach, IRL Press, Oxford, 1989, Chapter 3. M. Zimmerman, E.G. Yurewicz and G. Patel, Anal. Biochem. 70 (1976) 258. (a) R. Smith, E. Bissell, A. Mitchell and K. Pearson, Thrombosis Res. 17 (1980) 393. (b) E. Bissell, A. Mitchell and R. Smith, J. Org. Chem. 45 (1980) 2283. E. Bissell, A. Mitchell, K. Pearson and R. Smith, U.S. US 4, 388, 233 (1983) [Chem. Abstr. 99 (1983)84390g]. E. Sato, A. Matsuhisa, M. Sakashita and Y. Kanaoka, Chem. Pharm. Bull. 36 (1988) 3496. M. Zimmerman, B. Ashe, E. Yurewicz and G. Patel, Anal. Biochem. 78 (1977) 47. Torri and Co. Ltd. Jpn Kokai Tokkyo Koho JP a. 82, 75, 956 (1982); b. 82, 75, 957 (1982); c. 82, 75, 958 (1982) [Chem. Abstr. 97 (1982) 163507n, 163508p, 163509q]. Y. Kanaoka, T. Takahashi and H. Nakayama, Chem. Pharm. Bull. 25 (1977) 362. T. Morita, H. Kato, S. Iwanaga, K. Takada, T. Kimura and S. Sakakibara, J. Biochem. 82 (1977) 1495. S. Sakakibara, Jpn Kokai Tokkyo Koho 79, 03, 074 (1979) [Chem. Abstr. 91 (1979) 21130m].
96
[15] [16] [17]
C. Tzougraki / New Methods for Fluorometric Assays of Enzymes
K. Murakami, T. Ohsawa, S. Hirose, K. Takada and S. Sakakibara, Anal. Biochem. 110 (1981) 232. G. Smith, J. Ding and T. Peters, Anal. Biochem. 100 (1979) 136. Y. Kanaoka, T. Takahashi, H. Nakayama, T. Uevo and T. Sekine, Chem. Pliarm. Bull. 30 (1982) 1485.
[18] [19] [20] [21] [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] [55] [56] [57] [58] [59]
G. Kokotos and C. Tzougraki, J. Heterocyclic Chem. 23 (1986) 87. G. Kokotos and C. Tzougraki, Int. J. Peptide and Protein Res. 28 (1986) 186. G. Kokotos, R. Geiger and C. Tzougraki, Biol. Chem. Hoppe-Seyler 371 (1990) 835. K. Woods and M. Fooladi, J. Chem. Eng. Data 13 (1968) 440. R. L. Atkins and D. E. Bliss, J. Org. Chem. 43 (1978) 1975. M.C. Geokas, C. Largman, J.W. Brodrick, J. H. Johnson and M. Fassett, / Biol. Chem. 254 (1979)2775. N. Yoshida, M. T. Everitt, H. Neurath, R.G. Woodbury and T. C. Powers, Biochemistry 19 (1980)5799. C. Tzougraki, C. Noula, R. Geiger and G. Kokotos, Uebigs Ann. Chem. (1994) 365. S. Lampelo, T. Vanha-Perttula, J.Reprod. Fertil. 58 (1980) 225. N. Majkic-Singh, A. Vukovic, S. Spacic, A. Ruzic, M. Stojanov, I. Berkes, Clin. Biochem. 15 (1982) 152. L. Chapman, R. Burrows-Prakin, U.P. Rege, E. Silk, Br. J. Obstet. Gynaecol. 83 (1976) 238. P.A. Hensleigh, S.G. Cheatum, W. N. Spellacy, Am. J. Obstet. Gynecol. 129 (1977) 675. G. Gopalaswamy, N. Balasubramaniam, A. S. Kanagasabapathy, Aust. N. Z. J. Obstet. Gynaecol. 23(1983)79. G. Tiderstrom, D. Heinegard, Clin. Chim. Acta 88 (1978) 293. J.P.H. Burbach, J.L.M. Lebouille, J. Biol. Chem. 258 (1983) 1487. L.P. Aleksenko, V.F. Podznev, V.N. Orekhovich, Dokl. Akad. Nauk. SSSR 293 (1987) 728. S. Mizutani, Nagoya J. Med. Sci. 61 (1998) 85. M. Suzuki, T. Ueno, T. Takahashi, Y. Kanaoka, T. Okuyama, H. Furuya, T. Sekine, Clin. Chim. Acta 115 (1981) 223. H. P. MaSkova, G. Kokotos, C. Tzougraki and T. Earth, Collect. Czech. Chem. Commun. 54 (1989)2802. D. Watson, S. Gibbards, Clin. Biochem. 6 (1973) 60. H.P. MaSkova, J.P.H. Burbach, G. Kokotos, C. Tzougraki and T. Barth, Endocrine Regulations 26(1992)53. S. Wilk, Life Sci. 33 (1983) 2149, Mini-review. W.R. Welches, K.B. Brosnihan and C.M. Ferrario, Life Sci. 52 (1993) 1461, Mini-review. E. Szwajeer-dey, J. Rasmussen, M. Meldal and K. Breddam, J. Bacterial. 174 (1992) 2454. A. Kanatani, T. Yoshimoto, A. Kitazono, T. Kokubo and D. Tsuru, J. Biochem. 113 (1993) 790. M. Maes, F. Goossens, S. Scharpe, H.Y. Meltzer, P. d'Hondt and P. Cosyns, Biol. Psychiatry 35 (1994)545. K. Toide, M. Shinoda, A. Miyazaki, Rev. Neurosci. 9 (1998) 17. C. Tzougraki, G. Kokotos, H. P. Maskova, E. Anzenbacherova and T. Barth, Collect. Czech. Chem. Commun. 55 (1989) 1112 and references therein. J. Sommer, Biochim. Biophys. Acta 1173 (1993) 289. C. Noula, G. Kokotos, T. Barth and C. Tzougraki, J. Pept. Res. (1997) 46. P.J. Brynes, R. Andrade, D. Gordon, Anal. Biochem. 126 (1982) 447. M.J. Castilo, K. Nakajima, M. Zimmerman, J. Powers, Anal. Biochem. 99 (1979) 53. A. A. Gershkovich, V.V. Kholodovych, J. Biochem. Biophys. Methods 33 (1996) 135, Minireview. C. Charitos, C. Tzougraki and G. Kokotos, J. Pept. Res. 56 (2000) 373 and references therein. C.G. Knight, Methods Enzymol. 248 (1995), 18. G. Kokotos and C. Tzougraki, J. Chem. Soc. Perkin Trans. 2 (1991) 495. A. Yaron, A. Carmel and E. Katchalski-Katzir, Anal. Biochem. 95 (1979) 228. A. A. Gershkovich, Ukr. Biochem. J. 66 (1994) 10. K. Barlos, D. Gatos, J. Kallitsis, G. Papaphotiou, P. Sotiriou, W. Yao and W. Schafer, Tetrahedron Lett. 30 (1989) 3943. C.C Zikos and N.G. Ferderigos, Tetrahedron Lett. 35 (1994) 1767. C. Charitos, G. Kokotos and C. Tzougraki. J. Heterocyclic Chem. 38 (2001) 153. K. Paschalidou and C. Tzougraki. Lett. Pept. Sci. 1 (2001) 249.
97
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakas (Eds.) IOS Press, 2002
Enhancement of the Cytogenetic and Cytostatic Activity of Chlorambucil After its Esteric Connection with A Hecogenin Lactam V. Karayianni1, Ch. Camoutsis2, V. Soulakis2, D. Mourelatos1, E. Mioglou1, Z. lakovidou1 1
Laboratory of General Biology and Genetics, Aristotle University of Thessaloniki 2 Laboratory of Pharmaceutical Chemistry, University of Patras
Abstract. The reasons for the synthesis of the compounds studied in the present work are: a) The already known antineoplastic activity of Hecogenin in HeLa cells and b) the knowledge that the amide (-NHCO-) group in modified steroidal derivatives of Chlorambucil enhances the antineoplastic effects of the alkylating part of the molecule. SCEs (System Chromatids Exchange) have been employed as a sensitive indicator of unrepaired DNA damage after the damaging effects of the chemicals studied, while human lymphocyte Proliferation Rate Indices (PRIs) were used as a criterion of cytostatic activity. Our comprative study included the compounds: Hecogenin acetate (Hec) (1), Lactam of Hecogenin acetate (HecLac) (2), the ester of Hecogenin lactam with Chlorambucil (3), the ester of Hecogenin with Chlorambucil (4), and Chlorambucil (CBC) (5). The comparisons were done on a molar basis and at CBC concentrations which are considered to be very low (hypoclinical). The order of magnitude of cytogenetic activity of the compounds is: 3»4~2>5>1. In conclusion, and in accordance with the study's rational, we observed that the ester of CBC with HecLac appears with superior synergistic cytogenetic damage in comparison with CBC and HecLac acting independently.
98
V. Karayianni et al. / Enhancement of the Cytogenetic and Cytostatic Activity of Chlorambucil
Introduction Chronic lymphocytic leukemia appears more frequently from all other malignant hematological diseases in the Western countries, and occurs in the 30-40% of the patients that suffer from leukemias [1]. On the other hand, CBC is considered to be a potent chemotherapeutic for combating chronic lymphocytic leukemia and is nowadays used in clinical practice [1,2, 3]. CBC was submitted along with mitoxantrone and prednisone to a 62-year-old patient with a monoclonal mucosa-associated lymphoid tissue lymphoma with simultaneous manifestation in the colonial and bronchial mucosa and a complete year-long remission was achieved [4]. Moreover, it is used as a chemotherapeutic against nonHodgkin's lymphoma [2]. Hec, on the other hand, is a natural product of the hydrolysis of steroidal saponins, which are known for their antitumour effect. Steroidal saponins have shown cytotoxic action against cancer K562 cells in vitro [5] and in human squamous cell carcinoma cells (HSC-2) [6]. Moreover, they inhibited the in vitro growth of several human cancer cell lines, osteosarcoma (OST), pancreatic cancer (PANC-1), human gastric cancer (HGC-27), and pheochromocytoma (PC-12) cell cultures [7]. Mimaki et. al. also suggested that steroidal saponins showed cytostatic activity against human leukemia (HL-60) cells [8]. Sata et. al. refer the anticancer effect of steroidal saponins in P388 cells in vitro at 2.1 mg/ml [9]. Indeed, it was found that Hec inhibits the growth of cancer HeLa cell cultures [10]. Moreover, there are more reasons why Hec was selected for the study. Hec can produce cortisone [11, 12]. Also, the steroidal nucleus of Hec can be modified by the insertion of a N- molecule to the C ring. Previous studies have shown that the anticancer action of an alkylating agent like CBC is greatly enhanced when it is carried by a steroid molecule. In this study the SCEs and the PRI of the lymphocyte cultures of a healthy patient were calculated in order to compare the genotoxic and cytostatic effect of compounds 1-5 (Fig. 1).
H3CCOO
Hecogenin' acetate (3p-acetoxy-5a, 22a-spirostan-12-one) (1)
V, Karayianni et al. /Enhancement of the Cytogenetic and Cytostatic Activity of Chlorambucil
99
H3CCOO
Lactam of Hecogenin acetate (3(3-acetoxy-12a-aza-C-homo-5a, 22a-spirostan-12-one) (2)
•CHjCHjCH OH2CH2CN
dH2CHjC
Ester of Hecogenin lactam with Chlorambucil 4-[p-[bis(2-chloroethyl)amino]phenyl] butyric ester of 3p-hydroxy-12a-aza-C-homo-5a, 22a-spirostan-12-one (3)
CIH2CH2C.,
CIH2CH2C
Ester of Hecogenin with Chlorambucil 4-[p-[bis(2-chloroethyl)amino]phenyl] butyric ester of 3p-hydroxy-5a, 22a-spirostan-12-one (4) CH2CH2CH2COOH CIH2CH2C
CIH2CH2C
Chlorambucil 4-[p-[bis(2-chloroethyl)amino]phenyl] butyric acid (5)
Figure 1. Synthetic compounds used in the study.
100
V. Karayianni et al. /Enhancement of the Cytogenetic and Cytostatic Activity of Chlorambucil
Synthesis of the compounds.The synthesis of the 4-[p-[bis(2-chloroethyl) amino] phenyl] butyric esters of 3p-acetoxy-12a-aza-C-homo-5a, 22a-spirostan-12-one [13] and 3(ihydroxy-12a-aza-C-homo-5a, 22a-spirostan-12-one [14] was achieved by the mixed anhydride method. The mixed anhydride was prepared using 2,4,6-trichlorobenzoyl chloride as the condensing agent and triethylamine as an auxiliary base. The alcoholysis step was carried out in benzene and in the presence of 4-dimethylaminopyridine. The esters were isolated in good yields, whereas the corresponding 2,4,6-trichlorobenzoic acid ester, was isolated as by-product (Fig. 2).
X=-CONHor-C=O
Figure 2. Synthetic procedure of 4-[p-[bis(2-chloroethyl)amino]phenyl]butyric esters of 3p-acetoxy-12a-azaC-homo-5a, 22a-spirostan-12-one and 3p-hydroxy-12a-aza-C-homo-5a, 22a-spirostan-12-one.
Results The results of the SCE and the PRI measurements of compounds 1-5 in human lymphocyte cultures of a healthy donor are shown in Table 1. It is obvious that the ester of HecLac with CBC was by far the most effective of all compounds as it caused a synergistic increase on the SCE rate and decrease of the PRI compared to the control. Compounds 1-5 did not show synergistic genotoxic and cytostatic effect. More precisely, the ester of HecLac with CBC caused a significant increase on the SCE rate (P<0.02, t test) compared with HecLac, and compared with CBC (P<0.01, t test).
V. Karayianni et al. / Enhancement of the Cytogenetic and Cytostatic Activity of Chlorambucil
101
Moreover, the ester of HecLac with CBC caused a significant decrease of the PRI compared to HecLac (P<0.01, x2 test), and to CBC (P<0.001, x2 test). The results obtained by the use of Hec alone were very close to the SCE and PRI values of the control. Table 1. The SCEs and the PRI of compounds 1-5 in human lymphocyte culture PRI2 SCEs ± SEM1 Compound Concentration (uM) 2.57 13.10± 1.01 Control 0.5 2.52 1 14.48 ± 1.28 0.5 17.99 ± 1.84 2.37 2 0.5 25.06 ±2.18* (21.43) a 1.95** (2.42) b 3 0.5 4 2.63 0.5 18.70 ± 1.20 2.62 0.5 16.54 + 0.68 5 SCEs ± SEM= Sister Chromatids Exchange ± Standard Error of the Mean 2 PRI = M1+M2+M3+/ M1+M2+M3, where M1 is the number of cells in the first mitotic division, M2 in the second mitotic division, KCU M3+ in the third and other mitotic divisions. * Significant increase (P<0.02, t test) compared with HecLac, and (P<0.01, t test) compared with CBC. ** Significant decrease (P<0.01, x2 test) compared with HecLac, and (P<0.001, x2 test) compared with CBC. a Expected value if the increase above background for HecLac and CBC was independent and additive. b Expected value if the depression below background for HecLac and CBC was independent and additive.
Discussion The ester HecLac with CBC presented a remarkable synergism as a genotoxic and cytostatic agent in comparison with CBC and HecLac acting independently. CBC is a bifunctional alkylating agent with remarkable antitumour effect. It is believed that it causes interstrand DNA cross-links that stop DNA replication and it is considered to be highly mutagenic [16]. The study of Mimaki et. al. [10] showed the in vitro antitumour-promoter effect of three different types of Hec. They inhibited the incorporation of 32P into phospholipids of HeLa tumour cells at 5 jig/ml. It is possible that the presence of the lactam in the steroidal nucleus of Hec enhances the cytostatic and genotoxic effect of the ester. Previous studies on experimental animal mammary tumour and leukemia systems have shown a superior cytostatic action of the steroidal molecule after the insertion of the -NH-CO- group compared to the unmodified steroidal nucleus [17]. The -NH-CO- group probably becomes active and helps the ester binding to DNA and other biomolecules such as proteins. The lactam is transformed by a metabolic process or at least by an enzymatically catalyzed reaction to the active species: NHCO—> -N-H -C=O+. These species could attack the constituents of rodent and human cancer cells, acting synergistically with the alkylating congener [18]. This comparative study was made at CBC concentrations which are considered to be very low (hypoclinical). This is important because high concentrations of CBC can cause side effects, like myelosuppression, particularly leukopenia [19], or even hepatitis [20]. Moreover, low dosage could reduce CBC drug resistance [21]. Cancer and normal cells have the ability to excise and repair various types of damage to DNA since both cell types have similar DNA repair mechanisms. Therefore human lymphocyte cultures from normal subjects are a proper experimental material for studying potential chemotherapeutics [22]. A correlation between potency for SCE induction, antitumour activity and effectiveness in PRI depression by potential chemotherapeutics has been recently identified in vitro [23] and in vivo [24]. In the present study our results show that HecLac with CBC is an effective inducer of SCEs and of cell division delays, therefore further studies in different biological systems are warranted for establishing possible antitumour activity.
102
V. Karayianni et al. / Enhancement of the Cytogenetic and Cytostatic Activity of Chlorambucil
References [ 1 ] Losonczy, H., Orv-Hetil 139 (1998) 611 [2] Peters, F.P., Lalisang, R.I., Fickers, M.M., et. al., Ann Hematol 80 (2001))155– [3] Hovinen, J., Silvennoinen, R., Vilpo, J., Chem-Res-Toxicol 11 (1998) 92 [4] Sallach, S., Schmidt, T., Pehl, C., et. al.. Colon Rectim 44 (2001) 448– [5] Hu, K., Dong, A., Yao, X., Kobayashi, H., Iwasaki, S., Med 62 (1996) 573 [6] Mimaki, Y., Watanabe, K., Ando, Y., Sakuma, C., Sashida, Y., Furuya, S., et. al. J Nat Prod 64 (2001) 17 [7] Mimaki, Y., Sashida, Y., Kuroda, M., et. al., Biol Pharm Bull 18 (1995) 467 [8] Mimaki, Y., Kuroda, M., Ide, A., et. al., Phytochemistry 50 (1999) 805 [9] Sata, N., Matsunaga, S., Fusetani, N., et. al. Biosci Biotechnol Biochem 62 (1998) 1904 [10] Mimaki, Y., Kanmoto, T., Sashida, Y., et. al. Phytochemistry 41 (1996) 1405 [11] Cameron, A.F.B., Evans, R.M., Hamlet, J.S., et. al., J Chem Soc (1955) 2807 [12] Chapman, J.H., Elks, J., Wyman, L.J., Chemistry & Industry (1955) 603 [13] Mazur, R.H., J Am Chem Soc 81 (1959)1454 [14] Wall, M.E., Walens, H.A. J Am Chem Soc 80 (1958) 1984 [15] Goto, K., Maeda, S., Kano, Y., et. al., Chromosoma 66 (1978) 351 [16] Yaghi, B.M., Turner, P.M., Denny, W.A., et. al., Mut Res 401 (1998) 153 [17] Wall, M.E., Abernethy, G.S.Jr., Caroll, F.J., and Taylor, D.J., J Med Chem 12 (1969) 810 [18]Catsoulacos, P., Camoutsis, C., Papageorgiou, A., et. al., Anticancer Res 12 (1992) 1617 [19]Inoue, K., Ogawa, M., Horikoshi, N., et. al., Gan To Kagaku Ryoho 14 (1987) 2672 [20]Pichon, N., Debette-Gratien, M., Cessot, F., et. al., Gastroenerol Clin Biol 25 (2001) 202 [21] Sanderson, B.J., Johnson, K.J., Henner, W.D., Mutagenesis 16 (2001) 197 [22] Mourelatos, D., Cancer J 9 (1996) 136 [23] Papageorgiou, A., Tsavdaridis, D., Geromichalos, G.D., et. al., Cancer Detec Prev 25 (2001) 360 [24]Mylonaki, E., Mourelatos, D., Kotsis, A., Chemotherapy 44 (1998)121
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
103
In Searching for New Anti-Influenza A Drugs: Heterocyclic and Carbocyclic Aminoadamantanes Bearing a 2-Adamantyl Group With Potent Activity Against Influenza A Virus N. Kolocouris,1 A. Kolocouris,1 G. B. Foscolos,1 G. Fytas,1 E. Padalko,2 J. Neyts2 and E. De Clercq2 1
School of Pharmacy, Department of Phrmaceutical Chemistry, University of Athens, Panepistimioupoli-Zografou GR–15771 Athens, Greece. 2 Rega Institute for Medical Research, Katholieke Universiteit Leuven, B–3000 Leuven, Belgium. Abstract. The influenza A virus has been proved to be a serious threat for human life. Three pandemics have occurred in the last century, one of which the infamous "Spanish flu" of 1918 was responsible for more than 20 million deaths worldwide. The present drugs amantadine and rimantadine which are M2 virus ion channel blockers have both prophylactic and therapeutic activity. Since the danger of a future pandemic due to a new influenza A virus is real there is a need for more deep understanding of the SAR for aminoadamantane series. Heterocyclic and carbocyclic aminoadamantanes with a novel structure, i.e. with an amino group attached at C–2 position of the adamantane nucleus, were found to be potent antiinfluenza A agents in vitro. The synthetic pathways followed for their preparation are described. The lowest energy conformations of the most active compounds are depicted. The superposition of these conformers reveals their similarities and differences and can lead to the design of more active agents. The novelty of the new structures is discussed: molecules with an amino group and carbon substituent at 2adamantane position appear to be promising anti-influenza A agents. For the most active compounds amine nitrogen atoms lie in a distance of 1.5 - 2.5 A away from 2-adamantyl carbon. One of them exhibited even 230-fold higher activity than amantadine and another had a selectivity index (SI) that was more than 1045-fold higher. The intense effort which led to the described results will trigger future research and possibly the discovery of new anti-influenza A drugs.
104
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
Introduction Influenza A is a major respiratory tract disease affecting millions of people each year. Influenza A is characterized by the abrupt onset of constitutional and respiratory signs and symptoms (e.g., fever, myalgia, headache, severe malaise, nonproductive cough, sore throat and rhinitis) [1]. However, in some persons the infection can cause pulmonary or cardiac disease or lead to secondary bacterial pneumonia or primary viral pmeumonia. Epidemics of influenza occur during the winter months nearly every year and are responsible for an average of approximately 20,000 deaths per year in the United States [2,3]. Influenza A viruses have the ability to undergo changes by the mechanisms of antigenic drift and shift and new evolving strains can be a serious threat to the human population [4]. Thus, pandemic influenza A viruses appeared in 1918 ("Spanish" H1N1), 1957 ("Asian" H2N2) and 1968 ("Hong Kong" H3N2). Given that influenza shifts occur every 20-30 years and that a new lethal variant appeared in Hong Kong in 1997, the danger of future influenza A pandemics is real. The M2 virus ion channel blockers amantadine 1 and rimantadine 2 are antiinfluenza A drugs that inhibit virus replication at micromolar concentrations [5]. They inhibit influenza A virus replication by blocking the ion channel of the small virus membrane protein M2. Studies of the interaction of amantadine with the membrane spanning domain of M2 protein based on neutron diffraction, solid state NMR and dynamics simulations have revealed that the drug interacts with the pore of the channel. It has been proposed that possibly the ammonium group of amantadine forms a hydrogen bond with the His–37 imidazole nitrogen lone pair while adamantyl group fits in a lipophilic pocket formed in the vicinity of Gly–34 (Figure 1) [6–8].
Figure 1. Stuctural model of the M2 ion channel without (A) and with (B) the specific inhibitor, amantadine. The central pore reaches its widest diameter (-4 A) near the center of the bilayer, just above His–37.8
Amantadine 1 and rimantadine 2 are indicated for the prophylaxis and treatment of influenza A infection. Amantadine or rimantadine can cause CNS and gastrointestinal side effects and there is a need for more active aminoadamantanes with fewer side effects. By our best knowledge after the work of K. Lundahl et al. in 1972 [9] only few papers have appeared in the literature on antiviral aminoadamantanes or related compounds [10, 11]. Many heterocyclic and carbocyclic aminoadamantanes active against influenza virus A were synthesized in our laboratory during the last five years [12–19]. These compounds bear an amino group substitution at C–l and the most potent at C–2 positions of the adamantane nucleus. This finding is novel and in this review we will present the synthesis, biological results and a SAR study based on the conformational properties of compounds in series 3-8. Compounds in these series are relatively rigid and this is beneficial for superposition studies.
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
105
3a, 4a, 5a: R=H 3b, 4b, 5b: R=CH3 3c, 4c, 5c: R=C2H5
(CH2)n-N \R'
n = 0, 1
6a (n=0), 6d (n=1 : R=R'=H 6b (n=0), 6e (n=1 : R=H R=CH3 6c (n=0), 6f (n=1 R=R'=CH3
7a: R=H 7b: R=CH3 7c: R=C2H5
8a: R=H 8b: R=CH3 8c: R=C2H5 8d: R=n-C3H7
Results - Discussion Chemistry: The rigidity and crowding around 2-position of adamantane nucleus often makes chemical transformations rather difficult and the synthesis of rigid adamantane derivatives a challenging aim. The synthetic pathways used for the synthesis of the parent compounds in each series will be described below. 2-Nitroadamantane was the starting material for the synthesis of spiropyrrolidines 3 (Scheme 1) [13, 14]. This compound can be easily deprotonated to its carbanion, which undergoes 1,4-addition reactions with conjugated esters. Michael condensation of 2nitroadamantane 10 with ethyl acrylate in the presence of Triton B afforded ethyl 2-nitro-2adamantanepropanoate 11. Hydrogenation of the nitro ester 11 over Raney nickel catalyst provided, with concominant y-lactam formation, the spiro[pyrrolidine-2,2'-adamantan]-5one 12. Subsequent reduction of y-lactam 12 with LiAlH4 gave the parent spiropyrrolidine 3a. For the synthesis of the spiro[piperidine-2,2'-adamanantanes] 4 selective reduction of the methylester 14 with NaBH4 in a dioxane-water mixture gave the 2-nitro-2adamantanepropanol 15 [13, 15] (Scheme 2). Attempts at the direct conversion of alcohol 15 to nitrile 17 via known procedures which have been shown to proceed by activation of the alcohol function as the trimethylsilyl derivative or via the corresponding trifluoroacetate failed to succeed. Thus, the above nitroalcohol 15 was converted to its chloride 16 upon treatment with SOCl2 at 90 C. The nitrile 17 was obtained from the reaction of 16 with KCN in acetonitrile in the presence of 18-crown-6. Methanolysis of the nitrile 17 and hydrogenation of the intermediate nitroester 18 over Raney nickel catalyst afforded the
106
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
spiro[piperidine-2,2'-adamantane]-6-one 19 after 5-lactamisation in boiling ethanol. Reduction of the S-lactam 19 with LiAlH4 in DME led to the parent spiro[piperidine-2,2'adamantane] 4a. Scheme 1
Scheme 2
TritonB, t-BuOH
'
NaBH4|
14
10
CH2CH2CH2CI g
SOCl2
dioxane. h 25 oc
toluene 90 °C 15
.CH2CH2CH2CN 1)MeOH. gas HCI . reflux
KCN, 18-crown-6 acetonitrile, reflux
16
4b,c
= CH3
R = CH2CH3
The synthesis of the spiro[morpholine-3,2'-adamantanes] 5 is illustrated in Scheme 3 [13, 15]. Hydroxymethylation of the 2-nitroadamantane 10 afforded the 2-nitro-2-
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
107
adamantane-methanol 21. Catalytic reduction of the nitroalcohol 21 resulted in the 2amino-2-adamantamethanol 22, which was treated with bromoacetyl chloride to give the N-bromoacetamide 23. The bromoacetamide 23 was then cyclized in alkaline medium to afford the spiro[morpholine-3,2'-adamantane]-5-one 24, which was converted to the parent morpholine 5a by means of LiAlH4 reduction. Scheme 3 NO2
. Raney_
CH2OH
CH2OH
EtOH
22
•°1 *N
^s..
| H
^n
LiAlbJ4, THF
R'COCI, Et3N
reflux
ether
—
!*•
5a
25a,b: R1 = OEt R ' = CH3
5b,c: R = CH3 R = CH2CH3
The synthesis of the spiro[cyclopropane-l,2'-adamantan]-2-amines 6a-c and methanamines 6d-f was accomplished as depicted in Scheme 4 [14]. The [2+l]cycloaddition reaction of ethyl diazoacetate with 2-methyleneadamantane 26 in the presence of copper-bronze gave the ethyl spiro[cyclopropane-l,2'-adamantane]-2carboxylate, which was saponificated to the corresponding carboxylic acid 27. The action of ethyl chloroformate on the carboxylic acid 27, in the presence of triethylamine, afforded the corresponding mixed anhydride, which was then transformed to azid and isocyanate 28. Isocyanate 28 was then suitably transformed to amines 6a-f For the preparation of the compounds 7a-d the catalytic hydrogenation of the hydrochloride 30 over PtO2 catalyst led to the aminoalcohol 31 (Scheme 5) [17]. Reaction of alcohol 31 with SOCl2 gave the corresponding chloride 32 as hydrochloride. Elimination reaction of chloride 32 in refluxing KOH/EtOH solution produced 2piperideine 34 possibly via the unstable enamine 33. Imine 34 was treated with sodium borohydride to give the parent 2-(2-adamantyl)piperidine 7a.
108
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
Scheme 4 1) N2CHCO2Et, Cu heptane. A 2) NaOH
.OH CH3Mgl
KHSO 4
100 °C 170 mmHg
ether - THF
26
LiAIH4 CO2H
2) HNR1R2
29
27
6d-f
R1 = R2 = H R1 = H, R2 = CH3 R1 = R2 = CH3
1) CICO2Et/Et3N 2) NaN3 3) Toluene/A
NaCNBH3
LiAIH4 THF reflux
6b
28
6c
1) HCl-H2O 2) OH-
H NH2
6a
Scheme 5
•i.
JLf [^OHN cr
*2*
EtOH
KOH/EtOH, 0
33
CH2R 7b,c,d R=H, CH3, CH2CH3
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
N-Alkyl 2-pyrrolidinone 37 was prepared through N-alkylation of 2-pyrrolidinone 36 and used as starting materials for the preparation of the target compounds 8a-h (Scheme 6) [18]. Alcohol 38 was synthesized by the reaction of the suitable 1-substituted 2-oxo-3-'pyrrolidinyl lithium with 2-adamantanone. Subsequent dehydration and catalytic hydrogenation of the indermediate methylene lactam 39 afforded the 3-(2-adamantyl)-2pyrrolidinone 40. Reduction of lactam 40 with LiAlH4 yielded compounds 8b-h. Scheme 6
NaH, RX
H
3
DMF or benzene 70 °C
1.LDA, adamantanone, -70 °C » 2. H+/H2O
benzene reflux
38
36
N—R
N—R
H2, PtO2
N—R
LiAIH4
EtOH
39
TsOH OH
THF, reflux
40
8b-g
8b: R=CH3 8c: R=C2H5 8d: R=n-C3H7 Be: R=n-C4H9
The N-H pyrrolidine 8a was synthesized using the N-trimethylsilyl 2-pyrrolidinone 41 as starting material (Scheme 7). Lithiation of lactam 41 and subsequent reaction with 2adamantanone afforded alcohol 42. Compound 42 was treated with thionylchloride and potassium methoxide solution to give methylenelactam 44. Catalytic hydrogenation and subsequent LiAlH4 reduction resulted in the pyrrolidine 8a. The above described routes can be considered as general for the 3-substitution of y-lactams and pyrrolidines with cycloalkyl groups.
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
110
Scheme 7
1. LDA adamantanone, -70 °C
I
benzene, R.T.
2.
CHCl3, relux
H
41
36
42
N—H
44
N—H LiAlH4, THF
m reflux
45
8a
Antiviral activity evaluation - Structure activity relationship study: The activity (MIC50) and cytotoxicity (MCC50) of the aminoadamantanes 3–8 were examined against influenza A virus strains H1N1, H2N2 and H3N2 [14–18, 20]. In Figure 2 the most potent compounds against influenza A H2N2 virus, a strain with exquisite sensitivity to amantadine, are presented. Several compounds showed marked biological activity. Of the compounds of series 3-8 compound 3b had the highest activity, 230-fold higher than that of amantadine, while 4a had the highest selectivity index (SI>1045) [14–16]. This SI value is one of the highest known for anti-influenza A agents.
111
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
7 times more active than amantadine amantadine
I.
ipophilic scaffold rimantadine
SI > 310
SI = 1785
230 times more active than amantadine SI > 357
57 times more active than amantadine SI > 83.4
4.3 times more active than amantadine SI >1045
1.9 times more active than amantadine SI > 431
1.6 times less active than amantadine
3.9 times less active than amantadine SI = 58
SI = 147
101 times > more active than amantadine SI = 83.4
3.6 times > more active than amantadine SI = 125
CH2-Ni -HCI \u
6d
151 times more active than amantadine SI = 24.5
4.3 times more active than amantadine SI = 207
Figure 2. Antiviral activity and selectivity indices for the most potent compounds of series 3-8.
To investigate the SAR for the compounds of series 3-8 and for aminoadamantanes in general, we sought a conformational analysis study. The conformational analysis of compounds 3-8 was based on a combination of NMR spectroscopy and molecular mechanics calculations [21, 22] . The lowest in energy conformers for compounds 3b, 3c, 4a, 4b, 5a, 5b, 6b, 6d, 7a, 8a are shown in Figure 3 and are consistent with NMR data. Their conformational descriptors are summarized in Table 1. The detailed conformational analysis for some of the compounds of series 3-8 has been published in the literature [18, 23, 24].
112
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
4a
3c
5a
7a
Figure 3. Lowest energy conformers of compounds 3b, 3c, 4a, 4b, 5a, 5b, 6b, 6d, 7a, 8a derived from using a combination of NMR spectroscopy and molecular mechanics calculations.
113
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
Table 1. Lowest energy conformers of compounds 3b, 3c, 4a, 4b, 5a, 5b, 6b, 6d, 7a, 8a derived from a combination of NMR spectroscopy and MM+ calculations.18 Conformer descriptors
Compound
3b 3c 4a 4b 5a 5b 6b
conformation around free rotating bonds
2-Ad" group orientation
-
eq" eq"
heterocycle conformation E(l)',N-Me(ax) E(1), N-Me(ax)
ch" ch, N-Me(ax) ' ch ch, N-Me(ax)
-
gauche(+)-trans (C2-CH2 bond) ch e 7a trans (C3-C2' bond) 8a trans (C2-C2' bond) Notes and abbreviations: a Ad: adamantyl, equatorial, 'envelope with C-l out of plane, chair, e envelope with C-2 out of plane 6d
E(2)
The lowesr energy conformers were superimposed to reveal their similarities and differences (Figure 4). In general, for the most active compounds amine nitrogen atoms lie in a distance of 1.5 - 2.5 A away from the 2-adamantyl carbon. carbon framework in the vicinity of the 2-adamantyl carbon
Figure 4. Superposition of the lowest energy conformers of compounds 3b, 3c, 4a, 4b, 5a, 5b, 6b, 6d, 7a,
The biologically active compounds 3b, 3c, 4a, 4b, 5a, 5b, 6b, 6d are aminoadamantanes with an amino group substituent and a carbon substituent attached at
114
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
the C-2 position of the adamantane nucleus. The nitrogen atom is either directly attached (3b, 3c, 4a, 4b, 5a, 5b) or is remote from the 2-adamantyl carbon (6b, 6d).
HCI
It is important to note that while 1-adamantanamine is a specific antiviral drug, 2adamantanamine is inactive. The obtained results point out that a carbon substituent in the vicinity of the 2-adamantyl carbon can lead to a remarkable increase in antiviral activity (Figure 4). This result can possibly be rationalized in terms of the steric demands for optimum activity, i.e. a lipophilic group other than 2-adamantyl than can possibly fit better in the pocket around Gly–34.
Conclusion Compounds bearing an amino group substitution at the C-2 position of the adamantane nucleus and a carbon skeleton framework in the vicinity of the 2-adamantyl carbon showed increased anti-influenza A virus activity. For the most active compounds, the amine nitrogen atom lies in a distance of 1.5 - 2.5 A away from the 2-adamantyl carbon. The conformational analysis of the compounds reveal that M2 ion channel blockers with improved biological activity can be designed and synthesized. The efforts which led to the results described here will trigger future research and possibly the discovery of new anti-influenza A drugs. Future collaboration with structural biologists and biophysicists will help establishing this goal. Acknowledgments The research activities covered in this review were supported by research grants from the University of Athens, Greece.
N. Kolocouris et al. / In Searching for New Anti-Influenza A Drugs
References and Notes [1] N. J. Cox, K. Subarao, Lancet 354 (1999) 1277. [2] L. Simonsen, L. B. Schonberger, D. F. Stroup, N. H. Arden, N. J. Cox, The impact of influenza on mortality in the USA. In: L. E. Brown, A. W. Hampson, R. G. Webster, (Ed.), Options for the control of influenza III, Elsevier Science BV, Amsterdam, 1996; pp 26–33. [3] K.-J. Lui, A. P. Kendal, Am. J. Public Health 77 (1987) 712. [4] (a) J. K. Taubenberger, A. H. Reid, T. G. Fanning, Virology 274 (2000) 241. (b) J. K. Taubenberger, A. H. Reid, A. E. Kraftt, K. E. Bijwaard, T. G. Fanning, Science 275 (1997) 1793. [5] (a) A. J. Hay, A. J. Wolstenholme, J. J. Skehel, M. H. Smith, EMBO J. 4 (1985) 3021; (b) A. J. Hay, Semin. Virol. 3 (1992) 21; (c) L. H. Pinto, L. J. Holsinger, R. A. Lamb, Cell 69 (1992) 517. (d) K. C. Duff, P. J. Gilchrist, A. M. Saxena, J. P. Bradshaw, Virology 188 (1992) 14. [6] (a) K. C. Duff, R. H. Ashley, Virology 190 (1992) 485. (b) K. C. Duff, S. M. Kelly, N. C. Price, J. P. Bradshaw, FEBS Lett. 311 (1992) 256. [7] F. A. Kovacs, T. A. Cross, Biophys. J. 73 (1997) 2511. [8] (a) C. S. Gandhi, K. Shuck, J. D. Lear, G. R. Dieckmann, W. F. DeGrado, R. A. Lamb, L. H. Pinto, J. Biol. Chem. 274 (1999) 5474. (b) Figure 1 was reproduced after permission from J. Biol. Chem. [9] (a) K. Lundahl, J. Shut, J. L. M. A. Schlatmann, G. B. Paerels, A. Peters, J. Med. Chem. 15 (1972) 129, (b) R. Van Hes, A. Smit, T. Kralt, A. Peters, J. Med. Chem. 15 (1972) 132. [10] P. S. Manchard, R. L. Cerruti, J. A. Martin, C. H. Hill, J. H. Merrett, E. Keech, R. B. Belshe, E. V. Connell, I. S. Sim, J. Med. Chem. 33 (1990) 1992. [11] A. Garcia Martinez, E. Teso Vilar, A. Garcia Fraile, S. de la Moya Cerero, M. E. Rodriguez Herrero, P. Martinez Ruiz, L. R. Subramanian, A. Garcia Gancedo, J. Med. Chem. 38 (1995) 4474. [12] Burger's Medicinal Chemistry, in M. E. Wolff (Ed.), John Wiley & Sons, New York, 1998, Part II, pp 590–591. [13] A. Kolocouris, Ph.D Thesis (1995), available from National Documentation Center, Vas. Constantinou 48, Athens, Greece. [14] N. Kolocouris, G. B. Foscolos, A. Kolocouris, P. Marakos, N. Pouli, G. Fytas, S. Ikeda, E. De Clercq, E, J. Med. Chem. 37 (1994) 2896. „ [15] N. Kolocouris, A. Kolocouris, G. B. Foscolos, G. Fytas, J. Neyts, E. Padalko, J. Balzarini, R. Snoeck, G. Andrei, E. De Clercq, J. Med. Chem. 39, (1996) 3307. [16] G. Fytas, G. Stamatiou, G. B. Foscolos, A. Kolocouris, N. Kolocouris, M. Witvrouw, C. Pannecouque, E. De Clercq, Bioorg. Med. Chem. Lett. 7 (1997) 1887. [17] A. Kolocouris, D. Tatarides, G. Fytas, G. B. Foscolos, T. Mavromoustakos, N. Kolocouris, E. De Clercq, Bioorg. Med. Chem. Lett. 9 (1999) 3465. [18] G. Stamatiou, A. Kolocouris, N. Kolocouris, G. Fytas, G. B. Foscolos, J. Neyts, E. De Clercq, Bioorg. Med. Chem. Lett. 20 (2001) 2137. [ 19] Unpublished results. [20] S. Shigeta, K. Konno, T. Yokota, K. Nakamura, E. De Clercq, Antimicrob. Agents Chemother. 32 (1998)906. [21] A. E. Howard, P. A. Kollman, J. Med. Chem. 31 (1988) 1669. [22] Molecular mechanics were performed using the MM+ force field provided by the Hyperchem. This force field is an extension of MM2 force field. Molecular mechanics calculations give the most consistent results with experimental data for piperidine and pyrrolidine conformers. [23] A. Kolocouris, E. Mikros, N. Kolocouris, J. Chem Soc. Perkin Trans 2 (1998) 1701. [24] A. Kolocouris, J. G. Outeirino, J. E. Anderson, G. Fytas, G. B. Foscolos, N. Kolocouris, J. Org. Chem. 66 (2001) 4989. [25] P. E. Aldrich, E. C. Hermann, W. E. Meier, M. Paulshock, W. W. Prichard, J. A. Snyder, J. A. Watts, J. Med. Chem. 14 (1971) 535.
115
116
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Design, Synthesis and Conformational Properties of Linear Analogues of Human Myelin Basic Protein Epitope MBP1–11 S. Deraos1, T. Tselios1,1. Daliani2, P. Zoumpoulakis3, L. Probert2, A. Troganis4, P. Papathanassopoulos5, T. Mavromoustakos3 and J. Matsoukas1 1
2
Department of Chemistry, University of Patras,Patras, Greece 26500 Department of Molecular Genetics, Hellenic Pasteur Institute, Athens, Greece 11521 3 National Hellenic Research Foundation, Institute of Organic and Pharmaceutical Chemistry, 11635 Athens, Greece 4 Department of Chemistry, University of Ioannina, 45110 loannina, Greece 5 Department of Medicine, University of Patras, Patras, Greece 26500
Abstract. Experimental Allergic Encephalomyelitis (EAE) is induced in experimental animals immunized with Myelin Basic Protein (MBP). In EAE, residues 1–11 of myelin basic protein are the dominant disease-inducing determinants in PL/J and (PL/JxSJL/J)F1mice. Analogues of disease-associated epitopes have been identified which alter disease progression upon co-immunization. Based on the Human MBP1. 1 2 3 1 5 6 7 8 9 10 11 (analogue 11 sequence [Ala -Ser -Gln -Lys -Arg -Pro -Ser -Gln -Arg -His -Gly ] 1) we designed and synthesized linear analogues in which Lys at position 4 was replaced with Ala, [Ac-Alal-Ser2-Gln3-Ala4-Arg5-Pro6-Ser7-Gln8-Arg9-His10-Gly11] (analogue 2) or Tyr, [Ac-Ala1-Ser2-Gln3-Tyr4-Arg5-Pro6-Ser7-Gln8-Arg9-His10-Gly11] (analogue 3). In these analogues the N-terminus was acetylated. For the synthesis of linear MBP analogues, we resorted to the Fmoc/tBu methodology utilizing the 2 chlorotrityl chloride resin. In bioscreening assays we found that these linear analogues were not effective in inducing or inhibiting EAE in Lewis rats in contrast to PL/J mice, where these analogues were found active. However, it was noticed that analogue 2 when co-injected with guinea pig MBP72–85 (1020) gave a maximum clinical score for two additional days, in contrast to analogues 1, 3 which didn't alter effect caused by MBP72–85 (Fig.l).
S. Deraos et al. / Linear Analogues of Human Myelin Basic Protein Epitope
I
H
«
9
9
MBP 1–11
9
117
B
C—N-CH-G-HN-CHC—HN-CHC—HN-CH-C—«N—CH-C—OH | i i i |
OH
CHj
CLJ
C=0
6
r-u
CH
£H2
'
2
OH
' -2
'
2
'"
^
2
^
I
H
N"^)
^—NH
i
i..2
' *
2
C=NH
NH
CH2
NH2
C=NH NH2
11
] (analogue 1)
if H
II
99
9
ii
C—N-CH-C-HN-CHC—HN-CHC—HN-CH-C—HN—CH-C—OH 0
O
O
0
I /~^-
9
. . . . . . . H2N-CHC—HN-CHC—HN-CHC-HN-CHC—HN-CHC—NH CH3 CH2 CH2 CH3 CH2 V^ OH
.
CH22
,*u V"2
A,,
,
CH2 ' -^
QH
f*H
Vjrl
CH 22
i. .
|
CH ' V2
f'riz
M
n
V-NH
iMrl
£"2
C=0
n 2 CH ^ 2
=NH
^
NH2
C=NH NH,
Ac-Ala1-Ser2-Gln3-Ala4-Arg5-Pro6-Ser7-Gln8-Arg9-His10-Gly11 (analogue 2) O
0
,
,
9
9
i i
9
C—NH-CH-C—HN-CHC—HN-CHC—HN-CHC—HN—CH-C—OH
9
9
9
o
V
/^i
H2N-CHC—HN-CHC—HN-CHC—HN-CHC-HN-CHC—NH
CH3
CH2 OH
CH2 CH2 Cr
°
NH2
Jx,
CH2
CH2 J,H
CJ
AH' H2
V OH
? NH C=NH NH2
Y"2 QH
i CH2
i
6n2
fc
•"••<
i CH2
CH2
CH,
'
'
i
'
H
1
,"1
?=NH NH2
Ae-Ala1-Ser2-Gln3-Tyr4-Arg5-Pro6-Ser7-Gln8-Arg9-His10-Gly11] (analogue 3)
Figure 1: Chemical structures of the peptides
Introduction Multiple Sclerosis (MS) is a chronic inflammatory disease of the CNS characterized by focal T cell and macrophage infiltrates, demyelination and loss of neurologic function [1, 2]. MS is generally considered to be an autoimmune disease caused by neuroantigen-specific CD4+ T cells. Candidate autoantigens include constituents of the myelin sheath such as myelin basic protein (MBP) and proteolipid protein (PLP). Modern approaches towards the therapeutic management of MS involve the design and use of peptide analogues of disease-associated myelin epitopes to induce peripheral T cell tolerance [3, 4]. MBP epitopes have been identified to be sequences 1-11, 72-85, 87-99, 84-102, 143-168. In this study, MBP1–11
118
S. Deraos et al. / Linear Analogues of Human Myelin Basic Protein Epitope MBP 1
analogues have been synthesized and were tested for EAE effects in Lewis rats. Analogue 2 increased the clinical score of disease induced by MBP72–85, while analogues 1, 3 didn't change clinical effect caused by MBP72–85. Experimental section Chemistry: Synthesis of linear peptides 1, 2,3.
Fmoc-Gly-OH DMF/DIPEA
^ G.,
Fmoc Synthesis DIC/HOBl
Fmoc deprotech 20'J Piperidine r DMF
1 2 3
H;N-Ala-Set
1 Fmoc His(Tn) OH 2 Fmoc-Arg(Pbf)-OH Fmoc-Gln-OH Fmoc-Set(lBu) - OH Prc-OH noc.Arg(Pbf>-OH moc Lys(Boc> OH i«.AIa-OH moc-TyrfTn)-OH moc GlnOH moc ScrttBu) OH Ala OH
Arg(Pbf>Pro-Sef(tBu)-Gln-Arg(Pbf)-His(TnK;iy
H2N-Ala-SertlBu)-Gto-AU-Aig(PbO-Pn>Ser(tBu>-Gln-AiK(Pbf)-His(Tn)-Gly H2N-Ala-Ser(lBu)-Gln-Tyr(Trt)-Arg(Pbf)-Pro-Ser(tBu)-Gbi-Arg(PbO-His(Tn)-Gly
-^^
Acetylation with (CH3CO)2O/DIPEA X 10/XIO in DMF 1
Ac-Ala-Ser(tBu)-Gln-Lys(BochArg(Pbf)-Pro-Sef(tBn)-Gln-Arg(PbO-His(Tn>-Gly
2
Ac-Ala-Ser(tBu)-Gln-AU-Arg(Pbn-Pn>Ser(tBu)-Gln-Arg(Pbf)-His(Tn>-Gly
3 Ac-Ala-Ser(lBu)-Gln-Tyr(Tn)-Atg(PbO-Pro-Ser(tBu)-Gln-Arg(Pbf)-His(Tn)-Gij
^^ ^^ ^^
Cleavage with AcOH/TFE/DCM. 1/2/7
1 2 3
Ae-Ala-Ser(tBu)-Gto-Lys(Boc>-Arg(Pbf>-Pro.SerttBu)-Gte-Aig(Pbf-His(Tit)-Gly43H Ac-Ala-Ser(tBu)-Gln-Ala-Arg(Pbf-Pro-Sei(tBu)-Gln-Arg(Pbf>-His(Tn)-Gly-O Hc-Ala-Ser(tBu)-Gln-Tyr(Tn)-Arg(Pbf-Pro-Ser(tBu)-Gln-Arg(Pbf)-His(Tn)-Gly-OH Deprotection with TFA/DCM/EDTH/AIIISi 65/25/6/4 1 2
Ac-Ala-Ser-GlnLys-Arg-Pro-Ser-Ghi-Arg-His-Gly-OH Ac-Ala-Ser-Gln- Ato-Arg-Pro-Ser-Gln-Arg-His-Gly-OH
3
Ac-Ala-Ser-Gln-Tyr Arg-Pro Ser Gln Arg His-Gly OH
Scheme 1: Synthesis of linear peptide analogues 1, 2 and 3. The linear peptides were prepared on a 2-chlorotrityl chloride resin using solid-phase peptide synthetic methods [16, 17, 18]. The first Na -Fmoc (9-fluorenylmethyloxycarboxyl)-protected amino acid [Fmoc-Gly-OH was coupled to the resin in 1 h in the presence of diisopropylethylamine (DIPEA) in dichloromethane (DCM). The remaining peptide chain was assembled by sequential couplings of the Fmoc protected amino acids, in the presence of N,N'-diisopropylcarbodiimide (DIC) and 1-hydroxybenzotriazole (HOBt) in N.N-
S. Deraos et al. / Linear Analogues of Human Myelin Basic Protein Epitope
MBP 1-11
119
dimethylformamide (DMF) for 4 h.. The Fmoc protecting group was removed by treatment with piperidine (20% in DMF). The acetylation of N-terminal was achieved with acetic anydride/diisopropylethylamine (CH3CO)2O/DIPEA in DMF. The protected peptide resin was then cleaved with the splitting solution dichloromethane/acetic acid/2,2,2-trifluoroethanol (DCM/AcOH/TFE, 7:1:2 1h at room temperature). The deprotection of protected linear peptides was achieved with 65% trifluoroacetic acid (TFA) in DCM in the presence of 1,2 ethanedithiol (EDT) and anisole (Scheme 1). The purification was achieved using HPLC reversed-phase chromatography and the peptide purity was assessed by analytical HPLC and by mass spectrometry (ESIMS)18. Structure Elucidation and Conformational Analysis The structure and conformational analysis properties of the peptides under study, were obtained using a combination of 1D and 2D NMR spectroscopy. NOESY experiments showed the linearity of the molecules. Computational analysis using different minimization algorithms with Dynamics experiments were applied to find their low energy conformers. (Figure 2)
Figure 2: Low energy conformers of synthetic peptides 1, 2 and 3.
120
S. Deraos et al. / Linear Analogues of Human Myelin Basic Protein Epitope MBP 1–11
Biological data Female Lewis rats (220g) were immunized subcutaneously into the hind footpads with MBP72–85 (30ug) and each of the three linear peptide (500u,g). Peptides were dissolved in PBS and emulsified in an equal volume of Freund's complete adjuvant (CFA, Difco) containing 4mg/ml heat-killed Mycobacterium tuberculosis H37Ra (Difco). Clinical EAE was graded on a scale of 0–4 by established criteria as follows: 0, no discernable disease; 0.5, weight loss; 1, flaccid tail; 2, hind limb weakness; 3, paraplegia; 4, paraplegia with forelimb weakness, moribund (Figure 3).
10
12
Days
Figure 3: The modulatory effect of linear analogues 1–3on EAE upon co-injection with MBP72–85.
Discussion MBP is hypothesized to be a potential autoimmunogen in MS, largely because injection with MBP or MBP-reactive T cells induces Experimental Autoimmune Encephalomyelitis (EAE) in animal models. EAE is one of the best studied experimental animal models of MS, and represents an invaluable in vivo system for the evaluation of therapeutic approaches. The N-terminal peptide Acl–11 of MBP, induces EAE in mice that have the H–2u and (H–2u x H–2s) major histocompatibility complex (MHC) haplotypes, such as PL/J and (PL/JxSJL/J)Fl, but cannot do so in mice with the H–2S haplotype, such as SJL/J, because Acl–11 does not bind to I-As [5–8]. SJL/J mice, however, develop EAE when injected with the MBP peptide 89–101, which binds to IAs but not to I-Au. Several different immunotherapies have been used successfully in treating or preventing EAE [9]. Peptides, either the encephalitogenic epitopes themselves, analogs derived thereof, or unrelated peptides, have been used to prevent or treat EAE [5, 6, 10–13]. One example is Acl–1 l[4Ala], an analog in which the lysine at position 4 is replaced with alanine. Position 4 is a MHC contact, as Acl-1 l[4Ala] binds with 50 x higher affinity to I-Au than does Acl-11 and stimulates Acl-11-specific T cells in vitro with greater efficiency [5, 8]. Acl-1 l[4Tyr], an analog in which tyrosine replaces Lys at position 4, binds to I-Au with 1,500-fold higher affinity than does Acl-
S. Deraos et al. / Linear Analogues of Human Myelin Basic Protein Epitope
MBP 1–11
121
11 and stimulates Ac 1–11 -specific T cells in vitro more efficiently than does Acl1 i[4Ala] [8, 11]. Other studies have shown that arginine at position 5 also contacts the MHC [14, 15] whereas glutamine at position 3 and proline at position 6 contact the T cell antigen receptor (TCR) [5]. Interestingly, Acl-11[4Ala], when co-immunized with Acl-11, inhibits the development of EAE [5, 10] while by itself doesn't induce a proliferative response in vivo in (PL/J x SJL/J)F1 mice. In the present study we have synthesized linear analogues based on the epitope of MBP1–11. These analogues were tested in Lewis rats, looking for suppressors of EAE induced by the encephalitogenic MBP72–85 (1020). The linear peptides were prepared on a 2-chlorotrityl chloride resin using solid-phase peptide synthetic methods. Acetylation of peptides increases lipophilicity allowing cell surface permeability. Analogues didn't show any agonist activity in Lewis rats. Furthermore to the evaluation for agonist activity we have tested these analogues for antagonist activity by co-injection with encephalitogenic MBP72–85. Analogues 1, 3 didn't show any antagonist activity. Analogue 2 in co-injection with MBP72-85 sustains the time of maximum clinical score (4) for two additional days. References 1. Steinman, L. Cell (1996), 85, 299. 2. Martin, R.; McFarland, H.; McFarlin, D. Ann. Rev. Immunol. (1992), 10,153 3. Hafler, D.; Weiner, H. Immunol. Rev. (1995), 144, 75. 4. Hohfeld, R. Brain. (1997), 120, 865. 5. Wraith, D. C., Smilek, D. E., Mitchell, D. J., Steinman, L. & McDevitt, H. O. Cell (1989), 59, 247-255. 6. Gautam, A. M., Pearson, C. I., Sinha, A. A., Smilek, D. E., Steinman, L. & McDevitt, H. O. J. Immunol., (1992), 148, 3049-3054. 7. Mason, K., Denney, D. W., Jr. & McConneil, H. M. J. Immunol., (1995), 154, 5216-5227. 8. Fugger, L., Liang, J., Gautam, A., Rothbard, J. B. & McDevitt, H. O. Mol. Med., (1996), 2, 181-188. 9. Martin, R., McFarland, H. F. & McFarlin, D. E. Annu. Rev. Immunol. (1992), 10, 153-187. 10. Smilek, D. E., Wraith, D. C., Hodgkinson, S., Dwivedy, S., Steinman, L. & McDevitt, H. O. Proc. Natl. Acad. Sci. USA, (1991), 88, 9633-9637. 11. Metzler, B. & Wraith, D. C. Int. Immunol., (1993), 5, 1159–1165. 12. Samson, M. F. & Smilek, D. E. J. Immunol., (1995), 155, 2737-2746. 13. Brocke, S., Gijbels, K., Allegretta, M., Ferber, I., Piercy, C., Blankenstein, T., Martin, R., Utz, U., Karin, N., Mitchell, D., et al. Nature (London), (1996), 379, 343-346. 14. Gautam, A. M., Pearson, C. I., Smilek, D. E., Steinman, L. & McDevitt, H. O. J. Exp. Med., (1992), 176, 605–609. 15. Wraith, D. C., Bruun, B. & Fairchild, P. J. J. Immunol., (1992), 149, 3765-3770 16. Tselios, T.; Probert, L.; Daliani, I.; Matsoukas, E.; Troganis, A.; Gerothanassis, P.; Mavromoustakos, T.; Moore, G.; Matsoukas J. J. Med. Chem., (1999), 42, 1170. 17. Barlos, K.; Gatos, D.; Hondrelis, J.; Matsoukas, J.; Moore, G.; Schafer, W. Sotiriou, P. Liebigs Ann. Chem. (1995), 951. 18. Barlos, K.; Gatos, D.; Schafer, W. Angew. Chem., Int. Ed. Engl. (1991), 30,590. 19. Tselios, T.; Probert, L.; Kollias, G.; Matsoukas, E.; Roumelioti, P.; Alexopoulos, K.; Moore, G.; Matsoukas J. Amino Acids, (1998), 14, 333. 20. Hogg, M. Int. J. Mass Spectrom. Ion Phys. (1983), 49, 25.
This page intentionally left blank
Part II.
Role of Membrances in Drug Activity
This page intentionally left blank
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
125
The Effects of Vinblastine Sulfate on Dipalmitoylphosphatidylcholine Single and Multiple Bilayer Membranes H. Maswadeh1, C. Demetzos1,1. Daliani 2 , T. Mavromoustakos2, G. Nounesis3, A. Tsortos3 Department of Pharmaceutical Technology and Pharmacognosy, Panepistimiopolis, Zographou 15571, University of Athens, Greece 2 Institute of Organic and Pharmaceutical Chemistry, National Hellenic Research Foundation, Vasileos Constantinou 48, Athens 11635, Greece Institute of Radioisotopes and Radiodiagnostic Products, NCSR "Demokritos", Aghia Paraskevi 15310, Greece Abstract. The thermal and dynamic effects of vinblastine encapsulated in large uni- and multilamellar vesicles upon the phospholipid membranes have been studied for Dipalmytoylphosphatidylcholine (DPPC). The hydrophilic vinblastine sulfate molecules have been encapsulated using a pH gradient technique. MAS 13C NMR spectroscopy at various temperatures and a combination of high-precision differential scanning calorimetric techniques have been employed to study the effects upon the lipid phase transition sequence and especially upon the gel/liquid-crystalline phase transition. The calorimetric and spectroscopic results demonstrate that the encapsulation of vinblastine results in the abolishment of the Lp' to Pp' pretransition and most importantly in an increase of the molecular cooperativity of the single-bilayer phospholipid membranes along with an increase in the total enthalpy change (AH) for the main lipid phase transition. Computational analysis points out the intermolecular interactions between vinblastine and DPPC bilayers that can explain the thermograms and the NMR spectra.
Introduction Vinblastine (Figure 1) is a mitotic inhibitor isolated from the plant Catharanthus roseus G. Don. (Apocynaceae). It is used clinically in the treatment of leukemia and Hodgkin's disease, breast carcinoma, Wilm's tumor, Ewing's sarcoma and small-cell lung cancer, either alone or in combination with other chemotherapeutic agents [1]. Vinblastine enters into animal cells by diffusion through the plasma membrane. The permeability of the membrane depends upon the molecular order and thus the mobility of the membrane lipids
126
H. Maswadeh et al. / The Effects of Vinblastine Sulfate
[2–4]. The interactions of water-soluble drugs with phospholipid membranes have been studied extensively [5]. Their presence almost always causes domain formation and thus the alteration of the lipid molecular order in the gel phase by inducing fluidity [6]. The resulting interface increase within the bilayer leads to an analogous increase of its permeability and eventually to a leaky bilayer. The macroscopic thermodynamic parameters are also directly affected. The broadening of the heat capacity peak (Cp) at the gel/liquidcrystalline phase transition, the lowering of the transition temperature and the decrease of the total enthalpy are often encountered.
9'
6:
5'
3'
•C,H5
14
H3co2c
12
Scheme: Molecular Structure of vinblastine.
Previous studies of the interactions of vinblastine sulfate with phospholipid membranes focused upon the thermal effects of this antitumor agent on Dipalmytoylphosphatidylcholine (DPPC) multibilayer vesicles, and contrary to most other drugs, the presence of vinblastine was found to be followed by an increase in the total enthalpy change of the main lipid phase transition and occasionally by an increase of the transition temperature. [2–4]. While these results indicate a potentially interesting mechanism for reducing lateral heterogeneity in the membranes, the transition for the multilamellar vesicles was nevertheless characterized by reduced cooperativity. Since the low dimensionality is important for the dynamic heterogeneity of lipid bilayers, which in turn may be quite important for biological functions, the thermal effects of vinblastine sulfate upon single and multiple bilayer vesicles have been investigated. Additional structural and conformational information was gained by high-resolution solidstate NMR spectroscopy experiments at various temperatures. The detailed study of the interactions of vinblastine with phospholipids may improve the design of liposomal formulations for most efficient vinblastine delivery and may also be used to evaluate the way vinblastine interacts with the membranes of living cells.
H. Maswadeh et al. / The Effects of Vinblastine Sulfate
127
Results Differential Scanning Calorimetry: The high-precision microcalorimetric results for the interaction of DPPC and uni- and multilamellar vesicles with vinblastine sulfate are displayed in Figure 1. In Fig. 1A the Cp versus, temperature trace for pure DPPC is shown for a mixture of 65% large unilamellar (LUV) and 35 % multilamellar vesicles (MLV). The transition from the Lß' to the Pß' (ripple) phase is characterized by a very weak Cp anomaly at 33.5 °C. The contribution to this Cp peak comes exclusively from the multilamellar vesicles in accordance to previous studies and to our results for all-MLV samples. In unilamellar systems, dynamic lateral heterogeneity is inherent due to the low dimensionality [6]. This is emphatically demonstrated in the Cp anomaly describing the main lipid phase transition, which for the unilamellar system, has a Cp maximum at 41.2 °C and is described by a broadened peak [7]. On the other hand, for multilamellar vesicles the transition temperature (Tm) at 41.2 °C is characterized by a sharp Cp anomaly. When vinblastine sulfate is incorporated in the aquatic interior and the lipid membranes of the exact system displayed in Fig 1A, in molar ratio DPPC:vinblastine = 100:17, the effects are quite dramatic. As it can be seen from the Cp trace displayed in Figure IB, the weak Lp' —> Pß' peak can no longer be detected and both the uni- and multi lamellar peaks appear to have merged in a single, sharp anomaly at 41.76 °C characterized by a molar enthalpy change AH = 5.32 kcal/mol, which is larger than AH for either the unilamellar or the multilamellar system (Table 1). As a measure of the molecular cooperativity within the bilayers, and thus of the dynamic heterogeneity, the ratio of AH to the van't Hoff enthalpy (AHvn) has been calculated, which approximately gives the average number of correlated molecules in domains within the bilayer undergoing the transition [8]. The number of molecules within a cooperative unit (C.U.) is also listed in Table 1. The straightforward conclusions drawn from the results of Table 1 is that the presence of vinblastine sulfate in the interior of unilamellar DPPC vesicles induces a sharp increase of AH, Tm and the size of the C.U.. For MLVs, in accordance to previously published findings, there is an increase in AH accompanied by a decrease in Tm and the size of C.U. for the main phase transition and the abolishment of the pretransition peak.
45 50 30 35 40 45 50 Temperature Figure 1: (A) Cp versus T data for a mixture of single- and multiple bilayer DPPC vesicles, Tp is the gel to ripple-phase pretransition (Lß' to Pß'), Tms is the main-lipid phase transition for single and Tm for multiple bilayer vesicles. (B) Cp vs. T data for the same mixture as in (A), with vinblastine sulfate incorporated in the DPPC membranes at concentration x = 0.17 with respect to DPPC.
128
H. Maswadeh et al. / The Effects of Vinblastine Sulfate
Table 1: Quantitative thermal data for DPPC bilayers with or without vinblastine LUV
DPPC DPPC:vinblastine (100:17)
Tm (°C)
AH (kcal/mol)
41.20 41.76
5.32 7.77
C.U. (# of DPPC molecules) 76 180
Tm (°C)
41.20 39.40
MLV AH (kcal/mol)
7.51 10.44
DSC studies of multilamellar DPPC bilayers without or with the presence of x=0.17 vinblastine reconfirm the findings of the high-accuracy microcalorimetry. The DPPC bilayers exist in the gel phase for temperatures lower than 33 °C, and in the liquid crystalline phase for temperatures higher than 42 °C. At ca 35 °C the phospholipid bilayers exhibit the "ripple phase" and the peak corresponds to the pretransition temperature [9]. Contrary to the results for the single-bilayer vesicles, the presence of x=0.17 vinblastine causes the decrease of the phase transition temperature and an increase of the width of the Cp peak for the main lipid phase transition. In agerement with the single bilayer results, the presence of the drug causes the abolishment of the pretransition and an increase of ca 40% in AH. Such a thermotropic behavior of the drugs or other additive in membrane bilayers is not unique and is reported in the recent literature. For example the effects of ursodeoxycholic acid (UDCAH) and ursodeoxycholate (UDCA) on the thermotropic phase behavior of aqueous bilayer dispersions of DPPC buffered at pH 7.0, were examined by DSC. For concentrations of UDCA 25–60 mol-% similar thermal effects were observed as with those we report for vinblastine. The authors using theoretical analysis interpreted these results in terms of an induced interdigitated gel phase stabilized by DPPC:UDCA molecular interaction. The interdigitated gel phase exists in equilibrium with micelles, whose structures remain to be elucidated, of various UDCA:DPPC mole ratios. It is stated that in the interdigitated phase there is a larger van der Waals interaction between hydrocarbon chains that gives rise to a larger transition enthalpy and a more cooperative phase transition [10]. Another recently reported example in the literature is the one with grafted polyninyl pyrrolidone-palmityl (PVP-p) conjugate polymers [11]. A differential scanning calorimetry study of phosphocholines mixed with paclitaxel and its bromacylated taxanes showed that these bulky water-soluble molecules exert similar thermotropic behavior with vinblastine. The increase of AH was interpreted as a possible increase in the van der Waals interactions in the interior of the membrane bilayers. The possibility that taxanes could form micelles or emulsions with phospholipids which are not detectable under DSC conditions is also mentioned [12]. Other examples mentioned in the literature are those of synthetic polyphenols and ethylene oxide containing lipopolymers and tri-block copolymers on lipid bilayers of palmitophosphatidylcholine [13–14]. High Resolution I3C NMR spectroscopy: We have applied I3C MAS experiments to study the dynamic and thermotropic properties of bilayers containing DPPC with or without vinblastine. When an additive is incorporated into membrane bilayers different changes can be observed in the spectrum. These can be summarized to: (a) Changes in the peak intensity and the line-width due to induced membrane fluidity; (b) Change in the chemical shifts
129
H. Maswadeh et al. / The Effects of Vinblastine Sulfate
values of individual carbon nuclei of the membrane lipid due to the interaction with vinblastine molecules, (modified transition profiles); (c) Appearance of peaks from the carbon nuclei of the of the incorporated additive. A comparison of !3C NMR spectra of DPPC bilayers without and with vinblastine at temperature range of 25 °C-43 °C reveals the following: (a) The line-width of the peaks is narrower in the bilayers containing vinblastine; (b) Chemical shifts versus temperature plots of the major peaks corresponding to head-group, the interface and the hydrophobic region indicate that the presence of the drug causes mostly an increase of the chemical shift observed in the corresponding peaks of the DPPC bilayers alone. This is evidence of an ordering effect since the chemical shifts of the carbons in the membrane decrease with increasing temperature; (c) The peaks mainly in the region of 100–150 ppm, characterized by small intensity, demonstrate the presence of the drug in the membrane bilayers.
309 K
300 K
180 160 140 120 100 80 60 (ppm
40
20
0
-20
180 160 140 120 100 80 60 (ppm)
40
20
0
-20
Figure 2: High-resolution solid-state 13C NMR spectra of multilamellar DPPC (left) and DPPC/vinblastine (x. = 0.17) (right) at temperatures 25 - 43 °C.
Discussion There are two reports in the literature that examine the thermal effects of vinblastine in DPPC bilayers. However, the results obtained are controversial probably due to the different experimental conditions. In addition, no explanation was given for the obtained
130
H. Maswadeh et al. / The Effects of Vinblastine Sulfate
results [3,4]. We therefore found interesting to study more thoroughly the thermal effects of vinblastine in membrane bilayers using DPPC bilayers and combine the results with those obtained by NMR spectroscopy. In addition, a study is in progress in which the thermal and dynamic effects of vinblastine in different phospholipid bilayers will be studied in the presence and absence of cholesterol. The obtained thermograms can be explained if the intermolecular interactions between vinblastine and the DPPC bilayers are considered. The water-soluble amphoteric bulky molecule of vinblastine can be associated with different molecular interactions with DPPC bilayers. In particular, the HSO4" can interact electrostatically with N+(CH3)3 headgroup of the phospholipid, the hydrophobic regions of vinblastine can be associated with Van der Waals interactions with the acyl chains of the phosphoiipid and the positively charged molecule with the PO43- of the polar group. The molecule is anticipated to span both the polar and the hydrophobic region. Some of its polar groups (i.e. hydroxyl and methoxate) are enforced to fit in the hydrophobic core of the bilayer and may thus induce some destabilization of the molecule. However, the net effect is the stabilization of the membrane bilayer as it is depicted from the increase of C.U. and AH.
Acknowledgement This work was supported by grants from General Secretariat of Research and Technology and P.N. GEROLYMATOS S.A Pharmaceutical Company
References [1] I. S. Johnson, J. G. Armstrong, M. Gorman and J. P. Burnett, Cancer Res 23 (1963) 1390. [2] F. Berleur, V. Roman, D. Jaskierowicz, D. Davellose, F. Leterrier, L. M. T. Saraga and G. Madelmont, Biochem Pharmac 34 (1985) 3081. [3] L. T. M. Saraga, G. Madelmont, C. H. Legrand and S. Metral, Biochem Pharmac 30 (1981) 411. [4] L.T.M. Saraga and G. Madelmont, Biochim Biophys Acta 728 (1983) 394. [5] K. Jorgensen, J. H. Ipsen, O. G. Mouritsen, D. Bennett, M. J. Zuckermann, Biochim. Biophys. Acta 1062 (1991) 227. [6] O. G. Mouritsen and K. Jorgensen, Chem. Phys. Lipids 73 (1994) 3. [7] G. Nounesis, B.Ratna, S.Shin, R. S. Flugel, S.Sprunt, J. D. Litster, A. Singh, R. Shashidhar, S Kumar, Phys. Rev. Lett. 76 (1996) 3650. [8] J. T. Mason, Methods in Enzymology 295 (1998) 468. [9] M. J. Janiak, D. M Small, G. G. Shipley, Biochemistry 15 (1976) 4575. [10] M. T. Cotisel, I. W. Levin, J. Phys. Chem. B 101 (1997) 8477. [11] M. Savva, V. P. Torchillin, L. Huang, Journal of Colloid and Interface Science 217 (1999) 166. [12] S. Ali, S. Minchey, A. Janoff, E. A. Mayhew, Biophysical J. 78 (2000) 246. [13] N.-W. Huh, N. A. Porter, T. J. Mclntosh, S. A. Simon, Biophysical J. 71 (1996) 3261. [14] T. S. Baemark, T. S. Pedersen, K. Jorgensen, O. G. Mouritsen, Biophys. J. 73 (1997) 1479.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Stealth Liposomal Vinca Alkaloids (Vinorelbine and Vincristine) and in vitro Studies on Release by Buffer and Rat Plasma C. Demetzos Cancer Research Institute and Department of Pharmacology, University of California San Francisco, California, 94143-0450 Present address: School of Pharmacy Dept. of Pharmacognosy Panepistimiopolis Zografou 15771 University of Athens, Greece. This paper is dedicated to the memory of Professor Demitrios Papahadjopoulos
Abstract. This experimental article provides data concerning of the encapsulation of vinorelbine (VNR) in various liposomal composition which was achieved using different ammonium and sodium salts. The amount of VNR trapped inside the liposomes and the rate of release at 2 °C and 37 °C in buffer and in rat plasma up to 50% by volume at 37 °C, 45 °C and 55 °C (thermosensitivity liposomes), was determined by UV-vis spectrometry. VNR was encapsulated into Sterically Stabilized Liposomes (SSL) with high efficiency at 98%, using ammonium sulfate pH gradient method. The results concerning the rate of release, suggest that the lipid composition of the liposomes, the external solution (buffer or plasma) as well as the temperature play an important role of the drug release from the liposomes. Vincristine (VNC) was also studied in parallel experiments for comparative reasons.
Introduction The Vinca alkaloids (Fig. 1) are one of the most important class of antineoplastic drugs. Vincristine (VNC) and Vinblastine (VNB) the two important Vinca alkaloids were isolated from the periwinkle plant Catharanthus rosea (syn. Vinca rosea) [1]. The Vinca alkaloids exhibit major differences in toxicity, pharmakokinetic behavior and antitumor spectra, despite having only minor structural differences [2]. The mechanism of action of the Vinca alkaloids is already well established. They bind specifically to free tubulin dimers, disrupting the equilibrium between free tubulin dimers and assembled polymers. The toxicity of the Vinca alkaloids is also well described [3–5]. Vinorelbine (VNR) (5-noranhydrovinblastine) belongs to the Vinca alkaloids and has the molecular formula C45H54N4O8 . (2C4H6O6) and molecular weigh 1079.12. It is synthesized from anhydrovinblastine [6]. Its antitumor activity is equivalent to that of VNB and also has
132
C. Demetzos / Stealth Liposomal Vinca Alkaloids
synergistic activity with other drugs such as cisplatin and etoposide. VNR has reduced neurotoxicity compared with VNC and VNB as well as better hematologic tolerance [7,8]. Its cytotoxic activity is higher than VNC and VNB in P388 and L1210 leukemias. It has also shown a significant cytotoxic effect in B16 melanoma in contrast to other Vinca alkaloids, which were inactive. In Phase II studies VNR has shown a higher antitumor activity than VNC and VNB in breast, lung and ovarian carcinoma [9]. There is also extensive data confirming the activity of VNR in metastatic breast cancer [10].
Vincristine, R = CHO
MeO' Vjnorclhiiu'
Figure 1: Structures of Vinca alkaloids.
Liposome-encapsulated drugs appear to represent an increasingly useful method for delivery of chemotherapeutic agents. [11]. This technology is best exemplified in the case of doxorubicin, and several studies have shown that liposome-mediated alternations of this drug concerning cardiotoxicity and nephrotoxicity [12]. Recent studies using liposomeformulated doxorubicin has shown that its antitumor activity as well as its circulation time, are better than those obtained for the free doxorubicin [13, 14]. Vincristine one of the Vinca alkaloids, encapsulated into liposomes has been decreased its toxic side effect while increasing or maintaining therapeutic activity [15]. As a step toward the development of tumor-targeted vinorelbine formulation, we have studied loading of vinorelbine into sterically stabilized derivative of phosphadidylethanolamine (PEG-DSPE) liposomes (SSL) protected against reticuloendothelial clearance by addition of poly(ethylene glycol) (MW 1900). In this study we have investigated the ability of VNR to encapsulate into SSL liposomes, using ammonium and sodium salts with the same anion, the rate release in buffer and in 50% rat plasma as well as the potential benefit of a local hyperthermia treatment at 45 °C and 50 °C. This study presents the formulation of VNR into a liposome system that could be shown a high efficiency encapsulation, low drug release in buffers and in rat plasma 50%, as well as improvements on the pharmakokinetic properties of VNR. We therefore evaluated the properties of different phospholipids in order to prepare various types of liposomes, different interior buffers with alkaline or acidic pH. These parameters are important factors in the loading of lipophilic amines, such as Vincristine, into vesicles exhibiting a transmembrane pH gradient [14]. Vinorelbine was encapsulated into liposomes for the first time in order to explore its entrapment efficacy the release from the liposomes under various conditions as well as its ability to maintain high blood levels for extended
C. Demetzos / Stealth Liposomal Vinca Alkaloids
133
periods. Since the vinca alkaloids are cell-cycle-specific cytotoxic drugs, it may be predicted that liposomal formulation of VNR could be therapeutically advantageous. This study could be useful for the antitumor efficacy of VNR, to provide pharmacokinetic and bioability advantages after in vivo parenteral administration. Liposomal VNR is expected to be of wide utility in variety of human malignancies. Materials and Methods Materials: Distearoylphosphatidylcholine (DSPC), Egg phosphatidylcholine (EPC), rhodamine phosphatidylethanolamine (Rh-PE) and polyethylene glycol phosphatidylethanolamine (PEG-PE) were purchased from Avanti Polar Lipids , In. (Alabaster, Alabama). Cholesterol (CHOL), was obtained from Calbiochem (La Jolla, CA). Polycarbonate membrane was purcased from Poretics Corporation Livermore, CA, USA. Extruder device was purchased from Lipex Biomembranes, Vancouver, BC, Canada. Rat plasma (with heparin as anticoagulant) was obtained from 6-8 weeks old Sprague-Dawley male rats. Navelbine (Borroughs Wellcome, Co) as Vinorelbine tartrate was purchased from the Moffit Hospital Pharmacy (UCSF). Vincristine as Vincristine sulfate was purchased from Sigma Chemical Co (St. Louis, MO). Salts and buffers were purchased from Sigma Chemical Co. Preparation of liposomes and drug loading: Different unilamellar vesicles composed of DSPC, and EPC with CHOL and PEG-PE, were prepared from multilamellar vesicles, by membrane extrusion method (17, 18). The molar ratio used for DSPC or EPC/CHOL/PEGPE was 30:20:1.8. Rh-PE was added at mole ratio 0.03 moles. The lipids were mixed in chloroform and the solvent was removed under reduced pressure at 40 °C. Multilamellar vesicles were formed by vigorous shaking of the lipid film in an aqueous solution of 250 mM ammonium sulfate and 1 mM Deferoxamine (pH = 5.5). The mixture was then vortexed for a short time and then at 55 °C for 1h, and the preparation was treated by freeze-thaw for 10 times. The resultant large oligolamellar vesicles were extruded under pressure (200–300 psi), through double polycarbonate of 0.1 and 0.05 urn pore diameters 3 and 8 times respectively, using an extruder device heated at 55 °C. The particle diameter as measured by dynamic light scattering (Coulter Model N4) is presented in the Table 1. The amount of the phospolipids was determined by phosphate assay. Unentrapped ammonium sulfate was removed at room temperature by gel filtration through Sephadex (G-75) equilibrated with 100 m M MES buffer at pH = 5.5 NaCl 173 mM osmolarity of 600 mOs. Vinorelbine was encapsulated by the ammonium sulfate gradient method [19] as follows: Vinorelbine tartatre was in a solution (water for injection) (10 mg/ml). 100-ul of this commercial solution (100 ul = 1 mg) was added to the liposomes suspension (1.8 ml of MES 100 mM, pH = 5.5, NaCl and 173 mM, 600 mOs). The liposome VNR mixture was incubated in water bath at 55 °C for 30 min [19]. After incubation, unentrapped VNR was removed by passing through Sephadex (G-75), gel filtration chromatography equilibrated with 100 mM MES, buffer at pH = 5.5 and osmolarity of 600 mOs. Encapsulation of VNR was studied using different buffers as external solution as well as sodium salts with the same anions, in order to explore the role of internal and external solution and the role of the pH inside and outside of liposomes on the incorporation of VNR (Table 2). The amount of VNR trapped inside the liposomes was determined in a UV-vis spectometer (UV 160U SHIMADZU) at 270 nm wavelength [20], after adding the sample into C2H5OH/H20 4:1 (20 ul into 1 ml total). The % of incorporated VNR was 98%. Vincristine was encapsulated using DSPC/CH/PEG-PE 30:20:1.8 mole ratio and 0.03 moles Rh-PE. The ammonium sulfate gradient method was used as for VNR encapsulation. Vincristine sulfate was in solution (5 mg/ml) in water for injection. 200 ul of this commercial product (200 ul = 1
134
C. Demetzos / Stealth Liposomal Vinca Alkaloids
mg) was added to the liposomes suspension (2.2 ml of MES 100 mM pH = 5.5 NaCl 173 mM 600 mOs). The liposome VNC mixture was incubated in a water bath for 30 min at 55 °C [19]. After incubation, unetrapped VNC was removed by passing through Sephadex (G75) gel filtration chromatography, and the column was equilibrated with MES 100 mM pH = 5.5 NaCl 173 mM 600 mOs buffer. The amount of the drug trapped inside the liposomes was determined in a UV-vis spectrometer using 297 nm as a wavelength [20], after adding the sample into C2H5OH/H20 4:1 (20 ul into 1 ml total). The % of incorporated VNC was 97%. Method of assay for VNR and VNC release: Liposomes composed of DSPC/CH/PEGPE, with (NH4)2SO4 as an internal solution were used to determine the drug (VNR, VNC) release. We used as external buffers MES 20 mM pH = 5.4 NaCl 150 mM 300 mOs and HEPES 20 mM pH = 7.2 NaCl 144 mM 290 mOs as well as 50% rat plasma as external medium. VNR retention was measured at 2 °C and at 37 °C, using the above buffers (MES and HEPES). 50% rat plasma was used as external medium to measure the retention of VNR at 37 °C. VNC retention was measured at 37 °C using the above-described MES and HEPES buffers as well as 50% rat plasma at 37 °C as an external medium. The thermosensitivity of liposomes was tested also in HEPES 20 mM pH = 7.2 NaCl 144 mM 290 mOs, buffer at 55 °C and 45 °C, as well as in 50% rat plasma at 45 °C for VNR and VNC. Aliquots were assayed in all cases at the indicated times after passage of the liposomes over Sephadex G-75 or Sepharose 4B gel filtration column, to remove VNR or VNC free, from liposomes. In all the release experiments, we considered 100% retention of the drugs (VNR, VNC) the amount presents at 0 time of incubation. In other words after immediately gel filtration (Sephadex G-75 or Sepharose 4B, 1–2 min delay), of the sample of VNR or VNC at time 0 of incubation, we assumed that 100% the drug amount by UVvis absorption is still encapsulated in liposomes. Results Drug loading: Vinorelbine (VNR) was incorporated inside liposomes composed of DSPC/CH/PEGPE 30:20:.8 mole ratio, with high efficiency, at 98% within 30 min at 55 °C, with a mole drug/phospholipid (D/PL) ratio of 0.123 (Table 1). Table 1. Incorporation of Vinorelbine (VNR) and Vincristine (VNC) into liposomes using (NH4)2SO4 Drug
Liposome composition (molar ratio)
VNR
DSPCa30 CHOLb20 PEGPEC 1.8 EPCd 30 CHOL 20 PEGPE 1.8 DSPC 30 CHOL 20 PEGPE 1.8
VNR
VNC
Entrapment %
Average particle diameter (nm)
Standard deviation of panicle diameter (±nm)
Drug incorporation (ug/umole phospholipid)
Ratio of drug to phospholipid mole ratio
98
84.8
25.6
133.5
0.123
10
101.1
33.5
40.3
0.037
97
84.8
25.6
126
0.136
"DSPC: Dislearoylphosphatidylcholine; bCHOL: Cholesterol; c distearoylphosphatidylethanolamine derivatized at the amino position with polyethylene glycol; dEPC: Egg phosphatidylcholine;
C. Demetzos /Stealth Liposomal Vinca Alkaloids
135
We have used the ammonium gradient-loading method to achieve efficient loading, a method which first was used for anthracyclines [12]. As Table 1 shows, the VNR encapsulation is strongly dependent on vesicle composition. VNR was incorporated inside liposomes composed of EPC/CH/PEGPE 30:20:1.8 mole ratio, with an efficiency of only 10% within 30 min at room temperature with a mole D/PL ratio of 0.037 (Table 1). Ammonium gradient method was used for the VNR loading. Such low percentage values of VNR entrapment in EPC/CH/PEGPE liposomes can be justified on the basis of the 'fluid' liposome formulation, in contrast to the 'rigid' liposome composition (DSPC/CH/PEGPE) which produced high percentage encapsulated VNR. Vincristine (VNC) was incorporated inside liposomes composed of DSPC/CH/PEGPE (30:20:1.8) with efficiency as high as VNR, with a mole ratio D/PL of 0.136 (Table 1). Using different salts as internal solution and different buffers as external solution (Table 2) we explored the role of pH during VNR encapsulation into liposomes. Table 2. Incorporation of Vinorelbine (VNR) into liposomes with composition of :DSPC/CH/PEGPE 30:20:1.8 (mole ratio) Internal solution (mM) (pH) (mOs) Entrapment % External solution 250 5.5 537 96 MESa (pH5.5) (NH4)2SO4 50 (NH4)2S04 250 5.5 537 TES" (pH 7.2) (NH4)2HPO4 250 8.2 488 25 MES ( pH 5.5) 12 (NH4)2HPO4 250 8.2 488 TES (H7.2) 250 4.7 420 63 NH4H2PO4 MES (pH 5.5) 44 250 4.7 420 NH4H2PO4 TES (pH 7.2) 200 5.0 526 10 MES (pH 5.5) Na2SO4 Na2HPO4 170 5.0 572 6 MES(pH5.5) 70 250 5.6 432 NH4C1O4 MES (pH 5.5) 52 NH4C1 250 5.5 450 MES (pH 5.5) a
MES : 100 mM pH = 5.5 600 mOs (NaCl 173 mM) ; b TES : 100 mM pH = 7.2 597 mOs (NaCI 173 m M).
According to the obtained results (Table 2), we can say the following: MES 100 m M pH = 5.5 NaCl 173 mM 600 mOs, as an external solution and Nttt salts as ((NH4)2SO4, NH4H2PO4, NH4C1O4, NH4Cl) which provide acidic pH interior, cause high percentage of encapsulated VNR. On the contrary MES 100 mM pH = 5.5 NaCl 173 mM 600 mOs, as an external solution and NH4+ salts such as (NH4)2HPO4 which provide alkaline pH interior, cause low percentage of encapsulated VNR. TES 100 m M pH = 7.4 NaCl 173 mM 594 mOs, as an external solution , causes low percentage of encapsulated VNR. Using other cations, such as Na+, instead of NH4+ as internal solution, the % of encapsulated VNR is decreased dramatically (Table 2). Drug release in buffers and in 50% rat plasma: The ability of liposomes to retain VNR appears related to pH of the external solution, according to our results as well as to the temperature. The retention of VNR into liposomes was monitored at 2 °C (Table 3) and at 37 ° C (Table 4), using different external solutions (MES 20 mM pH = 5.4 300 mOs, HEPES 20 mM pH = 7.2 290 mOs). We found that the retention of VNR was higher in HEPES buffer (pH = 7.2) than in MES buffer (pH = 5.4).
136
C. Demetzos / Stealth Liposomal Vinca Alkaloids
Table 3. Vinorelbine (VNR) retention in buffers at 2 °C *. External Time Concentration of VNR Concentration of solution encapsulated (ug/ml) (days) phospholipid (umol/ml)
Ratio of VNR to phospholipid mole ratio %retention MES 0 42.9 0.492 0.080 100 MES 8 27.0 0.464 0.053 66.2 HEPES 0 38.0 0.610 0.058 100 HEPES 8 36.0 0.492 0.067 115.5 a A tube containing 400 ul total volume, composed of 200 ul liposomal VNR and 200 ul MES 20 mM pH = 5.4 NaCI 150 mM 300 mOs or 200 ul HEPES 20 mM pH = 7.2 NaCL 144 mM 290 mOs was allowed to stand for 8 days at 2 ° C. In both cases aliquots (200 ul) were withdrawn at time period indicated. Free VNR was separated from the encapsulated by gel filtration (G-75 Sephadex). Table 4. Vinorelbine (VNR) retention in buffers at 37 °C External Time Concentration of Concentration of Ratio of VNR to solution phospholipid (hours) VNR encapsulated phospholipid (umole/ml) mole ratio % retention (ug/ml) MES" 42.9 0 0.492 0.080 100 MES 1 0.077 96.8 40.8 0.488 26.5 MES 3 0.049 62.1 0.494 6 0.514 MES 0.046 57.6 25.6 24 17.3 MES 0.026 32.9 0.608 0.057 100 HEPESC 0 38.0 0.610 1 0.053 92.9 HEPES 38.6 0.666 0.059 103 HEPES 3 38.8 0.600 0.058 102 HEPES 0.634 6 40.0 0.066 115 24 HEPES 45.0 0.624 A tube containing 1000 ul total volume composed of 500 ul liposomal VNR and 500 ul MES 20 mM pH=5.4 NaCI 1540 mM 300 mOs or 500 ul HEPES 20 mM pH=7.2 290 mOs NaCl 144 mM was incubated for 24 h at 37 °C. Aliquots (200 ul) were withdrawn at various time periods. Free VNR was separated from encapsulated by gel filtration (G-75 Sepharose) ; b MES : 20 mM pH = 5.4 300 mOs ;c HEPES : 20 mM pH = 7.2 290 mOs.
We obtained similar results for VNC (Table 5). The drug retention was measured also using as external solution 50% rat plasma. We found for VNR (Table 6) and for VNC (Table 7) that the drug retention into liposomes was as high as in HEPES buffer. Additional results on the thermosensitivity of liposomal VNR (Table 8) indicate that liposomal VNR was stable at 55 °C and at 45 °C when the external solution was HEPES 20 mM pH = 7.2 mOs. However the release of VNR is much higher when the external medium is 50% rat plasma at 45 °C (Table 8). The results on the thermosensitivity of liposomal VNC (Table 9), were similar to those mentioned for VNR. Table 5. Vincristine (VNC) retention in buffers at 37 oC a. Concentration of Concentration External Time solution (hours) VNC encapsulated phospholipid (umole/ml) (ug/ml) 0.660 HEPES 0 65.5 1 20 mM 0.826 76.0 68.7 0.772 3 pH = 7.2 0.788 290mOs 6 73.8 24 52.4 0.580 51.7 0.676 MES 0 1 20 mM 43.6 0.708 0.796 p H=5.4 49.3 3 M 49.4 0.858 300 mOs 0.854 24 46.0
of
Ratio of VNC phospholipid mole ratio % retention 0.107 100 0.099 92.5 0.096 89.7 0.101 94.3 0097 90.6 0.080 100 0.066 80.4 0.066 81.0 0.062 75.3 0.058 70.4
to
C. Demetzos / Stealth Liposomal Vinca Alkaloids
137
a
A tube containing 1000 ul total volume composed of 500 ul liposomal VNC and 500 ul MES 20 mM pH=5.4 NaCl 150 mM 300 mOs or 500 ul HEPES 20 mM pH=7.2 290 mOs NaCl 144 mM was incubated for 24 h at 37 °C. Aliquots (200 ul) were withdrawn at various time periods. Free VNC was separated from encapsulated by gel filtration (G-75 Sephadex). Table 6. Vinorelbine (VNR) retention in 50% rat plasma at 37 °Ca of Time Concentration of VNR Concentration External phospholipid (umole/ml) encapsulated (ug/ml) (hours) solution 50% rat plasma in HEPES pH7.2
0 1 3 6 24
70.0 66.0 64.0 61.6 58.1
0.974 0.976 0.974 0.960 0.956
Ratio of VNR to phospholipid mole ratio % retention 0.066 100 0.062 94.9 0.060 92.2 0.059 90.1 0.056 85.3
a
A tube containigl000 ul total volume of 500 ul liposomal VNR in HEPES 20 mM pH = 7.2 NaCl 144 mM 290 mOs and 500 ul rat plasma was incubated at water bath at. 37 °C for 24 h. Aliquots (200 ul) were withdrawn at various time periods. Free VNR was separated from the encapsulated by gel filtration (G-75 Sepharose). Table 7. Vincristine (VNC) retention in 50% rat plasma at 37 °C a of Ratio Concentration of Concentration Time (hours) of VNC to External VNC encapsulated phospholipid phospholipid solution (ug/ml) (umole/ml) mole ratio % retention 0.852 0.097 100 77.0 0 50% 1 76.4 0.780 0.106 109 rat plasma 0.0802 3 69.3 0.093 95.8 in HEPES 0.772 57.9 0.081 83.5 6 pH 7.2 24 62.0 0.085 87.6 0.786 3
A tube containig l000 ul total volume of 500 ul liposomal VNC in HEPES 20 mM pH = 7.2 NaCl 144 mM 290 mOs and 500 ul rat plasma was incubated at water bath at 37 °C for 24 h. Aliquots (200 ul) were withdrawn at various time periods. Free VNC was separated from the encapsulated by gel filtration (Sepharose 4B). Table 8. Thermosensitivity of Vinorelbine (VNR) liposomes in HEPES buffer and 50% rat plasmaa External Temperature Time Concentration of VNR Concentration of Ratio of VNR to phospholipid solution (min) encapsulated (ug/ml) phospholipid (°C) (umole/ml) mole ratio %retention 55 HEPES 0 38.0 0.610 0.057 100 0.884 55 20 mM 30 58.0 0.060 105 pH=7.2 55 60 49.0 0.856 0.053 93.0 45 0.610 290 mOs 0 38.0 0.057 100 45 30 34.0 0.578 0.045 95.6 45 60 42.5 0.758 0.051 91.1 0.974 50% 45 0 70.0 0.066 100 rat 45 0.682 30 43.2 0.058 88.9 plasma 45 60 0.564 31.3 0.051 77.9 11
Thermosensitivity of VNR was examined using 300 ul liposomal VNR mixed with 300 ul HEPES 20 mM pH = 7.2 290 mOs. The mixture was incubated at 45 °C and at 55 °C for 60 min. 50% rat plasma (300 ul) in mixture with 300 ul liposomal VNR was incubated for 60 min at 45 °C. Aliquots for all the above mixtures were withdrawn at various time periods. Free VNR was separated from the encapsulated in all cases by gel filtration (Sepharose 4B).
C. Demetzos / Stealth Liposomal Vinca Alkaloids
138
Table 9. Thermosensitivity of VNC liposomes in HEPES buffer and 50% rat plasma * External solution
HEPES 20 mM 290 mOs pH=7.2 50% rat plasma
Temperature (°C)
Time (min)
55 55 55 45 45 45 45 45 45
0 30 60 0 30 60 0 30 60
Concentration of VNC encapsulated (ug/ml) 65.5 59.5 73.6 65.5 73.0 59.4 77.0 54.8 50.3
Concentration phospholipid (umole/ml) 0.660 0.582 0.744 0.660 0.800 0.582 0.852 0.622 0.790
of
Ratio of VNC to phospholipid mole ratio %retention 0.107 100 0.110 102 0.107 100 0.107 100 0.098 91.5 0.110 102 0.097 100 0.095 97.9 0.069 71.7
a Thermosensitivity of liposomal VNC was examined using 300 ul liposomal VNC mixed with 300 ul HEPES 20 mM NaCl 144 mM pH = 7.2 290 mOs. The mixture was incubated at 45 °C and at 55 °C for 60 min. 50% rat plasma (300 ul) in mixture with 300 ul liposomal VNC in HEPES 20 mM NaCl 144 mM pH = 7.2 290 mOs was incubated for 60 min at 45 °C. Aliquots for all the above mixtures were withdrawn at various time periods. Free VNC was separated from the encapsulated in all cases by gel filtration (Sepharose 4B).
Discussion Vinorelbine (VNR> is an important anticancer drug approved by the FDA as a single agent for NSCLC. There is now extensive data confirming the activity of VNR for metastatic breast cancer. Vinorelbine was incorporated into liposomes for the first time. The purpose of this study was to formulate liposomes, using buffers with different pH and rat plasma at different temperatures. Vinorelbine is a lipophilic weak base and this study investigated the parameters of VNR loading using different kind of salts as internal liposome solution and different kind of buffers as external solution. Vinorelbine was encapsulated in DSPC/CH/PEGPE vesicles using pH gradient-dependent entrapment procedure, under which the lipophilic cations are moved across a lipid membrane due to a pH gradient. In the case of EPC/CH vesicles using also pH gradient method, encapsulated VNR was decreased due to the 'fluid' composition of liposomes. The rate of release of liposomal VNR is important because rapid release of the drug from the vesicle carrier may obviate any benefits provided by long-circulating liposomal system. We therefore investigated at different buffers and at different temperatures the drug release as well as at 50% rat plasma. Thus the retention for VNR was found to be higher in HEPES 20 mM pH = 7.2 290 mOs and in 50% rat plasma, than in MES 20 mM pH = 5.4 300 mOs buffer. We have found that elevated temperatures did not affect the retention when external solution was HEPES buffer, but were required to obtain efficient release of the drug when the external medium was 50 % rat plasma. Vincristine is a weak lipophilic base. It is potent antineoplastic agent and has found that liposomal Vincristine is more potent than free drug, and is also less toxic. We used VNC results obtained from the literature in order to compare our results for liposomal VNR. VNR incorporation into liposomes (DSPC/CH/PEGPE) was as high as in VNC. Drug release was higher using MES 20 mM pH = 5.4 300 mOs buffer as external solution in both cases, much higher with VNR than VNC. The drug retention at 55 °C and 45 °C with external solution HEPES 20 mM pH = 7.2 290 mOs was high in both cases (VNR, VNC ). We obtained similar results for the drug release with 50% rat plasma as an external solution. For the comparison we used the Tables 4 , 6 and 8 (Figures 2, 3, 4) (VNR and Tables 5,7 and 9 (VNC) (Figures 5 and 6). Figure 7 uses data from Tables 6 and 7.
C. Demetzos / Stealth Liposomal Vinca Alkaloids
T jf
g.
n
—~~
•
1
Sf~<>
1\ i J.T
oTX
139
0 —- HEPHS BUFt-bK O ~O
RAT PLASMA 50% VffiS O UPPER
V
£>.-. -r -t- ~ " O - . , _
i
i"
,—>—~— ~ —,
TIME (HOURS) Figure 2: Release kinetics of encapsulated VNR in HEPES 20 mM pH=7,2, 290 mOs, in MES 20 mM pH=5,4, 300 mO and in 50% rat plasma at 37 ° C. Data obtained from Tables 4 and 6.
—D—
HEPES buffer (55°C)
—0—
HEPES buffer (45°C)
—-O— - RAT PLASMA 50% (4$°C)
20
40
60
80
TIME (MINUTE) Figure 3: Release kinetics of encapsulated VNR in HEPES 20 mM pH=7,2, 290 mOs at 55 ° C 45 ° C and in 50% rat plasma at 45 °C. Data from Table 8.
C. Demetzos / Stealth Liposomal Vinca Alkaloids
140
E3
VINORELBINE (VNR)
E3
VINCRIST1NE (VNC)
TIME (HOURS) *
Figure 4: Comparison of release kinetics of VNR and VNC in HEPES 20 mM pH=7,2, 290 mOs at 37 °C. Data obtained from Tables 4 and 6.
o, d
O
HEPES BUFFER
O
RAT PLASMA 5O%
----O - -
MES BUFFER
TIME (HOURS)
Figure 5: Release kinetics of encapsulated VNC in HEPES 20 mM pH=7,2, 290 mOs, in 50% rat plasma and in MES 20 mM pH=5.4, 300 m O s at 37 °C. Data obtained from Tables 5 and 7.
0
HEPES buffer 155 C!
o -
IIEPES buffer c 45t)
-o---- RAT PLASMA 50^(
,T
'O
i
i'lME 'MINI.1I
Figure 6: Release of encapsulated VNC in HEPES 20 mM pH=7,2, 290 mOs at 45 °C and in 50 % rat plasma at 45 ° C. Data obtained from Table 9.
C. Demetzos / Stealth Liposomal Vinca Alkaloids
141
VTNORELBINE (VNR) VINCRISTINE (VNC)
0
I
3
6
24
30
TIME (HOURS)
Figure 7: Comparison of release kinetics of VNR and VNC in rat plasma 50% at 37 ° C. Data obtained from Tables 6 and 7.
Further examination of the therapeutic activity as well as toxicological and pharmacological behavior of liposomal VNR will be needed in order to compare the therapeutic efficacy of liposomal system to the free VNR. Acknowledgements This study was funded by grants from the SPORE project of the University of California San Francisco.
[1] E. K. Rowinsky and R. C. Donehower, Pharmacol Ther. 52 (1991) 35. [2] D. Rahmani-Jourdheuil, F. Coloma, M. Placidi and R. Rahmani, J. Pharm. Sc. 83 (1994) 468. [3] R. S. Kaplan and P. H. Wiernik, Sew. Ongology 9 (1982) 103. [4] V. D. Jackson, H. B. Wells and J. N. Atkins, Amm. J. Med. 84 (1988) 1016. [5] E. Hamel and C. M. Lin, Arch. Biochem. Biophys. 209 (1981) 29. [6] P. Magnus and L. S. Thurston, J. Org. Chem. 56 (1991) 1166. [7] J. S. Lee, The Cancer Bulletin 42 (1990) 426. [8] C. A. Coltman, Seminar in Ongology. 21 (1994) 1. [9] L. Canobbio, F. Boccardo, and D. Guarneni, Eur. J. Cancer. 27 (1991) 804. [ 10] G. A Smith, Ongology. 9 (1995) 767. [11] D. Pathak, T. M. Allen, A. Gabizon, E. Mayhew, K. Matthay, S. K. Huang, K. D. Lee, M. C. Woodle, D. D. Lasic, C. Redemann and F. G. Martin, Proc. Natl. Acad. Sci. (USA) 88 (1991) 11460. [12] L. D. Mayer, M. B. Bally, H. Loughrey, D. Masin and P. R. Gullis, Cancer Res. 50 (1990) 575. [13] M. B. Bally, R. Naya, D. Masin, M . J. Hope, P. R. Guiles and L. D. Mayer. Biochim.Biophys. Acta 1023 (1990)133. [14] L. D. Mayer, L. C. L. Tai, M. B. Bally, G. N. Mitilenes, R. S. Ginsberg and P. R. Gullis, BBA. 1025 (1990)143. [15] A. Rahman, A. Kessler, N. More, B. Sikic, G. Rowden, P. Wooley and P. S. Schein, Cancer Res. 40 (1980)1532. [16] S. W. Murran, J. Liposome Res. 5(3) (1995) 523. [17] D. Papahadjopoulos, D. Allen and T. Gabizon, Proc. Natl. Acad. Sci. (USA) 88 (1991) 11460. [18] F. C. Szoka and F. Olson, Biochim. Biophys. Acta 601 (1980) 559. [19] G. Haran, R. Cohen, L. K. Bar and Y. Barenholz, Biochim. Biophys. Acta 1151 (1993), 201.
142
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Structural Elucidation, Conformational Properties and Effects in Membrane Bilayers of the Toxin Paralysin, ß-Ala-Tyr from Larvae of the Gray Flesh Fly I. Kyrikou.1.2 , T. Mavromoustakos , C. Poulos 'institute of Organic and Pharmaceutical Chemistry, Vas. Constantinou 48, Athens 11635 2 University of Patras, Department of Chemistry, Patras 26500, Greece Abstract. Larval extracts of the homometabolous insects (i.e.Neobelleria Bullata -Insecta Diptera), cause immediate paralysis, followed by death when injected into adult flesh flies. These extracts contain endogenous toxins (which are widely spread over the class of insects), called paralysins, and are present throughout all the development stages, with their concentration gradually increasing from larvae stage over pupation to late pharate adults. The above experimental observations indicate that paralysins have an active role in the metamorphosis. Due to the pharmacological importance of paralysins the stereo electronic properties responsible for their activity have been studied. In particular the known paralysin beta – Ala – Tyr was synthesized and its structure was elucidated using 1D and 2D NMR techniques. Its conformational properties were studied using a combination of 2D NOESY spectroscopy and molecular modeling. The thermotropic properties were also studied using solid - state NMR spectroscopy.
Introduction Acidic methanolic extracts of larvae obtained from nine different species were found to contain substances that cause a lethal effect in the adult stage of the same species and of other species. The compounds responsible for their lethal effects in adult are called paralysins. Two paralysins soluble in organic solvents and heat stable, were chromatographically purified to homogeneity by Shean-Jaw Chiou et al.[l] and they were identified by use of mass spectrometry and nuclear magnetic resonance respectively as ß-
/. Kyrikou et al. / Toxin Paralysin, ß-Ala-Tyr from Larvae of the Gray Flesh Fly
143
alanine - tyrosine (ß - Ala - Tyr) and as 3-hydroxy-kynurenine (3-HK). The quantities of ßAla-Tyr and 3HK in the insect appear to increase steadily during larval development with peak values prior to the pupal stage [2]. ß -Ala - Tyr was reported by Levenbook in 1969 [3] (Fig. 1), to be synthesized in the fat body and to accumulate in the larval haemolymph up to the moment of the formation of the white puparium. After that, its concentration drops to almost undetectable levels, because hydrolases from the fat body degrade the dipeptide into the resulting amino acids The exact role of free ß-Ala or of any of the ß-Ala containing derivatives is unknown. It is believed that ß-alanylation of the neurotransmitters octapamine, tyramine or dopamine leads to storage forms or inactivation products. Their site of action probably is on the nervous system since they cause paralysis and they may also interfere on the bioelectric activity of a yet unknown neurotransmitter receptor. This study aims to explore the stereo electronic properties responsible for the activity of ßAla – Tyr and also its dynamic properties in membrane bilayers, using solid - state NMR spectroscopy, due to its pharmacological importance. Its structure was elucidated using ID and 2D NMR techniques and the conformational properties were studied using a combination of 2D NOESY spectroscopy and molecular modeling. In addition its dynamic properties in phospholipid bilayers were studied using high-resolution 13C-MAS, in an attempt to investigate its physicochemical interactions with membrane bilayers.
©
1
'-to
ß-Ala-Tyr
14 15 16 CH2CH2(CH2)i0CH2CH2CH3
CH2CH2(CH2)10eH2CH2CH3 3
14 15 16
DPPC
Figure 1: Molecular structures of beta-alanyl-tyrosine and DPPC.
Materials and Methods Materials: DMSO (99,95%) ampoules were purchased form Merck (Darmstadt, F.R. Germany) and ultra - precision NMR tubes from Peypin (France). Dipalmitoyl-glycero-sn3-phosphorylcholine (DPPC) was obtained from Avanti Polar Lipids Inc., AL, USA.
144
I.
Kyrikou et al. / Toxin Paralysin, ß-Ala-Tyr from Larvae of the Gray Flesh Fly
Solid State NMR Spectroscopy: High-resolution NMR spectra were obtained at 100.63 MHz on a Bruker MSL 400 NMR spectrometer capable of high-power 'H-decoupling and equipped with magic angle spinning unit. The spinning rate for MAS NMR experiments used was 2.5 kHz, and was kept constant for the temperatures 25–43 °C. These temperatures cover all mesomorphic states of DPPC bilayers. Each spectrum was an accumulation of 1000. The lipid content for the two samples used in the experiments was the same (~40mg). The delay time was 6s, the 90° pulse width was 5 us and the acquisition time was 40 ms. Chemical shifts values of DPPC bilayers were obtained from literature [4]. Liquid NMR Spectroscopy: The high-resolution spectra were obtained using Bruker 300 AC instrument. All data were collected using pulse sequences and phase cycling routines provided in the Bruker library of pulse programs. Data processing, including sine-bell apodization, Fourier transformation, phasing, symmetrization, and plotting were performed using Bruker software packages. !H NMR spectra were recorded using the following acquisition parameters: pulse width (PW) 3.0 us, spectral width (SW) 2513 Hz, data size (TD) 32K, recycling delay (RD) 1.0 s, number of transients (NS) 16, and digital resolution 0. 076 Hz pt-1. 13C NMR spectra were performed with PW 2.1 us, SW 20000 Hz, TD 8K, RD 2.5 us, NS 24576 and digital resolution 2.441 Hzpt-1. The !H - 1H correlation Spectroscopy (COSY) was recorded using the following acquisition parameters: recycling delay (Dl)l s, DO increment 3 us, spectral width in F2 2487.56 Hz and in F1 1243.78 Hz. The data sizes were 1K and 2K in FI and F2, respectively, and the data were zero-filled in FI prior to 2D Fourier transformation to yield a 2 x 2K data matrix. The spectrum was processed using sine-bell window function both in F1 and F2 (WDW=S) and the data were symmetrized about the diagonal (SYM). Two - dimensional (2D) phase - sensitive 1H - 1H nuclear Overhauser enhancement (NOESYPH) spectra were recorded using the following acquisition parameters: Dl = 1s, D0 = 3us, SW in F2=4761.9 Hz and F1=2380.95 Hz. Several mixing times (0.20 - 1.0 s) were used in NOESYPH experiment, of which 0.35 s gave the most prominent NOE results. Digital resolution was 2.0 Hz pt-1 in both directions. Molecular modeling: Computer calculations were performed on a Silicon Graphics O2 workstation using Quanta 97 version of the Molecular Simulation Incorporated (MSI) program. The conformational energy of the dipeptide ß - Ala - Tyr (Fig 1) was first minimized and then subjected to molecular dynamics experiments to explore its lower energy conformers, with the use of NOE constraints [6]. The molecular dynamics calculations were run for the molecule using simulation time 6 ps at a temperature of 2000 K and a dielectric constant (£=45) that simulates the DMSO environment. Family structures were generated using the dihedral angle criterion. The lowest energy conformers from each family were considered as the representatives ones. Results and discussion Structural identification of ß- Ala - Tyr: Table 1 shows the chemical shift values, for the 1 H-NMR spectrum of the molecule in DMSO solvent. Observed peaks are referenced to tetramethylsilane (TMS). The peak identification was confirmed by integration of the peaks, a 2D COSY spectrum and 2D NOESY. In the same table I3C-NMR assignment is shown. Such an assignment for the dipeptide is necessary in order to assign the solid-state spectra of phospholipid bilayers containing the dipeptide.
/. Kyrikou et al. / Toxin Paralysin, ß-Ala-Tyr from Larvae of the Gray Flesh Fly
145
Table 1: 'H-NMR and nC-NMR Chemical shifts in DMSO for ß-Ala-Tyr Assignment H-5 H-8, H-11 H-9, H-10 H-7 H-14, H-3 H-15 H-4 H-l, H-2
•
Chemical Shifts DMSO D2O (literature) 8.16 7.02 7.17 6.63 6.86 4.06 4.63 3.36 3.17 3.05 2.64 3.01 2.94 2.64 2.64 2.23 3.16
Assignment
c-r C-2' C-3' C-4' C-5' C-6' C-7'
c-8',c-ir C9',C-10' C-12'
Chemical Shifts DMSO D2O (literature) 31.7 33.64 35.6 36.46 169.97 171.9 54.5 56.90 225.25 36.69 36.0 129.72 128.6 129.89 130.6 114.84 115.5 155.54 154.5
Chemical Shifts DPPC bilayers (25°C) * 36.37 170.01 57.36 37.22 130.16 131.13 116.08 154.91
hidden in the DPPC (CH2)10 peak
Conformational properties of ß- Ala - Tyr: The energy of the dipeptide was first minimized using the algorithm of Steepest Descents (SD) to approach its local minimum and then using the Newton Raphson (NR) method to reach the local minimum. The minimized structure was used as the starting conformer in the dynamics experiments [5]. From the observed NOEs the Hl0 - H4, H9 - H4, are critical for the conformational properties of the molecule. They establish a spatial proximity between the alkyl chain and the phenyl ring. The observed NOEs were used in the dynamics experiments as distance constraints. During the molecular dynamics procedure the molecule was heated at a temperature of 2000 K, and then was equilibrated at the final temperature obtained during heating. Finally, the equilibrated structure was simulated and a dynamics trajectory was produced. Using the dihedral angle criterion family structures were generated for each procedure [10]. From the RMSD map generated the cluster threshold used was 54.574 and included two families. The values of the flexible dihedral angles of the lowest energy conformer derived from the family are represented in Fig. 2 T, = -178.40° T2= 6.28° T3 = 3.40° T4= 70.21°
Figure 2: The lowest energy conformer obtained from the molecular dynamics procedure simulating DMSO environment with the use of NOE constraints.
C - NMR spectroscopy: To gain more information about the structure of the membrane bilayers and the effect of the dipeptide when inserted into DPPC (Fig. 1) bilayers solid state 13C-NMR spectroscopy was applied [7]. Each spectrum consists of two regions, namely, carbon atoms in the hydrophobic region (10–40 ppm) and those in the glycerol backbone region (40–80 ppm), and esterified carbonyls (near 170 ppm).
146
/. Kyrikou et al. I Toxin Paralysin, ß-Ala-Tyr from Larvae of the Gray Flesh Fly
When a molecule is present in the phospholipid bilayers it can cause the following modifications to the I3C-NMR spectrum, (a) Changes in peak intensity and line-width due to the modified membrane fluidity, (b) changes in chemical shift values of individual carbon nuclei of the membrane lipid due to modified phase transition profiles, (c) appearance of a specific subset of peaks from carbon nuclei of the incorporated peptide. When the bilayer is below the phase transition (gel phase) the carbonyl signal is broad and asymmetric. Above the phase transition, where the fluidity of the membrane is increased this peak becomes narrower. Other peaks also above the phase transition become narrower and additional peaks are resolved. Above the main phase transition temperature several changes are observed not only in the lineshape of the peaks but also in their chemical shift. For examples, an upfield shift of the (CH2)n peaks is observed which reflects an increase in the acryl chains population of gauche conformations [8]. High -Resolution I3C-NMR spectroscopy: Representative I3C/MAS-NMR spectra for the preparations under study are shown for the hydrophobic, polar and backbone regions (Fig. 3) and esterified carbonyl regions (Fig. 4). The preparations containing the dipeptide show additional peaks (labeled on the top of the peaks) due to its presence in the membrane bilayers. DPPC
DPPC /p-Ab-Tyr
T= 43«C
T=40iC
T=38«C
T=36«C
T=33«C
T=30"C
T=25«C
Figure 3: The hydrophobic, polar and backbone regions of Ala-Tyr bilayers in the temperature range 25–43°C.
I3
C-MAS spectra of DPPC, DPPC/(x=0.20)
147
/. Kyrikou et al. / Toxin Paralysin, ß-Ala-Tyr from Larvae of the Gray Flesh Fly
DPPC
DPPC/ß-Ala-Tyr
T=43°C
T=40°C
T= 38 °C ^
T=36°C
T=33°C
= 30°C
j ;. w/A^ 1 r^ 160
.'60
Figure 4: Esterified carbonyl regions of temperature range of 25-43°C.
140
120
100
ppm C-MAS spectra of DPPC, DPPC/(x=0.20) DPPC bilayers in the
13
For each temperature the chemical shift values of -CH3, C-15', C-3', (CH2)10, -N(CH3)3 and C-11in the DPPC bilayers with and without the dipeptide were measured and compared in Fig. 5. For DPPC the chemical shifts of all carbons show a consistent upfield shift taking place at 33°C, coinciding with the pretransition temperature. This interpretation confirms results obtained by other authors [9]. The incorporation of the dipeptide does not affect the pretransition temperature.
148
/. Kyrikou et al. / Toxin Paralysin. ß-Ala-Tyr from Larvae of the Gray Flesh Fly
DPPC DPPC/ p-Ala-Tyr CH-,
C-15
C-3
CO OS
'
__
(CH 2 )iO
N(CH 3 ) 3
C-ll
Temperature Figure 5: Chemical shift changes versus temperature of the hydrophobic. polar and backbone carbon peaks over the temperature range 25 - 43°C for DPPC and DPPC / (J-Ala-Tyr bilayers.
/. Kyrikou et al. / Toxin Paralysin, B-Ala-Tyr from Larvae of the Gray Flesh Fly
149
The chemical shift changes versus temperature plots reveal various other observations, (a) In the gel phase the presence of drug molecule causes upfiled shift in the headgroup interface and the (CH2)10 regions of the hydrophobic region, It does not affect the terminal group and the C-15' (or to-1) carbon adjacent to the terminal group. These results show that the dipeptide does not reach deep into the center of the bilayers in the gel phase. (b) The presence of the drug causes broadening in the hydrophobic core (CH2)10 and C-3' regions, biphasic effect in the polar region (N(CH3)3, C-11') and almost no effect in the deep hydrophobic region (CH3, C=15'); (c) The drug causes upfield and downfield effects showing that causes simultaneously stabilizing and destabilizing effects in a bilayers that exists in the liquid crystalline phase. Only C-15' is not affected in the liquid crystalline phase with the presence of the drug. Its incorporation is very sensitive with temperature and in the head-group and terminal methyl-group a biphasic effect was observed (upfield at 38 °C and downfield at 43°C). This may interpreted that the dipeptide is more flexible in the liquid-crystalline phase with no fixed topography. References [1] Chiou S-J, Kotanen S., Cerstianes A, Daloze D., Pasteels J., Lesage A, Drifhout W. Verhaer P., Dillen L., Clayes M., De Meulemeester H., Nuttin B., De Loof A., Schoofs L., Biochem. Biophys. Res. Comm., 246 (1998) 457. [2] Chiou S-J, Cerstiaens A., Kotanen S.P., De Loof A., Schoofs L., J. Insect Phys., 44 (1998) 405. [3] Levenbook L., Bodnaryk R.P. and Spande T. F. Biochem. J., (1969) 837. [4] Wen-guey Wu, Lang-Ming Chi, Biochimica. et Biophysica. Acta, 1026 (1990) 225. [5] T. Mavromoustakos, M. Zervou, D. Panagiotopoulos, E. Theodoropoulou, J. Matsoukas, D. Karussis, J. Pharma. Biomed. Anal. 16 (1998) 741. [6] T. Mavromoustakos, E. Theodoropoulou, M. Zervou, T. Kourouli, D. Papahatjis., J. Pharma. Biomed. Anal. 18 (1999) 947. [7] T. Mavromoustakos, E. Theodoropoulou, Chemistry and Physics of Lipids, 92 (1998) 37. [8] T. Mavromoustakos, I. Daliani, Biochimica. et Biophysica. Acta, 1420 (1999) 252. [9] T. Mavromoustakos, E. Theodoropoulou and De-Ping Yang. Biochimica. et Biophvsica. Acta, 1328 (1997) 65. [10] Leach A.R. (1998), Molecular Modeling (Addison Wesley Longman Ltd, London), 1-11
150
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Effects of NSAIDs in Membrane Bilayers T. Mavromoustakos1, I. Kyrikou1, A. Kapou1 and D. Kovala-Demertzi2 1
Institute of Organic and Pharmaceutical Chemistry, Vas. Constantinou 48, Athens 11635 2 University of Ioannina, Chemistry Department, loannina 45110
Abstract. The thermal effects of non-steroidal antiinflammatory drugs (NSAIs), aspirin, piroxicam and tenoxicam, in DPPC bilayers without and with cholesterol are studied. All drugs affected the pre- and main transition temperatures of DPPC bilayers without and with cholesterol. Their effects can be summarized to the lowering of the phase transitions and causing an increase of the breadth of the transitions at low concentrations. At higher concentrations their effect is augmented and the pretransition for most of DPPC/NSAID bilayers is abolished. The effect of pH did not appear to modify significantly the thermal effects of these drugs.
Introduction Aspirin, or acetylsacylic acid, is synonymous for first aid relief of pain, fever, and inflammation. The 100-years old and most popular drug is facing new competition. The unique strength of acetylsacicylic acid is its nonselective behavior and lack of specificity for a single target. It is the portfolio of different properties that makes acetylsacylic acid such a unique drug. The therapeutic effect of acetylsacylic acid is based on a covalent modification of cyclooxygenase and the inhibition of the first step of prostaglandin synthesis. Recently, two different cyclooxygenase isoforms have been characterized COX-1 and COX-2. Inhibition of COX-1 or COX-2 leads to very different pharmacological effects. The COX-1 inhibition is predominantly responsible for anti-thrombotic effects, while antiinflammatory effects are mediated mainly through COX-2. New studies from the last two years revealed that in addition to arthritis and pain, cancer and neurodegenerative diseases like Alzheimer's disease could potentially be treated with COX-2 inhibitors. It has been suggested that the anti-inflammatory and most of the analgetic effects of NSAIDs result from an inhibition of PGs synthesized via COX-2 [1-4]. It was reported in the literature that acetylsalicylic acid (ASA) was 5-10 fold more potent to reduce fever when was intragastrically administered in a lipid-associated state. In another report it was shown that the half-life of ASA was prolonged when the agent was chemically associated with DMPC because of its reducing conversion to salicylic acid [5]. For the above reasons the ASA membrane interactions were studied using fluorescence polarization and NMR spectroscopy. The fluorescence polarization results showed that ASA causes disordering of the membrane bilayers in a concentration depending fashion and lowering of AH. The NMR data showed that ASA was not inserted into membrane bilayers
T. Mavromoustakos et al. / Effects of NSAIDs in Membrane Bilayers
151
and aromatic rings interacted with membrane bilayers. In addition, the mobility of acetylsalicylic acid was restricted by membrane and the ASA changed membrane viscosity. It was proposed that the association of ASA with membrane might involve both electrostatic and hydrophobic forces. In another study it was shown that aspirin, benzoate and salicylate caused significant increases in membrane conductance [6–8]. These results triggered our research interest to apply DSC and molecular modeling in order to further study the effects of ASA with membrane bilayers and to compare its thermal effects with NSAIDs piroxicam and tenoxicam shown in Figure 1. Piroxicam, [4-hydroxy-2-methyl-N-pyridin-2-yl)-2H-1,2-benzothiazine-3carboxamide-1,1-dioxide], H2pir, is a potent and extensively used non-steroidal antiinflammatory, anti-arthritic drug with a long biological half-life. To date, piroxicam is among the top ten NSAIDs in the market. Tenoxicam [4-hydroxy-2,methyl-N-2-pyridyl2H-thieno(2,3-e)-l,2-thiazine-3-carboxamide-1,1-dioxide], is a new non-steroidal drug which has anti-inflammatory, analgetic and antipyretic effects: The drug is widely used in the treatment of rheumatic diseases. It is a derivative of oxicam with a thiophene ring replacing the benzene ring in piroxicam. Both piroxicam and tenoxican are the most famous members of this group and are nonselective inhibitors like aspirin We have developed an interest in NSAIDs, the co-ordination chemistry and anti-inflammatory properties of NSAIDs with transition and non-transition metal ions [9,10]. DPPC bilayers were used for these studies because its thermal effects and dynamic properties are well studied. In addition, the incorporation of various drugs in the spontaneously phospholipid bilayers formed after hydration have been studied extensively in an attempt to understand the molecular interactions between drugs with phospholipids. The presence of an additive in membrane bilayers affects the thermodynamic parameters that govern a thermogram such as the maximum of the main phase transition or the pretransition (Tm), the heat capacity of the peaks (Cp) and the line-width (Tm1/2) [8–11]. The nature of the DSC thermograms can be understood if the total intermolecular effects (i.e. interfacial, hydrogen bonds and nonspecific hydrophobic and electrostatic interactions) between the additive and the phospholipid bilayers are considered. Membrane preparations were formed when the drug was in a neutral and acidic environment (pH=2.5) in order to explore any differential effect of anionic or non- anionic forms of the molecules in membrane bilayers. The effect of the cholesterol in membrane bilayers containing NSAID was also studied. Cholesterol is a major constituent of membrane bilayers and DPPC/cholesterol bilayers is often more distinctive system to differentiate the thermal properties of drugs. [12].
Aspirin
Piroxicam
152
T. Mavromoustakos et al. / Effects of NSAIDs in Membrane Bilayers
OH
O
Tenoxicam Figure 1: Chemical structures of NSAIDs
Results Differential Scanning Calorimetry: Differential scanning Calorimetry (DSC) is a fast and relatively inexpensive technique that allows the study of the thermotropic properties of the membranes in the absence and presence of bioactive molecules. Therefore, it is used in our laboratory as a diagnostic technique to investigate differential effects that may be caused by the incorporation of additives. When such differential effects are observed, then techniques that offer complementary and more detailed information on the thermal and dynamic properties of membranes, with or without the presence of additives are applied. Figure 2 shows DSC scans of DPPC and DPPC/NSAID bilayers. The DPPC bilayers exist in the gel phase (Lß') for temperatures lower than 33 °C, and in the liquid crystalline phase for temperatues higher than 42 °C (IV). In between 33-42 °C the phospholipid bilayers exist in Pß or ripple phase. The obtained DSC scan of fully hydrated DPPC multibilayers shows a pretransition centered at 35 °C and a peak maximum at 41.2 °C. The main phase transition is accompanied by several structural changes in the lipid molecules as well as systematic alterations in the bilayer geometry, but the most prominent feature is the trans-gauche isomerization-taking place in the acyl chain conformation. The average number of gauche conformers indicates the effective fluidity, which depends not only on the temperature, but also on perturbations due to the presence of a drug molecule intercalating between the lipids. At low concentration of x=0.01 (1%-molar ratio) all NSAEDs affect in a similar way the thermal properties of DPPC bilayers, by causing marginal broadening of the pretransition and they cause narrowing of the breadth of the phase transition. The addition of higher concentration of NSAID (x=0.05) augments their thermal effects. In particular, at this concentration aspirin abolishes the pre-transition temperature and causes broadening of the phase transition temperature, effects not observed by piroxicam and tenoxicam. Only piroxicam at this concentration causes significant lowering of the phase transition temperature. At x=0.10 aspirin causes further broadening of the phase transition temperature. Tenoxicam affects only the pretransition by broadening it and piroxicam causes broadening both to the pre- and main transition temperatures. At x=0.20 aspirin does cause a lowering of the phase transition and no further broadening of the phase transition. In contrary, tenoxicam causes a significant broadening and more lowering of the phase transition as well as abolishement of pre-transition. Piroxicam also abolishes the pretransition and causes the most lowering of the phase transition among three preparations. It appears that aspirin causes the most significant effect up to x~=0.10 and lowest at the higher concentration of x=0.20. The drug molecules under study do not appear to affect the
T. Mavromoustakos et al. / Effects of NSAIDs in Membrane Bilayers
153
enthalpy change (AH) of the main phase transition of DPPC bilayers (see quantitative results on Table 1). Figure 3 shows DSC scans of the same NSAIDs on DPPC bilayers using identical concentrations and buffer (pH=2.5). Interestingly, similar results have been observed as in aqueous DPPC bilayers for piroxicam and tenoxicam. Aspirin had the most significant effect in all concentrations summarized to:(a) lowering of the phase transition; (b) increase of the breadth of the phase transition and (c) abolish of the pre-transition at lower concentration. These results show that the effect of pH is only marginal. An exception as is described occurs at x=0.20 with aspirin which causes more significant thermal effects on DPPC bilayers at pH=2.5. This probably reflects that aspirin is more active in membrane bilayers when it is in non-anionic form rather than in anionic form. Figure 4 shows the effects of NSAIDs in DPPC/cholesterol (x=0.10) bilayers. DPPC/cholesterol bilayers show a phase transition centered at 41.4 °C and no pretransition. The presence of NSAID in this bilayer results in the broadening of the width of phase transition and a slight lowering of the phase transition temperature. The most significant effect in the broadening of the phase transition is observed with aspirin. These results show that cholesterol at low concentrations does not interfere with the way the studied NSAIDs perturb the membrane bilayers. Piroxicam and tenoxicam appear to affect in an almost identical way the DPPC/cholesterol bilayers. The effects of NSAIDs were also studied in high cholesterol content. DPPC/cholesterol (x=0.15) shows a considerable broad peak. The presence of the NSAID causes inhomogeneity of the sample, since it causes significant broadeninig and additional peaks (different domains) that probably consist of DPPC/cholesterol, DPPC/NSAID or DPPC/NSABD/cholesterol.
Figure 2: DSC scans of NSAIDS 'when bilayers are formed with the use of water.
154
T. Mavromoustakos et al. / Effects of NSAlDs in Membrane Bilayers
10
"^ 2 0' 3 I0 T4 I0 ' 50 I ' 60 I 10 I ' 20 I ' 30 I ' 40I ' 501^
60
I"!
10
' I ' I ' I '
2 0 3 0
40
SO
Figure 3: DSC scans of NSAIDS when bilayers are formed with the use of buffer pH=2.5. +(0.05) + Cholesterol x=0.15
+(0.10) + Cholesterol x=0.10
DPPC
DPPC
10
20
30
40
Temperature
50
60
10
IMI 20
30
' 40M FT SO
Tempemtore
Figure 4: DSC scans of NSAIDS when bilayers are formed with the use of water and cholesterol
60
T. Mavromoustakos et al. / Effects of NSAIDs in Membrane Bilayers
155
Table 2: Values of Phase Pretransition and Transition Temperatures (Tm), Half-Width Temperature and Enthalpy Changes (AH) of DPPC with or without NSAIDS with the use of water.
samples DPPC alone DPPC + aspirin (x=0.01) DPPC + aspirin (x=0.05) DPPC + aspirin (x=0. 10) DPPC + aspirin (x=0.20) DPPC + tenoxicam (x=0.01) DPPC + tenoxicam (x=0.05) DPPC + tenoxicam (x=0.10) DPPC + tenoxicam (x=0.20) DPPC + piroxicam (x=0.01) DPPC + piroxicam (x=0.05) DPPC + piroxicam (x=0.10) DPPC + piroxicam (x=0.20)
Tml/2
Tm
AH
0.50 0.50 0.50 0.60 0.75 0.40 0.40 0.50 1. 15 0.50 0.60 0.85 0.90
42.571 41.715 41.179 40.287 39.363 41.729 41.378 41.236 38.285 41.066 39.316 38.985 36.558
41.838 41.930 41.470 40.826 39.502 42.169 41.340 41.470 43.670 42.502 42.316 41.266 42.128
Table 3: Values of Phase Pretransition and Transition Temperatures (Tm), Half-Width Temperature and Enthalpy Changes (AH) of DPPC with or without NSAIDS with the use of buffer.
samples DPPC alone DPPC + aspirin (x=0.01) DPPC + aspirin (x=0.05) DPPC + aspirin (x=0. 10) DPPC + aspirin (x=0.20) DPPC + tenoxicam (x=0.01) DPPC + tenoxicam (x=0.05) DPPC + tenoxicam (x=0.10) DPPC + tenoxicam (x=0.20) DPPC + piroxicam (x=0.01) DPPC + piroxicam (x=0.05) DPPC + piroxicam (x=0.10) DPPC + piroxicam (x=0.20)
T m1/2
Tm
AH
0.65 0.60 1.00 1.35 1.40 0.50 0.50 0.70 1.20 0.55 0.60 0.70 0.90
42.20 42.94 41.69 40.63 36.53 42.04 41.82 41.24 38.82 42.87 41.69 40.85 39.85
42.09 39.95 40.60 41.44 43.62 41.63 41.20 41.54 41.00 40.84 42.31 41.07 40.87
Table 4: Values of Phase Pretransition and Transition Temperatures (Tm), Half-Width Temperature and Enthalpy Changes (AH) of DPPC / CHOL with or without Compound with the use of water.
samples DPPC/CHOL (90: 10) alone DPPC/CHOL (90:10) + 1 (x=0.10) DPPC/CHOL (90:10) + 2 (x=0. 10) DPPC/CHOL (90:10) + 3 (x=0.10) DPPC/CHOL (85:15) alone DPPC/CHOL (85: 15) + 1 (x=0.05) DPPC/CHOL (85:15) + 2 (x=0.05) DPPC/CHOL (85:15) + 3 (x=0.05)
Tml/2
Tm
0.50 0.70 0.45 0.55 0.70 2.95 7.60 6.90
41.37 40.49 40.14 40.49 41.74 40.02 32.538 32.54
AH 32.28 29.30 28.15 26.25 18.33 14.50 3.61 3.59
156
T. Mavromoustakos et al. / Effects of NSAIDs in Membrane Bilayers
Discussion According to the thermograms the three drugs present a similar thermotropic behaviour inside the membrane model. Theoretical calculations confirm that their topography is similar. Each molecule is located near the head-group of the DPPC molecules forming two hydrogen bonds with it. Aspirin is a smaller molecule and is situated lower, towards hydrophobic region. This localization may explain the most perturbing effects in comparison to the other two NSAIDs.
Figure 5: Topography of the nsaids, (a) aspirin, (b) piroxicam, (c) tenoxicam inside the model membrane bilayer
The four oxygen atoms, the pyridyl and imine nitrogens for piroxicam and tenoxicam and the two oxygen atoms for aspirin, exhibit the maximum electron density and negative charge. The highest effective charge and the highest electron density values for the four oxygen atoms and for the pyridyl and imine nitrogen for piroxicam and tenoxicam and for the two oxygen atoms for aspirin show strong electron-donor properties and could rationalize a network of inter-, intra- hydrogen and non-hydrogen or polar bonding [10]. The localization of piroxicam and tenoxicam in the head-group region explains their significant effect on the pre-transition and only marginal effect on the main-transition. The better accommodation of tenoxicam versus piroxicam explains its lowest perturbing effects. Acknowledgement We are thankful to GSRT for the funding of this research activity through PENED 99 99ED442. We also thank HELP EPE for the generous gift of piroxicam and tenoxicam.
T. Mavromoustakos et al. / Effects of NSAIDs in Membrane Bilayers
157
References [1] Beuck, M. Nonsteroidal Antiinflammatory Drugs., Angew. Chem. Int. Ed. 38 (1999) 631. [2] Tonnessen, T.I., Aas, A.T., Sandvig, K., Olsnes, S., J. Pharmacol. Exp. Ther. 248 (1989) 1197. [3] Herchuelz, A., Derenne, F., Deger, F., Juvent, M., Ganse, Van E., Staroukine, M., Verniory, A., Boeynames, J.M., Douchamps, J., J. Pharmacol. Exp. Ther. 248 (1989) 1175. [4] Lenard, M.L., Carlos, U., Amy, L., J. Pharmacol. Exp. Ther. 277 (1996) 1221. [5] Snow, I.B., Maass, A.R., Nephron 23 (1979) 15. [5] Muhammad, J.H., Janes, A.R. J. Pharm. Pharmacol. 45 (1993) 3496. [6] Fujisawa, Kadama, T., Masuhara, E. 62 (1983) 803. [7] Li, J., Huang, H., Zhou, M., Ning, S., Jiang, X., Peng, Y., Zhao, K., Biochem. Mol. Biol. Int.. 47 (1999) 665. [8] De, S., Ghosh, A.K., Basu, R., Nandy, P., Phys. Med. Biol. 41 (1996) 383. [9] D. Kovala-Demertzi, J. Inorg. Biochem. 79 (2000) 153. [10] M. A. Demertzis, S. K. Hadjikakou, D Kovala-Demertzi, A. Koutsodimou and M. Kubicki, Helvet. Chim. Acta, (2000), 2, 787-2801; S. K. Hadjikakou, M.A. Demertzis, J. R. Miller and D. KovalaDemertzi, J. Chem. Soc. Dalton Trans, (1999) 663. [11] Gutknecht, J., Mol. Cell. Biochem. 114 (1992) 3. [12] T. Mavromoustakos, E.Theodoropoulou, D. Papahatjis, t. kourouli, De-ping Yang, M. Trumbore, A. Makryannis, Biochim. et Biophys. Acta, 1281 (1996) 235.
This page intentionally left blank
Part III.
Conformational Analysis of Bioactive Compounds
This page intentionally left blank
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
The Structure of Endotoxin-Neutralizing Peptides Bound to LPS P. Pristovsek and J. Kidric National Institute of Chemistry, Hajdrihova 19, SI-1000 Ljubljana, Slovenia
Abstract Lipopolysaccharide (LPS, endotoxin) induced Gram-negative sepsis and septic shock remain lethal in up to 60 % of cases, and LPS antagonists that neutralize its endotoxic action are the subject of intensive research. In the last decade peptidic antagonists have become increasingly important in providing leads for treatment of LPS-mediated diseases. In an effort to determine the molecular motifs of LPS specific binding by antiseptic peptides we have modeled the complexes of two proteins (LALF and BPI) with LPS based on a proposed common binding motif of LPS to proteins [Ferguson et al. (2000) Structure 8, 585] that has been extended with the hydrophobic contacts of the LPS acyl chains and non-polar protein side-chains. The models of two peptides (polymyxin B and a synthetic fragment of LALF) bound to LPS have been derived using their bound conformations as determined from transferred NOE studies, and the same extended binding motif. The proposed models can be used for design of peptides with improved antiseptic activity.
Introduction A key feature of innate immunity in mammals is the ability to limit the infectious challenges rapidly [1]. One of the major ligands recognized by the innate immune system is lipopolysaccharide (LPS, endotoxin), a common constituent of Gram-negative bacterial outer membranes. LPS is an amphiphile consisting of the lipid A, a core oligosaccharide and an outer polysaccharide composed of repeating hetero-oligosaccharide sub-units; the lipid A is a hydrophobic, lipid-rich moiety that harbors the endotoxic principle of LPS and is the most highly conserved part of the structure, typically with two glucosamines, two phosphate esters and six fatty acid chains [2] (Fig. 1). Failure to contain the bacterial infection can result in Gram-negative sepsis as a result of the release of LPS [3]. Very low concentrations of LPS (0.01 nM) are required to incite septic shock, a multiple organ dysfunction syndrome that is associated with high mortality in intensive care unit patients causing 2 million deaths in the world annually [4]. Septic shock is initiated by mediators released from host cells stimulated by LPS, of which tumor necrosis factor a (TNF-a) appears to be the most important one [5]. Antibiotics used to treat the bacterial infection can actually be harmful because they can stimulate the release of endotoxin [6]. Endotoxin
162
P. Pristovsek and J. Kidric / The Structure of Endotoxin-Neutralizing Peptides'
exerts its pathophysiological reactions by the induction of a specific cellular recognition of agonistic LPS/lipid A which is initialized by the combined extracellular actions of LPS binding protein (LBP), the membrane-bound or soluble forms of CD 14 and the newly identified Toll-like receptor 4 (TLR4)*MD-2-complex (for a recent review see [7]). Many strategies to overcome the endotoxic shock, including neutralizing antibodies, soluble cytokine receptors, various endotoxin binding factors and antibacterial agents that inhibit lipid A biosynthesis, have been tested with mixed results [8-12]. Recent developments involve synthetic antagonists structurally based on natural lipid A and those lipid A structures known to be devoid of endotoxic activity [13], and LPS-neutralizing peptides that are either derived from natural antimicrobial peptides or from proteins that bind LPS [14], [15]. The latter strategy is increasingly important despite of known difficulties with rapid degradation of peptides in the organism, and provides leads to substances that may be clinically useful in the treatment of Gram-negative bacterial sepsis and shock. Investigations of the interactions between LPS (or lipid A) and oligopeptides were motivated by the finding that polymyxin B (PmB, mo-K'TK'-cyclo-[K'K'fLK'K'T]; mo: methyl octanoate; K': diaminobutyric acid (Dab); f: D-Phe; the cycle is closed between the side-chain of Dab 4 and the main chain of Thr 10, Fig. 2), a cyclic, cationic peptide antibiotic, binds to lipid A [16] with an apparent dissociation constant in the uM range [17] and neutralizes its pathogenicity. Unfortunately, PmB is toxic and can not be used for therapy. A series of peptides designed to mimic the primary and secondary structure of PmB was synthesized and tested for binding and detoxification of LPS / lipid A [18],[19]. Other peptides that bind and detoxify LPS are derived from proteins that bind LPS, e.g. LBP [20],[21], limulus anti-LPS factor (LALF) [21],[22], bactericidal permeabilityincreasing protein (BPI) [21],[23] and heparin binding protein (HBP) [24]. A LPS-binding motif consisting of alternating series of positively charged and hydrophobic residues of the peptides forming a positively charged amphipatic loop has been put forward based on the X-ray structure of LALF [25]. The structures of two other proteins that bind LPS are also known from X-ray studies, i.e. HBP [26] and BPI [27]. However, neither the three-dimensional structures of the peptides nor the structural aspects of the lipid A-peptide interaction at the atomic level are known. The investigation of the molecular determinants of lipid A specific binding by the peptides mentioned above have been hampered by the amphiphilic character of LPS causing typical aggregate formation in water that makes structural studies very difficult. The interaction of LPS with its cognate binding proteins has been structurally elucidated in the single case of the X-ray crystallographic structure of LPS in complex with the integral outer membrane protein FhuA from Escherichia coli K-12 [28]. Four positively charged residues of FhuA were identified that appear to provide most of the important electrostatic interactions with LPS [29]. Their arrangement was proposed as a common LPS binding motif and was used to identify LPS-binding sites on proteins known to interact with LPS.
Figure 1: Chemical structure of lipid A.
Figure 2: Chemical structure of polymyxin B (PmB).
P. Pristovsek and J. Kidric / The Structure of Endotoxin-Neutralizing Peptides
163
In an effort to determine the molecular motifs of LPS/ lipid A specific binding by peptides neutralizing LPS toxicity we are investigating their complexes by NMR and molecular modeling. We have determined the LPS-bound structure of PmB with the aid of the transferred nuclear Overhauser enhancement (NOE) effect [30] and proposed a model of the complex [31]. We have employed a similar methodology with a synthetic 16-residue fragment of LALF (residues 36-47 with a synthetic disulfide bridge, GCKPTFRRLKWKYKCG, LALF-16) that neutralizes LPS in vitro [22], and determined a preliminary bound conformation [32]. In this paper we use the electrostatic motifs of binding proposed in [29] and additional hydrophobic contacts of the lipid A fatty acid chains with non-polar regions on the surface of LALF and BPI to develop models of complexes with LPS. The derived models and the conformations of PmB/LALF-16 bound to LPS determined from transferred NOE studies are used to propose models of the peptideLPS complexes which can be used for development of peptides with improved inhibition of endotoxicity. Experimental For the modeling of the LPS - LALF and LPS - BPI complexes the data on common (electrostatic) binding motifs from [29] was employed using the X-ray structures of LALF [25] and BPI [27]. The three-dimensional structure of LPS from E. coli strain K-12 (without the outer polysaccharide and the acyl chain at position 3 of the proximal glucosamine) was taken from the X-ray structure of FhuA [28]. The docking was performed using a similar procedure as described in detail elsewhere [31]. Initially the electrostatic and H-bond contacts of the protein and the two phosphate groups of LPS were set up as proposed in [29]. The model was refined using the hydrophobic contacts of the lipid A acyl chains with non-polar regions on the protein surfaces. During the optimization a maximal reduction of accessible hydrophobic surface on both molecules was sought while keeping both molecules rigid. Only in the final stage the LPS molecule was allowed to move in the energy minimization using a distance-dependent dielectric constant employed in order to simulate, at least in part, electrostatic screening by the solvent. For the modeling of the LPS - PmB and LPS - LALF-16 complexes the same LPS coordinates and the coordinates of the peptides bound to LPS determined from transferred NOE studies were used [31], [32]. The surface of the peptides was scanned for the common electrostatic motif [29] and used for initial attachment of LPS to the peptide. The model of the complex was refined using the same procedure as described above. The Insight-II/Discover program package of (MSI, 9685 Scranton Rd, San Diego, CA 92121-3752, USA) on a Silicon Graphics Indy workstation using the Consistent Valence Force Field (cvff [33]) was used for all calculations.
164
P. Pristavsek and J. Kidric / The Structure of Endotoxin-Neutralizing Peptides
Results and Discussion The common electrostatic binding motif of LPS [29] to two proteins with known X-ray structure (LALF and BPI [27]) was deduced from the only X-ray structure that involves a molecule of LPS bound to a protein [28]. Although many contacts of positively charged side-chains of the FhuA protein can be identified that bind to lipid A via H-bonds or electrostatic interactions, only four of them (Lys306 and Lys351 at 4'-phosphate, Arg382 and Lys439 at 1-phosphate) are part of the proposed common electrostatic binding motif [29]. In the models of the complexes of LPS with LALF and BPI (Fig. 3 and 4) the electrostatic and H-bond interactions to the phosphate groups come from Arg41, Arg40, Lys47 and Lys64, and Lys42, Lys48, Lys92 and Lys99, respectively. Our model was optimized using reduction of accessible hydrophobic surface on both molecules; additional hydrophobic contacts of the lipid A acyl chains with Thr38, Phe39, Leu42 and Trp44 on LALF and Tyr54, Pro35, Tyr37, Alal65 and His50 on BPI could be proposed in the present work.
Figure 3: Model of the complex of LPS (stick model) with LALF (dark surface: polar sidechains).
Figure 4: Model of the complex of LPS (stick model) with BPI (shown in part; dark surface: polar side-chains).
The bound conformation of PmB [31] and the preliminary structure of LALF-16 [32] display an amphiphilic character caused by spatial separation of polar and non-polar residues. These structures have been determined from NMR structure refinement using data from transferred NOE experiments. The latter occurs in case of weak binding of a smaller ligand (in this case: peptide) to a large molecule or assembly (in this case: LPS aggregate); the cross-relaxation in the bound species is much larger than in the free ligand and is transferred to the latter through chemical exchange. The intra-molecular NOEs of the bound ligand therefore occur at the resonance frequencies of the free ligand and give structural insight to the bound conformation of the ligand [30]. The transferred NOE, however, is measured in conditions of approx. 10:1 peptide:LPS molar ratio, and does not contain any information about the interactions with LPS. The model of the complex was therefore obtained using the bound conformation of the peptides and the coordinates of LPS [28]. The positively charged side-chains (Dab1, Dab8 and Dab9 in the case of PmB, and Lys3, Arg7, Arg8 and Lys14 in the case of LALF-16) served for the electrostatic contacts in agreement with the common motif proposed for the proteins (see above). The model of the complex was optimized using reduction of accessible hydrophobic surface on both molecules. The final models of the complexes (Fig. 5 and 6) displayed additional hydrophobic contacts of the acyl chains of LPS with Phe6, Leu7 and the octanoate aliphatic
P. Pristovsek and J. Kidric / The Structure of Endotoxin-Neutralizing Peptides
165
chain of PmB, and with Thr5, Phe6, Leu9 and Trpl1 of LALF-16, respectively. It should be noted that the non-polar contacts in LALF-16 occur to residues that correspond exactly to the ones in the full molecule of LALF. The importance of the hydrophobic contacts in the LPS-peptide complexes has been indicated by calorimetric titration studies that expose the entropic drive to complexation [34].
Figure 5: Model of the complex of LPS (stick model) with PmB (dark surface: polar sidechains),
Figure 6: Model of the complex of LPS (stick model) with LALF-16 (dark surface: polar sidechains).
Similarly to LPS bound to FhuA the core oligosaccharide and the outer polysaccharide of LPS were not involved in binding either to any of the proteins or peptides emphasizing and confirming the role of the lipid A portion for specific binding. It is known that lipid A alone possesses the endotoxic properties of LPS [2]; similar structural studies of lipid A with the peptides, however, have not been successful because of its low solubility in water. The experimental studies of antiseptic peptides in complex with LPS using NMR are difficult because of the amphiphilic nature of the peptides that tend to aggregate at concentrations necessary for measurement; additionally, conditions for the transferred NOE effect are not always attainable. In absence of any experimental data the modeled complexes of LPS and the proteins/peptides presented in this work are not final but remain a useful tool to understand the action of LPS at atomic level, demonstrating the importance and power of molecular modeling. The proposed models of PmB - LPS and LALF-16 LPS complexes will contribute to the understanding of lipopolysaccharide recognition at the molecular level and can serve in structure based anti-septic drug design. Acknowledgment We wish to thank Prof. D. Hadzi (NIC, Slovenia) for introducing us to the field. We thank A. Hoess for the coordinates of LALF. We are grateful to Prof. L. Szilagyi (Univ. of Debrecen, Hungary) for carefully reading the manuscript. This work was supported by the Ministry of Education, Science and Sport of the Republic of Slovenia. References R. Medzhitov, C. A. Janeway, Cell 91 (1997) 295. E. T. Rietschel, T. Kirikae, Schade.F.U., U. Mamat, G. Schmidt, H. Loppnow, A. J. Ulmer, U. Zahringer, U. Seydel. F. di Padova, M. Schreier, H. Brade, FASEB J. 8 (1994) 217.
166
[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] [32] [33] [34]
P. Pristovsek and J. Kidric / The Structure of Endotoxin-Neutralizing Peptides
R. J. Ulevitch, P. S. Tobias, Annu. Rev. Immunol. 13 (1995) 437. E. T. Rietschel, O. Westphal, in H. Brade, S. M. Opal, S. N. Vogel, D. C. Morrison (Eds.): Endotoxin in health and disease, Marcel Dekker, Inc., New York 1999, p. 1. J. C. Mathison, E. Wolfson, R. J. Ulevitch, J. Clin. Investig. 88 (1988) 1925. J. L. Shenep, R. P. Barton, K. A. Morgan, J. Infect. Dis. 151 (1985) 1012. C. Alexander, E. T. Rietschel, J. Endotoxin Res. 7 (2001) 167. W. J. Christ, O. Asano, A. L. C. Robidoux, M. Perez, Y. Wang. G. R. Dubuc, W. E. Gavin, L. D. Hawkins, P. D. McGuinness, M. A. Mullarkey, M. D. Lewis, Y. Kishi, T. Kawata, J. R. Bristol, J. R. Rose, D. P. Rossignol, I. Kobayashi, I. Hishinuma, A. Kimura, N. Asakawa, I. Yamatsu, Science 268 (1995)80. R. L. Greenman, R. M. Schein, M. A. Martin, R. P. Wenzel, N.R., N. R. Maclntyre, G. Emmanuel, H. Chmel, K. B. Kohler, M. McCarthy, J. Plouffe, J. A. Russell, X. S. S. group, J. A. M. A. 266 (1991) 1097. M. A. Rogy, L. L. Moldawer, H. S. Oldenburg, W. A. Thompson, W. J. Montegut, S. A. Stackpole, A. Kumar, M. A. Palladino, M. N. Marra, S. F. Lowry, Ann. Surg. 220 (1994) 77. J. Wherry, R. Wenzel, E. Abraham, R. Allred, R. Balk, R. Bone, H. Levy, S. Nasraway, T. Perl, H. Silverman, R. Wnderink, T. T. M. S. Group, Chest 104 (1993) 3S. H. R. Onishi, B. A. Pelak, L. S. Gerckens, L. L. Silver, F. M. Kahan, M.-H. Chen, A. A. Patchett, S. M. Galloway, S. A. Hyland, M. S. Anderson, R. H. Raetz, Science 274 (1996) 980. D. P. Rossignol, L. D. Hawkins, W. J. Christ, S. Kobayashi, T. Kawata, M. Lynn, I. Yamatsu, I. Kishi, in H. Brade, S. M. Opal, S. N. Vogel, D. C. Morrison (Eds.): Endotoxin in health and disease, Marcel Dekker, Inc., New York 1999, p. 699. M. Porro, in H. Brade, S. M. Opal, S. N. Vogel, D. C. Morrison (Eds.): Endotoxin in health and disease, Marcel Dekker, Inc., New York 1999, p. 403. P. Pristovsek, J. Kidric, Mini Rev. Med. Chem 1 (2001) 409. D. C. Morrison, D. M. Jacobs, Immunochem. 13 (1976) 813. S. A. David, B. Bectel, C. Annaiah, V. I. Mathan, P. Balaram, Biochim. Biophys. Acta 1212 (1994) 167. A. Rustici, M. Velucchi, R. Faggioni, M. Sironi, P. Ghezzi, S. Quataert, B. Green, M. Porro, Science 259 (1993) 361. M. T. Demitri, M. Velucchi, L. Bracci, A. Rustici, M. Porro, P. Villa, P. Ghezzi, J. Endotoxin Res. 3 (1996) 445. A. H. Taylor, G. Heavner, M. Nedelman, D. Sherris, E. Brunt, D. Knight, J. Ghrayeb, J. Biol. Chem. 270 (1995) 17934. R. J. Battafarano, P. S. Dahlberg, C. A. Ratz, J. W. Johnston, B. H. Gray, J. R. Haseman, K. H. Mayo, D. L. Dunn, Surgery 118 (1995) 318. C. Ried, C. Wahl, T. Miethke, G. Wellnhofer, C. Landgraf, J. Schneider-Mergener, A. Hoess, J Biol. Chem. 271 (1996) 28120. B. H. Gray, J. R. Haseman, Infect. Immun. 62 (1994) 2732. H. A. Pereira, I. Erdem, J. Pohl, J. K. Spitznagel, Immunol. 90 (1993) 4733. A. Hoess, S. Watson, G. R. Silber, R. Liddington, EMBO J. 12 (1993) 3351. L. F. Iversen, J. S. Kastrup, S. E. Bjorn, P. B. Rasmussen, F. C. Wiberg, H. J. Flodgaard, I. K. Larsen, Nature Struct. Biol. 4 (1997) 265. L. J. Beamer, S. F. Carroll, D. Eisenberg, Science 76 (1997) 1861. A. D. Ferguson, E. Hofmann, J. W. Coulton, K. Diederichs, W. Welte, Science 282 (1998) 2215. A. D. Ferguson, W. Welte, E. Hofmann, B. Lindner, O. Hoist, J. W. Coulton, K. Diederichs, Structure 8 (2000) 585. G. M. Clore, A. M. Gronenborn, J. Magn. Reson. 48 (1982) 402. P. Pristovsek, J. Kidric, J. Med. Chem. 42 (1999) 4606. J. Kidric, K. Feher, L. Szilagyi, P. Pristovsek, Peptides 2000 : Proceedings of the Twenty-sixth European Peptide Symposium, 24-27 August 2000 (Montpellier, France), Conformational analysis of peptides neutralising LPS pathogenicity - NMR and molecular modelling study, pp. 79 (2000). P. Dauber-Ogusthorpe. V. A. Roberts, D. J. Ogusthorpe. D. J. Wolff, M. Genest, A. T. Hagler. Proteins Struct. Funct. Genet. 4 (1988) 31. S. Srimal, N. Surolia. S. Balasubramanian, A. Surolia. Biochem. J. 315 (1996) 679.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
167
Synthesis and Conformational Analysis of Steroidal Carriers of Alkylating Agents with the Use of Two-Dimensional Nuclear Magnetic Resonance A. Kapoua, T. Mavromoustakosb, S. G. Grdadolnikc and S. Nikolaroopoulosa a b
Lab. of Pharmaceutical Chemistry, Department of Pharmacy, University of Patras, Greece Institute of Organic and Pharmaceutical Chemistry, National Hellenic Research Foundation, Greece. a National Institute of Chemistry, Hajdrihova 19, P.O.B. 30 SI-1115 Ljubljana, Slovenia
Abstract. The steroidal esters 3p-acetoxy-5-androsten-7-one, 3p-acetoxy-5-androsten-7,17dione, 3p-acetoxy-17a-aza-D-homo-5-androsten-17-one and 3p-acetoxy-I7a-azaD-homo-5-androsten-7,17-dione were synthesized. Among these synthetic compounds the ones that are chemically combined via an esteric bond with alkylating mustards such as chlorambucil exhibit significant anticancer activity and under strength toxicity as has been shown by in vitro and in vivo tests. In an effort to implicate the conformation of the steroidal moiety with the structureactivity relationships of these combinations and thus rationalize the design of new entities, ID- and 2D-NMR spectra were recorded and fully assigned.
Introduction Fighting and defeating cancer has constituted one of the major projects of physicians, biologists, medicinal chemists and other scientists during the last 50 years. Although considerable progress has been made in both diagnosis and treatment, many types of cancer remain incurable and as lethal as ever. The multiformity of this disease calls for a diversity in treatment approaches and accounts for the combined use of surgery, chemotherapy, radiation therapy and radioimmunotherapy.
168
A Kapou et al. / Synthesis and Conformational Analysis of Steroidal Carriers
A large number of anticancer drugs have been applied, some of which extremely specific for a certain type of cancer, others with multiple properties used in several types of the disease. Alkylating agents have been widely used ever since their anticancer properties were recognized just after World War I, when they were used as chemical weapons. They are still being used alone or in combination with other agents in myelogenic leukemia, Hodgkin disease, lung, testicle, ovarian and breast cancer, as well as in several lymphomas [1]. Unfortunately, their remarkable effectiveness is moderated by the serious side effects that accompany the therapy. The biochemical target of alkylating agents is DNA; however they cannot distinguish between tumor-cells and normal cells. Aiming to minimize their toxicity via a more specialized targeting, many researchers have chemically combined different alkylating agents with a diversity of carrier-molecules such as aminoacids , chlorokine and quinacrine , hydrocarbons, purines and pyrimidines, steroids [2–4] and others, with diverse effectiveness. Steroidal molecules seem to comprise a number of physiochemical properties that palliate the toxicity of alkylating agents, allowing for a dose increase of the latter. For this reason, two such combinations, Estramustine and Prednimustine were commercially available for a period of time. In particular, the beneficial effects of steroids added to the alkylating agents can be summarized to: (a) The presence of the steroid in the molecule increases the tumor uptake of the alkylating agent into hormone-dependant tumors; (b) It increases the lipophilicity of the alkylating agent facilitating its transition into the cell and later into the nucleus; (c)-It is speculated, that once the drug approaches the DNA, the steroidal part of the molecule, interacts with its receptor and enables approximation between the alkylating agent and the DNA-helix; (d) Steroids are also thought to metabolically protect the alkylating-part during its course through the body, minimizing the toxicity [1]. The four steroids (I-IV) (Figure 1) were used in combination with several alkylating agents since it is already known that the nature and the conformation of the steroidal skeleton play a critical role in the exhibition of the anticancer activity. The compounds, which were derived with the use of structure I proved completely inactive, while the corresponding compounds derived from II, III and IV proved significantly active. 3D structure-activity relationships based on NMR data would probably lead to a better understanding of stereoelectronic properties that contribute to the bioactivity of such combinations. Taking into account the complexity of steroid 1H-NMR spectra, we proceeded, at a first step to the complete assignment of 1D 1H-NMR and I3C-NMR of compounds I-IV by recording and analyzing the corresponding 1H-13C HSQC, 1H-I3C HMBC, DQF-COSY and ROESY 2D-spectra.
A. Kapou et al. / Synthesis and Conformational Analysis of Steroidal Carriers
AcO''
AcO* 3b-Acetoxy-5-androsten-17-onc
-^
^/
169
-o
3b-Acetoxy-5-androsten-7,17-dione II
AcO
3b-Acetoxy-17a-aza-D-homo-5-androsten-l7-one
3b-Acetoxy-17a-aza-D-homo-5-androsten-7,17-dione
III
IV
Figure 1. Alkylating agents are esterified at the 3a-position of the steroidal carrier-molecules, replacing the acetoxy-group.
Synthetic Chemistry Protection of the hydroxyl-group of dehydroepiandrosterone (DHEA) by acetylation in the presence of acetic anhydride/pyridine produced 3B-acetoxy-5-androsten-17-one (I). Treatment of I with t-BuOOH in dichloromethane, in the presence of CuI, afforded the 3pacetoxy-5-androsten-7,17-dione (II) (Figure 2).
DHEA
i. Ac2O / Pyridine; ii. t-BuOOH, Cul / CH2C12 Figure 2. The synthetic course for steroids I and II
The presence of the 17-keto group facilitated the introduction of a heteroatom in ring D of compound I. Thus, the lactamic steroids III and IV were obtained with the use of 3bacetoxy-5-androsten-17-oxime (Ia) as the synthetic intermediate. Oxime Ia was prepared by the reaction of I with an excess of hydrochloric hydroxylamine in a mixture of absolute ethanole/pyridine. A Beckmann rearrangement of oxime Ia in cold 1,4-dioxane and in the presence of SOCl2, produced 3p-acetoxy-17a-aza-D-homo-5-androsten-17-one (III) and a by-product IIIa, with an open D ring and bearing an exocyclic double bond and a nitrile group. The lactamic steroid III was then subjected to allylic oxidation in dichloromethane with the use of t-BuOOH and CuI as catalysts, affording the 3p-acetoxy-17a-aza-D-homo-5androsten-7,17-dione (IV) (Figure 3).
170
A. Kapou et al. / Synthesis and Conformational Analysis of Steroidal Carriers N-OH
la
1
AcO
AcCT
v
^"
U
Acer
IV
^
v
HI,
III
i. NH2OH.HCl / Pyridine / Abs. EtOH; IL SOCl2 / Dioxane, 0°C; in. t-BuOOH / Cul / CH2Cl2 Figure 3. The synthetic course for steroids III and IV
NMR Spectroscopy: The 1H-NMR ID-spectra of compounds I-IV, as well as the 1H-I3C HSQC, 1H-13C HMBC, DQF-COSY and ROESY 2D-spectra were recorded at a concentration of 15-20 mg/ml in CDC13 at 298 K, on a VARIAN 600 MHz. ROESY spectra were recorded with a selection of a mixing time of 150 msec. The strategy followed for the full assignment of the obtained spectra involved first the analysis of HSQCs coupled with reported data from the literature [5–13]. Then, by means of the analysis of HMBCs, 13C-18, 13C-19, 13CH3CO-, l3C-5, 13C-6, CH313CO-, 13C-7 and 13 C-17 were assigned and the respective long-range 13C-1H couplings were recognized. For example, distinction between CHs-18 and CH3-19 was made, by their different coupling on HMBC (1H-1a, 1B and 9 for 13C-19 and 1H-12a, 12p and 14 for 13C-18) (Figure 4a,b).
X: -Hor=O
A. Kapou et al. / Synthesis and Conformational Analysis of Steroidal Carriers
171
Figure 4. Characteristic long-range C-H couplings (a) observed on the HMBC spectrum of II (b)
3b-acetoxy-5-androstene-7,17-one
Figure 5a shows a representative ROESY spectrum of 3p-acetoxy-5-androstene-17-one, fully assigned. The most important spatial proximities, that is, the axial-axial couplings (<4.5 angstroms) shown on figure 5b, were observed on all ROESY spectra.
172
A. Kapou et al. / Synthesis and Conformational Analysis of Steroidal Carriers
Figure 5b. On the ROESY spectrum of II (a) the most important steroidal axial-axial couplings (b) can be observed.
Table 1 gives a detailed 1H and 13C NMR assignment of the steroids. It can be easily observed that the 1H and 13C chemical shifts of the first two rings are similar in I and III and, II and IV. Thus it can be concluded that when one edge of the molecule is chemically modified, predictions concerning the chemical shifts of distant atoms can be quite safely made. On the contrary, the introduction of a new group in a central part of the molecule seems to affect all atoms. Table 1. 1H and 13C chemical shifts of compounds I-IV
Compound Atom
I 13
// 1
C 1
H a:1,11(m) b:1,83(m) a:133(m) b:1,55(m)
13
C
IV
III 1
1
H a:1,26(m) b:1,95(m) a:1,97(m) b:l,67(m)
36,7-363
73,6
"C
13
C
1
H a:1,13(m) b:133(m) a:135(m) b:1,56(m)
353
H a: 1,23(m) b:1,93(m) a:1,97(m) b:1,67(m)
a:4,58(m)
72,0
a:4,68(m)
a:2,34(m) b:2,27(m)
37,5
a:2,54(m) b:2,42(m)
1
36,7-36,9
2
27,7
3
73,7
a:4,58(m)
71,9
a:4,69(m)
4
38,0
a:2,30(m) b:2,27(m)
373
a:236(m) b:2,46(m)
5
140,0
-
164,8
-
139,6
-
163,0
-
6
121,9
5,38(d)
126,5
5,72(s)
121,7
5,36(d)
126,4
5,72(s)
7
30,8
a:1,62(m) b:2,07(m)
2003
-
31,2
a:1,59(m) b:2,16(m)
200,2
-
2
35,9 27,2
3
27,6
8
31,4–31,5
l,62(m)
44,3
2,37(t)
32,3
1,37(m)
44,6
2,18(t)
9
50,2
1,62(m)
49,9
1,55(in)
49,4
1,12(m)
49,0
l,66(m)
10
36,7–37,01
-
38,5
-
36,7-36,83
-
38,9
-
11
20,3
12
a:1,63(m) b:1,44(m) a:1,25(m) b:1,80(m)
20,5 30,6
a:1,72(m) b:1,55(m) a:1,22(m) b:1,83(m)
20,9 39,7
a:1,67(m) b:1,37(m) a:1,51(m) b:1,63(m)
20,6 38,4
a:1,70(m) b:1,46(m) a:1,47(m) b:1,62(m)
13
47,5
-
473
-
54,4
-
54,0
-
14
51,7
1,26(m)
45,7
l,62(m)
48,0
1,26(m)
41,4
1,75(m)
15
21,9
a:1,92(m)
24,1
a:2,78(m)
19,9
a:1,85(m)
21,4
a:2,71(m)
173
A. Kapou et al. / Synthesis and Conformational Analysis of Steroidal Carriers
b:1,49(m) a:2,04(m) b:2,43(m)
b:1,74(m) a:2,10(m) b:2,43(m)
b:1,45(m) a:2,34(m) b:2,45(m)
b:1,42(m) a:2,41(m) b:2,51(m)
16
35,8
17
220,9
-
220,3
-
171,9
-
172,5
-
18
13,5
0,84(s)
13,7
0,86(s)
22,0
1,14(s)
22,7
1,15(s)
19
19,3
1,01(s)
17,4
1,21(s)
19,2
0,97(s)
16,7
1,21(s)
CH3-CO
21,4
2,01(s)
213
2,02(s)
21,4
2,01(s)
21,2
2,02(s)
CH3-CO
170,5
-
170,2
-
170,4
-
170,2
N-H
-
-
-
-
-
5,83(s)
-
35,6
30,6
30,6
6,19(s)
Catoms that could not be not resolved
References [1] S. Nikolaropoulos, D. Tsavdaridis, E. Arsenou, A. Papageorgiou, E. Karaberis and D. Mourelatos, Anticancer Res., 20 (2000) 2745. [2] G.R Vavasour., H.I. Bolker and A.F. McKay, Canadian J. Chem., 30 (1952) 933. [3] M.E. Wall, Jr. Abernethy, F.I. Carroll and D.J. Taylor, J. Med. Chem., 12 (1969) 810. [4] P. Catsoulacos and I. Boutis, Cancer Chemother. Rep., 57 (1973) 365. [5] Z. Szendi, P. Forgo and F. Sweet, Steroids, 60 (1995) 442-446. [6] H. Duddeck, D. Rosenbaum and M. Hani, A. Elgamal, B. E. Fayez, Magn. Res. Chem, 24 (1986) 999. [7] H. Schneider, U. Buchheit, N. Becker, G. Schmidt and U. Siehl, J. Am. Chem. Soc., 107 (1985) 7027. [8] D. N. Kirk, H. C. Toms, Ch. Douglas and K. A. White, J. Chem. Soc. Perkin Trans.2 (1990) 1567. [9] J. W. Blunt, and J. B. Stothers, Org. Magn. Res., 9 (8) (1977) 439. [10] J. C. Gramain and J. C. Quirion, Magn. Res. Chem., 24 (1986) 938. [11] G. A. Revelli and E. G. Gros, Steroids, 58 (1993) 181. [12] E. Gacs-Baitz, G. Kalaus, P. Gyory, Org. Magn. Res., 22 (11) (1984) 736. [13] V. S. Bogdanov, E. G. Cherepanova, T. M. Fadeeva, A. M. Turuta and A.V. Kamernitskii, Academy of Science of the USSR, 5 (1984) 961.
174
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Structure Elucidation and Conformational Properties of Irbesartan P. Zoumpoulakis1, M. Zervou1, S. G. Grdadolnik2, T. Mavromoustakos1 1
Institute of Organic and Pharmaceutical Chemistry, National Hellenic Research Foundation. Vas. Constantinou 48, Athens, 11635, Greece ' National Institute of Chemistry, Hajdrihova 19 P.O.B. 30 SI-1115, Ljubljana, Slovenia
Abstract Although there have been significant efforts for prevention and treatment, hypertension continues to be one of the major causes of death in the developed countries. A variety of drugs have been deployed for the last 15 years. The last generation of the antihypertensives, were developed to interfere in the Renin-Angiotensin System (RAS) by blocking the binding of the vasoconstictive hormone Angiotensin II to the AT1. receptor site. The first beneficial drug of this category, approved in the market, was losartan (COZAAR) followed by other, - similar in structure -antihypertensive drugs (sartans). All of them act in almost the similar way by binding on the AT, receptor but their biological activity varies significantly. In an effort to comprehend the molecular basis of their bioactivity, the stereoelectronic features of their pharmacophoric segments have been compared. Thus, a research activity was initiated by our laboratory to explore the conformational properties of these drugs. In previous studies, the structural features of losartan (COZAAR) and eprosartan (TEVETEN) were examined. In this study, the conformational properties of irbesartan (KARVEA) and its superimposition ability on losartan and eprosartan are explored.
Introduction The two major systems which regulate the cardiovascular function are the rennin-angiotensin system (RAS) and the sympathetic nervous system (SNS). Throughout the path of the RAS system, the octapeptide ANG II is synthesized. This peptide elicits potent vasocontsticitive effects when interacting with AT1 receptor; a member of the superfamily of the G-protein-coupled receptors, already cloned from rat, pig and human libraries [1].
P. Zoumpoulakis et al. / Structure Elucidation and Conformational Properties of
Irbesartan
175
Among the categories of potent antihypertensive drugs, the most recent one includes molecules which prevent ANGII from binding the AT1 receptor and are called ANGII receptor antagonists (AT1 antagonists). In order to obtain information on the regiochemical and stereochemical requirements for productive binding at the active site of the AT1 receptor, an effort was made to study the conformational analysis of synthetic peptides and non-peptide mimetic AT1 antagonists. The conformational properties of the first AT1 antagonist losartan, to the AT1 receptor have been already studied. In particular, the structural features which determine the pharmacophoric segments of losartan are examined. These are: a) conformation of biphenyltetrazole; b) orientation of hydroxymethylimidazole relative to the biphenyl template; and c) the n-butyl chain conformation. Losartan was superimposed with Cterminal region of AT1 antagonist sarmesin since its design was based on its similarity with side groups that constitute the C-terminal part of Angiotensin II. Superimposition of losartan with AT1 antagonist sarmesin showed that: (i) losartan's hydroxymethylimidazole matched with sarmesin's imidazole of His6; (ii) losartan's nbutyl chain was in a spatial proximity with n-butyl chain of sarmesin's Ile5 carbon chain; (iii) losartan's tetrazole was in close vicinity in space with sarmesin's isosteric carboxylate of Phe8 and (iv) losartan's spacer phenyl ring matched with Sarmesin's pyrrolidine group of Pro7. Interestingly, losartan mimics the y-turn formed around Pro7 in sarmesin [2]. Recently, similar superimposition study was sought with the c-[Sar1Lys3-Glu5-Ile8] ANG II. Losartan as sarmesin mimics the y-turn formed around Pro7 in c-[Sar1-Lys3-Glu5-Ile8] ANG II [3]. Our studies have been extended to the detailed conformational analysis of irbesartan, another effective blood pressure lowering agent. Compared to other sartans, irbesartan appears to possess unique actions among the AT1 blockers. Many recent biological studies have shown that single dose of irbesartan caused greater and more sustained blockade on the AT1 receptor than the other sartans [4]. Unlike losartan, irbesartan doesn't metabolizes into active metabolites. The present study will aid in exploring the conformational similarities and differences of irbesartan with the AT] antagonists losartan and eprosartan. Figure 1.
EPROSARTAN
IRBESARTAN
Figure 1: Structures of the three drugs which act as AT1 antagonists. Losartan, eprosartan and irbesartan.
Their superimposition similarities and differences of losartan and eprosartan with Cterminal parts of sarmesin has been already studied [5]. The final aim of these studies is
176
P. Zoumpoulakis et al. / Structure Elucidation and Conformational Properties of Irbesartan
to explore their conformational dynamic properties in membrane bilayers and docking to AT1 receptor. These under progress studies may shed light to the molecular basis of their action. Irbesartan was kindly offered by the pharmaceutical company BristolMyers Squibb. Results and discussion Structure identification of Irbesartan: Figure 2 depicts the H NMR spectrum of irbesartan in DMSO solvent. Observed peaks are referenced to TMS. The assignment of the peaks is shown on the top of the spectrum. The proton chemical shifts of irbesartan are assigned following standard procedures and using homonuclear DQF-COSY and ROESY in combination with 1H-13 C HSQC and 1H-13C HMBC experiments.
10 6,6a,6b,6c DMSO, 7600
7200
6800
6400
6000
5KB
5200
4800
4400
4000
3600
3200
2800
2400
2000
1600
1200
0900
13,14,16,17
Experimental 22
7700 7.650 7603 7550 7500 7450 7400 7350 7300 7250 7200 7150 7100 7050
Simulated
7700 7650 7600 7550 7500 7450 7400 7350 7300 7250 7200 7150 7100 7050
Figure
2:
Proton
NMR
spectra
of
irbesartan and
simulation
of
the
aromatic region
P. Zoumpoulakis et al. / Structure Elucidation and Conformational Properties of
Irbesartan
177
Conformational properties of Irbesartan: The conformational properties of the molecule depend on the observed NOEs (Table I). These NOEs suggest a conformation similar to the one of losartan. Table I: NOE and their intensities for the molecule of irbesartan
#
Protons Intensity Distance (A) 7-11 2.168 vS 2.898 2 7-(13,14,16,17) S 2.167 11-(13, 14,16,17) vS 3 2.683 4 19-(13, 14,16,17) S 1: light, m: medium, s: strong, vs: very strong
1
Conformational Search: A low energy conformer of irbesartan, derived using a combination of NMR spectroscopy and Computational Analysis, as shown in Figure 3.
Figure 3: Conformation of irbesartan
Superimposition on Eprosartan and Losartan: The low energy conformer of irbesartan derived from the combination of NMR spectroscopy and computational analysis was superimposed with losartan and eprosartan. The superimposition involved the following equivalent matching groups: (i) the nitrogen atoms of the imidazole ring of the three molecules, (ii) eprosartans' carboxylate group with the isosteric tetrazole of losartan and irbesartan. The superimposed molecules showed a nice overlapping. RMS value for the superimposition with losartan was 0.06 and for that with eprosartan was 0.5. The biphenyl tetrazole systems were matched while the butyl chain of irbesartan seems to favor a position farther of the phelyl rings than the molecule of losartan and eprosartan. Figure 4.
178
P. Zoumpoulakis et al. / Structure Elucidation and Conformational Properties of Irbesartan
Figure 4: Superimposition of irbesartan with losartan (right) and eprosartan (left).
Conclusions A systematic exploring of the conformational properties of AT| antagonists constitutes a basis for the attempt to comprehend their stereoelectronic properties which govern their antihypertensive action. Combination of 2D NMR spectroscopy and computational analysis help to explore the lowest energy conformers of various AT1 antagonists. The interaction of these conformers with the membranes as well as with the AT1 are already in progress in order to shed light on the way of action of these drugs. The research plan is rationalized from the fact that AT1 antagonists act on the membrane helices of AT1 receptors. Therefore, the mechanism of action of these molecules is energetically favored to involve their incorporation and then diffusion into membrane bilayers to reach the receptor site. The anchoring of pharmacophore segments in the receptor may determine the molecular requirements for bioactivity and may lead to novel structures that possess better activity. Acknowledgments This work was supported by a) the Ministry of Energy and Technology of Greece (EPET II/115, PENED 2001), b) Workpackage WP9 "Conformational analysis of peptidomimetic drugs" of the Program of the Center of Excellence at National Institute of Chemistry in Ljubljana Slovenia sponsored by European Commision, contract no: ICA1–CT–2000–70034.
P. Zoumpoulakis et al. / Structure Elucidation and Conformational Properties of
Irbesartan
179
References [ 1] M. De Gasparo, K.J. Catt, T. Inagami, J.W. Wright, Th. Unger, Pharmacol. Rev. 52 (2000) 415. [2] T. Mavromoustakos, A. Kolocouris, M. Zervou, P. Roumelioti, J. Matsoukas, R. Weisemann., J. Med. Chem. 42 (1999) 1714. [3] L. Polevaya, T. Mavromoustakos, P. Zoumboulakis, S. G. Grdadolnik, P. Roumelioti, N. Giatas, Ilze Mutule, T. Keivish, D. Vlahakos, E. Iliodromitis, D. Kremastinos, Bioorg. Med. Chem. 9 (2001) 1639. [4] L. Mazzolai, M. Maillard, J. Rossat, J. Nussberger, H.R. Brunner, M. Burnier, Hypertension 33 (1999) 850. [5] P. Zoumpoulakis, S.G. Grdadolnik, J. Matsoukas, T. Mavromoustakos, (in press) J. Pharmac. Biol. Anal.
180
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
1
H and 13C NMR Assignment and
Conformational Studies of a Myelin Basic Protein Epitope 74-85 Implicated in Multiple Sclerosis A. Tzakos1, A. N. Troganis2, T. Tselios4, A. Bonvin3, N. van Nuland3, S. Deraos4, J. Matsoukas4 and I. P. Gerothanassis1 1
Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece. Department of Biological Applications and Technologies, 45110 Ioannina, Greece 3 Bijvoet Center for Biomolecular Research, Department of NMR Spectroscopy, Padualaan 8, 3584 CH Utrecht, the Netherlands Department of Chemistry, University of Patras, 26500 Patras, Greece
2
Abstract: Detailed 1H and 13C NMR chemical shift assignments, 2D 1H-1H NOESY experiments, and conformational properties of two potent linear dodecapeptide analogues of Myelin Basic Protein are reported. The two analogues Gln74 -Lys75-Ser76-Gln77-Arg78-Ser79-Gln80-Asp8l-Glu82-Asn83Pro84-Val85 (MBP74-85) and Gln74-Lys75-Ser76-Gln77-Arg78-Ser79-Gln80-Ala81Glu82-Asn83-Pro84-Val85 (Ala8lMBP74-85), which induce and inhibit respectively Experimental Autoimmune Encephalomyelitis [EAE, the animal model of Multiple Sclerosis (MS)], differ only in the aminoacid residue at position 81 (Asp or Ala) resulting in relatively similar folded conformations with different activities. Both peptides show strong tendency for aggregation even at low concentration in DMSO solution. The aggregation mode is, mainly, through head to tail inter-molecular interactions. 2D 1H-1H NOESY experiments at sufficiently low concentrations (< 2 mM) where both peptides exist mainly in the monomeric state indicate a number of significant interresidue NOEs, such as strong NOE connectivities between pVal 85 -yGln 74 in MBP74-85 and BPro84 -yGln74 in Ala8lMBP74-85. Restricted Molecular dynamics based on NOE constrains indicate the close proximity of Lys75/Arg78/Asp81 side chain observed in agonist MBP74-85 but not in antagonist Ala 81 MBP74-85, which may account for the triggering of the disease.
A. Tzakos et al. / 1H and 13C NMR Assignment and Conformational Studies
181
Introduction
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS), which is believed to be mediated by autoreactive T cells [1-3]. The activation of resting T cells reactive with CNS antigen is thought to constitute the primary autoimmune event in multiple sclerosis. Myelin Basic Protein (MBP) plays an integral role in the structure and function of the myelin sheath [4-6]. It was the first agent in brain or spinal cord homogenates, which was discovered to be responsible for experimental allergic encephalomyelitis (an animal model for the human multiple sclerosis disease) [7–9]. Some of the most important functions of MBP include stimulation of phospholipase C activity [10,11], actin polymerization in conjunction with Ca2+-calmodulin [12,13] tubulin stabilization [14] or even potential regulatory roles as transcription factors [15]. Knowledge of the tertiary structure of MBP would facilitate a better understanding of how it carries out its diverse functions. Recently, the threedimensional structure of MBP was reported from molecular modeling and electron microscopy data. Furthermore, it is of critical importance to investigate the structure of the epitopes of the protein, revealed in the segments 1-14, 22-34, 43-68, 67-75, 75-82, 83-96, 90-99, 114-121, 118-131, 125-131, 130-137 and 131-140 [16-20] which have antigenic properties. Characterization of the molecular features of antigenic sites could provide insights into the immunogenic properties that can be used as valuable tools for the design of synthetic peptides that can act as new vaccines or artificial regulators of the immune response. Linear and cyclic analogues of the 72-85 epitope have been synthesized in order to identify pharmacophoric groups and develop a molecular model, which could be useful in drug design of potential therapeutics [21,22]. This paper addresses the question of the structural features of the enchephalitogenic epitope MBP74-85 of the segment 74 - 85 (Gln74-Lys75-Ser76-Gln77-Arg78-Ser79-Gln80-Asp81Glu82-Asn83-Pro84-Val85 (MBP74-85)) of the guinea pig MBP and the synthetic peptide Ala81MBP74-85 (Gln74-Lys75-Ser76-Gln77-Arg78-Ser79-Gln80-Ala81-Glu82-Asn8l-Pro8485 81 Val (Ala MBP74-85)), which inhibits the Experimental Autoimmune Encephalomyelitis (EAE). Materials and Methods Synthesis: The synthesis of linear MBP analogues was achived using Fmoc/tBu methodology. 2-Chlorotrityl cloride resin and Na-Fmoc amino acids were used for the synthesis as described previously [23–27]. Peptide purity was assessed by analytical HPLC (Nucleosil-120 C18, reversed phase, 250X4.0mm), mass spectrometry (FABMS, ESIMS) and amino acid analysis. 1
H- and 13C- NMR Spectroscopy: Samples of the MBP74-85 and Ala 8l MBP 74-85 analogues were dissolved in DMSO-d6 at 2mM concentration, pH 5.7, 295 K and dispensed into 5-mm tubes. NMR spectra were recorded on a Bruker AMX-400 spectrometer at 300K. The chemical shifts are reported with respect to the resonance of the solvent. All proton 2D spectra were acquired using the TPPI method for quadrature detection, with 2K x 512 complex data points and 48 scans per increment for 2D TOCSY and 160 scans for 2D NOESY experiments, respectively. The mixing time for
182
A. Tzakos et al. / 1H and 13C NMR Assignment and Conformational Studies
TOCSY spectra was 100 ms and for NOESY 350 ms. Phase - sensitive 2D NOESY was used for specific assignment and for estimation of proton - proton distance constrains. Data were processed by using the NMRPIPE software [28]. The tl dimension was zerofilled to give 1 x 1 K real data points, and 45° phase shifted square sine - bell window function was applied in both dimensions. A mixing time of 350 ms provided sufficient cross-peak intensity without introducing spin-diffusion effects and 2D - NOESY were carried out with concentrations of five and one mM of compound MBP74-85 in DMSO in order to verify aggregation head to tail intermolecular NOEs. Interproton distances were derived from quantification of NOE cross-peak intensities in NOESY experiments. Amide NH solvent accessibility was deduced from chemical shift temperature coefficients. I3 C NMR spectra were obtained at 100.6 MHz, and the assignment of the peaks was based on 1H, 13C HMQC and HMBC experiments. The spectra were acquired with 2K x 256 points, 72 and 160 scans per increment, respectively. The tl dimension was zero- filled to 1K, to give an 1K x 1K real points and 0° square cosine - bell window function was applied in both dimensions. Structure Calculations: The presence of conformational averaging in linear peptides can complicate the calculation of singular structures, which are based on the use of intraresidue and sequential NOEs. For this reason, only medium- and long-range NOE cross-peaks were considered in the calculations. NOE cross peaks were separated into three distance categories according to their intensity. Strong NOE enhancements were given an upper distance constraint of 3.0 A, medium NOE enhancement a value of 4.0 A and weak NOE enhancements 5.5 A. All calculations were performed with CNS [29] using the ARIA setup and protocols [30]. A simulated annealing protocol in Cartesian space was used starting from an extended conformation [30]. The structures were subjected to a final refinement protocol in explicit water by solvating them with a 8 A layer of TIP3P waters [31]. Results and Discussion 1
H and 13C Assignment: Amino acid spin systems were identified by locating networks of characteristic connectivities in the 2D TOCSY spectra [32] and 13C resonances were identified with the use of 1H - I3C HMQC and HMBC spectra. The complete lists of 1H and I3C chemical shifts and the temperature coefficients of the backbone NH amide proton resonances were measured in DMSO-d6 are shown in Tables I-FV and in Figure 1. Agonist
Antagonist
Figure 1. Plot of temperature coefficients against the aminoacid residues of the MBP74-85 agonist and the Ala 8l MBP 74-85 antagonist.
A. Tzakos et al. / 1H and 13C NMR Assignment and Conformational Studies
183
Table I: 1H NMR assignments and chemical shifts (5, ppm) and NH temperature coefficients (-A8/AT) of the Agonist Linear Analogue of MBP74-85 in DMSO-d6 at pH 5.7 and 295 K
GIn4
8.10
4.26
1.94 1.90a
2.12
Arg5
8.03
4.32
1.70
1.50
Ser6 Gln7
7.98 8.08
4.31 4.23
Asp8
8.18
4.52
Glu9
7.75
4.24
Asn10
8.18
4.76
Pro 11
-
4.42
Val12
7.88
4.03
3.60 1.91 1.76 2.69 2.51 1.89 1.74 2.54 2.35 1.99 1.88 2.04
3.10
7.52
7.21 6.80 7.28 7.15 7.02
.
2.13
7.23 6.77
5.16 5.51
5.51 5.63
-
_
5.42
2.22
.
5.05 7.38 6.88
- 1.87 1.83 0.88 0.86
5.42
3.61 -
6.05
Ambiguous assignment. Table II: 13C NMR assignments and chemical shifts (5, ppm) of the Agonist Linear Analogue of MBP74-85 8 AA CO a P Y Residues Glnl 51.79 27.01 168.25 173.59 30.36 52.47 31.42 26.62 38.68 Lys2 171.35 22.10 54.94 61.57 Ser3 170.08 Gln4 52.31 27.90 174.08 170.61 31.36 24.84 171.22 52.03 156.74 29.06 40.40 Arg5 54.94 Ser6 61.57 169.98 Gln7 52.31 27.73 31.18 173.99 171.66 174.02 Asp8 170.20 49.55 35.96 29.94 Glu9 171.15 51.60 27.59 180.49 169.69 47.72 173.41 Asn10 36.86 Pro 11 171.48 59.16 28.88 24.16 46.66 172.74 57.39 Val12 18.20 29.66 19.12 Table III: 1H NMR assignments and chemical shifts (8, ppm) and NH temperature coefficients (-AS/AT) of the Antagonist Linear Analogue of MBP74.85 (Ala81 MBP74-85) in DMSO-d6at pH 5.7 and 295 K AA NH a -AS/AT 8 NH2 Y P (ppb/K) Glnl 8.20 3.85 1.94 2.23 7.41 1.90 6.95 4.37 Lys2 8.60 1.71 1.33 1.54 2.76 7.76 3.32 _ Ser3 8.15 4.33 3.61 5.63 Gln4 8.09 4.25 1.95 2.13 7.23 4.81 1.74 6.78 7.26 6.51 8.03 4.32 Arg5 1.70 1.50 3.10 7.53 7.13
A. Tzakos et al. / 1H and 1 3 CNMR Assignment and Conformational Studies
184
Ser6 Gln7
7.97 8.07
4.30 4.23
Ala8 Glu9
7.99 7.87
4.24 4.23
Asn10
8.13
4.76
Val12
Table IV: MBP74-85 AA Residue Glnl Lys2 Ser3 Gln4 Arg5 Ser6 GIn7 Ala8 Glu9 Asn10 Pro 11 Val12
.91 .73 .20 .87 1.71
7.89 13
4.03
6.24 7.20
7.23 6.78
2.13
2.22
-
2.53 2.35 2.00 1.97 2.04
4.42
Pro 11
7.01
-
3.59
1.91 1.83 0.88 0.86
-
6.24 4.96
7.39 6.89
5.44
-
3.62
-
5.89
C NMR assignments and chemical shifts (5, ppm) of the Antagonist Linear Analogue of
CO
a
P
Y
6
168.22 171.28 170.04 170.68 171.15 169.92 171.12 171.92 171.07 169.70 171.45 172.72
51.77 52.43 54.92 52.29 52.15 54.92 51.99 48.21 51.44 47.66 59.12 57.35
26.99 31.42 61.60 27.89 29.09 61.60 27.68 17.93 27.62 36.85 28.87 29.66
30.35 22.08
173.54 26.60
-
-
31.35 24.81
174.04 40.39
-
-
31.26
173.94
-
-
30.03 170.80 24.16 18.17 19.11
177.50
46.64
-
e .
;
38.66
156.70
-
Homonuclear 1H — 1H NOE and Molecular Structure Calculations: Structure elucidation and conformational properties of the analogues MBP74-85 and Ala8lMBP74-85 were obtained through 1H-1H NOESY experiments. A summary of some representative medium- and long-range inter-residue backbone NOE connectivities is presented in Table V. These NOE constrains were used in the ARIA/CNS set up [29, 30] for structure calculations. A simulated annealing protocol in Cartesian space was used starting from an extended conformation. Force constants were scaled throughout the protocol following the default ARIA/CNS set up [29, 30] and the structures were subjected to a final refinement protocol in explicit water. The resulting structures were energy minimized with 100 steps of Powell steepest descent minimization and the structure stereochemistry was evaluated through PROCHECK [33]. These calculations yielded 20 low-penalty structures, which satisfied all the co-ordinate geometry, and NMR distance constrains. This family of refined structures was compared and the root-mean square deviation (rmsd) of backbone atoms measured throughout all segments in the molecules. Both the N- and Cterminal regions exhibited some conformational averaging, but the segment Ser 76-Asn83 appeared to maintain a more consistent conformation. Backbone atoms of the twelveresidue segment Gln74-Val85 were superimposed and were found to possess an rmsd value equal to 0.9 A. A stereo view of the backbones of the superimposed structures for the agonist enchephalitogenic epitope MBP74-85 and the synthetic antagonist analogue Ala 8l MBP 74-85 is shown in Figure 2A and 2C respectively. Similar folded conformations (closed
A. Tzakos et al. / 1H and 13C NMR Assignment and Conformational Studies
185
loops) can be observed for both analogues. From the twenty refined structures of the agonist MBP74-85 and antagonist analogue Ala8lMBP74-85 we selected for simplicity one structure for each analogue (Figure 2B and 2D respectively). The selection criterion was the rmsd value of the structure, which has the lowest deviation from the average rmsd. The sidechains of Lys75, Arg78, Asp81 in the case of the agonist MBP74-85 are in close proximity. In the case of the antagonist Ala 81 MBP 74-85 the sidechains Lys75, Arg78 seems to be also in close proximity but the sidechain of Ala 81 appears to be in the opposite site. Figure 3 shows a stylised view of superposition of the peptide structure of agonist (blue) and antagonist (red) with the representation of the sidechains of Lys75, Arg78, Asp81 (agonist) and Lys75, Arg78, Ala81 (antagonist). The close proximity between the side chains Lys75, Arg78, Asp81 of MBP74-85 is manifested, whereas the antagonist represents a different behaviour as the sidechains of Lys75 and Arg78 are in opposite direction than those of the agonist and further the sidechain of residue at position 81 (Ala81) is in opposite direction and far from the sidechains of Lys75 and Arg78 . The more compact conformation of the agonist analogue (that brings the sidechains of Lys75, Arg78, Asp81 in close proximity) compared with the conformation of the antagonist is further supported from the deviation of the amide temperature coefficients between the agonist and the antagonist showing in Figure 1. Specifically, all the residues of the agonist that are located in the Lys75-Glu82 block, appear to be more protected than the NH protons of the antagonist and so the Ala81 MBP74-85 (antagonist) could obtain in that block a more open structure without the close proximity of Lys75 , Arg78 , Ala 81 .
Table V. List of Representative NOEs Used in the Distance Geometry Structure Determination for the Trans Isomers of the Agonist and Antagonist Linear Analogues of MBP74-85. Antagonist Magnitude of NOE Agonist Magnitude of NOE 1 aProll-aVall2 1 aAsp8-aAsn 10 aAsn10-8Proll aAsnlO-8Proll s s pAsnlO-5Pro11 m pAsnlO-pSer6 m yPro11-yVal12 1 m pAsnlO-aSer6 m pVal12-yGlnla pGlu9-yVal12 m 1 NHVal12-NHGln4b m 6Lys2-NHGIn4 NHGlu9-nNHArg5b s NHLys2-nNHArg5b s NHGlnl-nNHArg5 b m m NHGlnl-nNHArg5 b NHGlnl-otProll" m yGlnl-yProll a m CNHLys2-aGlnl 1 1 NHLys2-yGlni NHVall2-pProll 1 NHLys2-pGlnl m 1 CONHAsnlO-yVall2 CONHGlnl-yLys2 1 1 1 eNHArg5-5Lys2 eNHArg5-5Lys2 a Some contribution due to intermolecular head-to-tail association was observed. b Exchange cross peak
186
A. Tzakos et al. / 1H and I3C NMR Assignment and Conformational Studies
Figure 2. Stereo view of the backbone structures of the agonist (A and B) and antagonist (C and D) linear
analogues of MBP74-85.
agonist
antagonist
antagonist
Figure 3. Superposition of agonist and antagonist analogues of MBP74-85 denoted by arrows
Acknowledgements This work was supported by the Ministry of research and Technology of Greece (EPET II 15, PENED 1999) References [1] Martin, R.; McFarland, H.; McFarlin, D. Annu. Rev. Immunol. 10 (1992) 153. [2] Steinman, L. Cell 85 (1996) 299. [3] Ota, K.; Matsui, M.; Milford, EL.; Mackin, GA.; Weiner, HL.; Hafler, DA. Nature (Land). 346 (1990) 183. [4] Deber, C. M. & Reynolds, S. J. Clin. Biochem. 24 (1991) 113. [5] Kirschner, D. A., Inouye, H., Ganser, A. L. & Mann, V. J.Neurochem. 53 (1989) 1599. [6] Moscarello, M. A. in Cell Biology and Pathology of Myelin: Evolving Biological Concepts and Therapeutic Approaches (Devon, R. M., Dooucette, R., Juurlink. B. H. J., Nazarrali, A. J., Schreyer, D. J. & Verge, V. M. K.) 1986, Plenum Press, New York [7] Carnegie, P. R. Biochem. J. 123 (1971) 57. [8) Eylar, E. H., Brostoff, S., Hashim. G., Cacaam, J. & Burnett, P. J. Biol. Chem. 246 (1971) 5770. [9] Kies. M. Annn. N. Y. Acad. Sci. 122 (1965) 161. [10] Tompkins. T. A. & Moscarello, M. A. Arch. Biochem. Biophys. 302 (1993) 476. [11] Tompkins. T. A. & Moscarello. M. A. Biochim. Biophys. Ada 1206 (1994) 208.
A. Tzakos et al. / 1H and 13C NMR Assignment and Conformational Studies
187
[12] Chan, K. F. J., Robb, N. D. & Chen, W. H. J. Neurosci. Res. 25 (1990) 534. [13] Dobowolski, Z., Osinksa, H., Mossakowska, M. & Barylko, B. Eur. J. Cell Biol. 42 (1986) 17. [14] Pirollet, F., Derancourt, J., Haiech, J., Job, D. & Margolis, R. L. Biochemistry 31 (1992) 8849. [15] Staugaitis, S. M., Colman, D. R. & Pedraza, L. BioEssays 18 (1996) 13. [16] Sires, I. R., Hruby, S., Alvord, E. C. Jr., Hellstrom, L, Hellstrom, K.-E., Martenson, R. E. et al., Science 214 (1981) 87. [17] Fritz, R. B. & Chou, C.-H. J. J. Immunol. 130 (1983) 2180. [18] Carnegie, P. R., Dowse, C. A. & Linthicum, D. S. J. Neuroimmunol. 5 (1983) 125. [19] Hruby, S., Alvord, E. C. Jr, Martenson, R. E., Debler, G. E. Hickey, W. F. & Gonatas, N. K. J. Neurochem. 44 (1985) 637. [20] Hruby, S., Alvord, E. C. Jr, Groome, N. P., Dawkes, A. & Martenson, R. E., Mol. Immunol. 24 (1987) 1359. Martenson R. E., J. Neurochem. 46 (1986) 1612. [21] Tselios T., Daliani I., Probert L., Deraos S., Matsouka E., Roy S., Pires J., Moore G., Matsoukas J. Bioorg. Med. Chem. 8 (2000) 1903. [22] Tselios T., Daliani I., Deraos S., Thymianou S., Matsouka E., Troganis A., Gerothanassis I., Mouzaki A., Mavromoustakos T., Probert L., Matsoukas J. Bioorg. Med. Chem. Let. 2000, In Press [23] Tselios, T.; Probert, L.; Kollias, G.; Matsoukas, E.; Roumelioti, P.; Alexopoulos, K.; Moore, G.; Matsoukas J. Amino Acids 14 (1998) 333. [24] Tselios, T.; Probert, L.; Daliani, I.; Matsoukas, E.; Troganis, A.; Gerothanassis, P.; Mavromoustakos, T.; Moore, G.; Matsoukas J. J. Med. Chem 42 (1999) 1170. [25] Tselios, T.; Daliani, I.; Matsoukas, E.; Roumelioti, P.; Alexopoulos K.; Probert, L.; Pires, J.; Moore, G.; Matsoukas, J. (Ed. J. Matsoukas and T. Mavromoustakos), Biomedical and Health Research Series, V. 22, p. 255–264, Ios Press 1999 [26] Barlos, K.; Gatos, D.; Schafer, W. Angew. Chem., Int. Ed. Engl. 30 (1991) 590. [27] Barlos, K.; Gatos, D.; Hondrelis, J.; Matsoukas, J.; Moore, G.; Schafer, W.; Sotiriou, P. Liebigs Ann. Chem. (1995) 951. [28] Delaglio, F., Grzesiek, S.; Vuister, G. W.; Zhu, G.; Pfeifer, J. and Bax A. J; Biomol. NMR 6 (1995) 277. [29] Brunger, A.T., Adams, P.O., Clore, G.M., DeLano, W.L., Gros, P., Grosse-Kunstleve, R.W., Jiang, J.-S., Kuszewski, J., Nilges, N., Pannu, N.S., Read, R.J., Rice, L.M., Simonson, T., & Warren, G.L., Acta Cryst. D54 (1998) 905; Linge, J.P. & Nilges, M. J. Biomol. NMR 13 (1999) 51; Nilges, M. & O' Donoghue, S., Prog. NMR Spectroscopy 32 (1998) 107. [30] Linge, J.P. & Nilges, M. J. Biomol. NMR 13 (1999) 51. [31] Jorgensen, W., Chandrasekar, J., Madura, J., Impey, R., and Klein, M. J. Chem. Phys. 79 (1983) 926. [32] Wutrich Kurt, NMR of Proteins and Nucleic Acids 1986, Wiley Interscience [33] Laskowski R. A., MacArthur M. W., Moss D. S., and Thorton J. M., J. Appl. Cryst. 26 (1993) 283.
188
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) 1OS Press, 2002
Conformational Analysis of Biomolecules in Solution: NMR - 3D Structures Biological Implications G. A. Spyroulias, S. Papazacharias, A. Galanis and P. Cordopatis Laboratory of Pharmacognosy and Chemistry of Natural Products, Department of Pharmacy, University of Patras, Patras, Greece
Abstract. Over the last decade the devolepement on technology and methodology in NMR Spectroscopy was achieved in concert with the detailed analysis and exploitation of NMR data, especially these that can provide geometrical information (NOEs - Nuclear Overhausser Enhancement, paramagnetism, relaxation times, 3 J H N H a couplings constants) in such a way that structural constrains could be extracted and used in the determination of 3D structures of biomolecules in solution. These structures are characterized by resolution comparable to that provided by X-ray in solid state. The assignment of 2D, 3D or even 4D NMR spectra is the stage where geometric information for the biomolecule becomes available. The main volume of the information is extracted from connectivities between protons close to each other in a distance up to 5.0-5.5 A. The secondary structure could be identified through characteristic sequential NOE signals between HN, Ha and Hp protons, while tertiary structure is calculated through computational procedures using NOE-derrived distance constraints. The strategy of solution structure determination of biomolecules is presented below for the case of the synthetic bioactive peptide of h/r Corticotropin Releasing Hormone (CRH, 41 aminoacids). Investigation of the structural characteristics of this peptide in solution, has a great impact not only in the study of its Structure-Fuction relationship, but also in the design and synthesis of new analogues. Rational modification and/or change of residues that play a key role in the overal charge of the molecule as well as in the folding and stabilization or destabilization of its tertiary structure, significantly contributes to monitor its reactivity towards the structural features.
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
189
Introduction It is beyond any doubt the fact that the profound understanding of the function of a biological macromolecule it is strongly coupled with the way that its polypeptide chain folds in order to adopt a well-defined tertiary structure. The two techniques that could afford the three-dimensional structure of a biomacromolecule is the X-ray crystallography in the solid state (single crystals) and nuclear magnetic resonance (NMR) spectroscopy in solution. NMR overcomes the drawbacks arised in the way to obtain suitable crystals for diffraction (time-scales, preparation of heavy-atoms derrivatives, etc.) and from the structural restriction imposed on the solid state. At the same time the size of macromolecular structures that can be solved by NMR has been dramatically increased over the past few years, due to the recent development in the technology of the instrumentation and methodology. At Brookhaven Protein Data Bank (PDB) [1] at the end of 1999 had been deposited the coordinates of more than 1000 NMR structures of proteins and nucleic acids which their size reach up to 35 kDa while the vast majority of them range between 2 and 20 kDa [2]. Theoretical Background Every proton possess a property known as magnetization, and when is found in a static magnetic field B0, the magnetization lies parallel to Bo (defined as z direction). To record a conventional 1D NMR spectrum, a radio-frequency pulse B\ is applied, which rotates the magnetization away from z-axis towards the x-y plane. A free-induction decay is recorded immediately after the pulse and yields the conventional spectrum after Fourier transformation. Limitation arised from signal overlapping in 1D spectra, when the number of protons are increased significantly, can be overcome by extending the measurements into a second dimension (or even into a third or fourth). 2D NMR experiments are based to the classic experimental scheme [3] consisting of a preparation period, an evolution period (t1) (during which the spins are labelled according to their chemical shift), a mixing period (during which the spins are correlated with each other), and finally a detection period (t2). A number of experiments are recorded with successively incremented values of the evolution period t\ to generate a data matrix s(t1,t2). 2D fourier transformation of this matrix yields the desired 2D frequency spectrum S(w1, w2). In most homonuclear 2D experiments, the diagonal corresponds to the 1D spectrum, and the symmetrically placed cross peaks on either side of the diagonal indicate the existence of an interaction between two spins while the type of this interaction depends on the type of the experiment. Thus, in a correlation experiment (COSY [4]) experiment, the cross peak arises from through-bond scalar correlations, while in a nuclear Overhauser enhancement (NOE) experiment, they arise from through-space correlations.
190
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
Figure 1. Flow-chart illustarting the various steps of the general strategy applied in Biomolecular NMR and Solution Structure Determination using the modules of DY ANA and AMBER programs.
In the initial stage of any investigation of a macrobiomolecule by NMR spectroscopy, each nuclear magnetic resonance must be associated with a specific nucleus in the molecule under investigation. Resonance assignments must be sequence specific: each resonance must be assigned to a spin in a particular amino acid or base in the protein or in nucleic acid, respectively. NMR spectroscopy provides three type of information useful for spectral assignments: (a) through-bond interactions (via scalar couplings in COSY [4], TOCSY [5] spectra), (b) through-space interactions (via dipolar couplings such as in NOESY [6] spectra), and (c) chemical environment (via the chemical shift). The strategies employed for resonance assignments depend on whether only homonuclear 1H NMR spectra are available (unlabeled proteins) or I3C and I5N heteronuclear correlation spectra are also available (isotopically labeled proteins). The summary of the general strategy employed to solution structure determination of biomacromolecules is illustrated at Figure 1.
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
191
Resonance Assignment Strategies. Among the various steps of the multidimensional procedure of Solution Structure Determination, the methods used for the resonance assignment are of great interest. Concerning the majority of resonances that are to be assigned, due to the 1H nuclei, should be underlined that with only few exceptions, correlations resulting from 1H-1H scalar couplings can be observed only between protons separated by two, three or four bonds in protein (amino acids) or in nucleic acids (bases).
Figure 2. Sequential connectivities observable in NOESY experiments for a tripeptide fragment.
The cross-peaks in 1H-1H correlation NMR spectra occur between protons within the same amino acid residue or spin system, but not between protons in different residues (since the interresidue 4JH,+iNH,a coupling constant is neglible). This observation guides the procedure by which 1H resonance assignments are made. Scalar correlation experiments, such as COSY [4] or DQF-COSY [7] and TOCSY [5] experiments are used to identify resonance positions within each aminoacid spin system, while the NOESY [6] experiment is used to sequentially connect the various amino acid spin systems (see Figure 2). Protons
NH
Ha
Helix B-strand Coil 5(ppm) Chart l.Dependence of 1H chemical shifts in amino acids on protein/polypeptide secondary structure in water.
Initially, the 1H resonances are categorized on the basis of their chemical shifts. As indicated in Figure 3 for the vast majority of residues in a protein or a polypeptide in aqueous solution, the backbone amide protons (NH) resonate between 10.0 and 7.0 ppm, the backbone ot-protons (Ha) resonate between 6.0 and 3.5 ppm, aliphatic sidechain protons resinate between 3.5 and 1.0 ppm while protons of methyl groups resonate at chemical shift values smaller than 1.5 ppm. There is also a dependence of 1H
192
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
chemical shifts on amino acid type and on protein secondary structure summarized in the work of Wishart et al [8] (see Chart 1) and analysis of the chemical shift values for Ha backbone protons could provide valuable insight and even a prediction for the element of the secondary structure (coil, a-helix, B-strand) according to the Chemical Shift Index [9]. The backbone amide protons are usually the best resolved set of resonances within a protein 1H NMR spectrum; thus the Ha and side-chain resonance positions are most readily determined in scalar correlation experiments by the observation of direct and relayed cross-peaks to the amide protons [10]. This could be the starting point of the principal process of determining 1H resonance assignments developped and proposed by Wuthrich and coworkers [11] at mid 1980's.
Figure 3. Regions of a 2D NMR spectrum characterized by the statistical chemical shift ranges of the various types of 1H resonances in proteins/polypeptides.
The first stage of spectral data analysis makes use of scalar couplings (combined use of COSY and TOCSY NMR maps) to establish each intraresidue group of NH, Ha, and aliphatic side-chain resonances that are grouped in an individual spin system typical for each of the twenty natural amino acids. Recognizing the cross-peaks pattern and analyzing the chemical shifts of the resonances one can attribute the group of signal to a certain residue; thus identifying each one of the residues of the polypeptide sequence. Aromatic protons of tyrosine, phenylalanine, tryptophan and histidine residues as well as side-chain amide protons of glutamine and asparagine residues are not scalarcoupled to the remainder of the side chain. Consequently, association of the side-chain and backbone resonances of these spin systems has to be made on the basis of intraresidue NOE correlations. The second step of resonance assignment involves the detection of medium and long-range through-space connectivities detectable in NOESY spectra. For this reason the proton resonances assigned in TOCSY spectra, are transferred to the NOESY maps, and each individual amino acid is connected with the neighbouring, in the sequence
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
193
and/or in space, residues by using through-space dipolar coupling (NOE) interactions. Is such a way the sequential connection of the spin system could be accomplished. In preliminary steps inter-residue connectivities invlolving the NHi, Hai,, HBi, and NHi, i+1 (i+1 is the next to i residue in the sequence) could be detected and the arised crosspeaks could be assigned with, practically, no ambiguities for a polypeptide comprising up to 100 amino acids. At this point it should also be noted that statistical analysis of the proton positions inferred from X-ray crystal structures of proteins has shown that NH, Ha, and/or HB\ of residues adjacent in the primary polypeptide sequence are found in short distances one to each other, and could give rise to observable NOE signals. NMR spectra of larger proteins suffer from signal overlap and the procedure of the resonance assignment when only 1H-1H homonuclear spectra are available becomes very complicated if not unfeasible. The sequential NOEs connectivities are usually denoted with the short-hand notation dNN, daN, and dBN, respectively [12] and usually illustrated in a typical diagramm as that presented below for the case of the investigated system of a CRH analogue (see below Figure 5). Identification of a series of sequential NOE interactions could connect not only two neighboring residues together but could also result to an identified peptide fragment of the polypeptide comprised by 4, 5 or 6 residues. As gradually, more and more spin systems are added to the identified core of residues, the formed sequence will eventually match to a unique section of the primary, known a priori, ammo acid sequence. At this advanced steps the spin systems, are assigned sequence-specifically. Detection of NH-NH, Ha-NH, and HB-NH dipolar connectivities is not observed only for sequential residues but also could give well defined NOE cross-peaks between nonsequential residues as a consequence of the secondary structure and the overal folding of the polypeptide chain [13]. As mentioned above, the ambiguities in the assignment process is limited when the identification of sequential NH-NH, Ha-NH, and H(3-NH NOEs takes place since sequential residues are found, in crystals, to the right distance and the suitable orientation/position to give rise to a well defined and intense dipolar couplings. Additionally, the sequential ordering of spin systems must fit in the primary sequence and NH-NH, Ha-NH, or Hp-NH NOEs between residues that are never adjacent in the primary sequence result from longer-range (of the order i, i+j, when i=1 and j > 5) contacts. In the limit, the assignment encompass all spin systems, and self consistency is the best criterio for the validity of the acquired results [11]. The detected and assigned long range connectivities are of crucial interest for the calculation of the three-dimensional structure of biomacromolecule since they provide with structural information/constraints for the contacts between parts of the polypeptide that are distant in the primary sequence but becomes close when the chain folds, and adopts the final tertiary structure. NMR-derived Structural Restraints and Structure Determination. Chemical shifts, scalar coupling interactions, and dipolar relaxation are strong indicators of occured conformational changes in biomolecules. Analysis of these parameters yields structural information that upon the appropriate manipulation could be incorporated as NMR-derrived structural restraints to the calculation of the 3D structure of the molecule. The major volume of structural information is extracted by the interproton interactions which give rise to NOE peaks in 2D 1H homonuclear spectra. The dipolar cross-relaxation rate constant is proportional to the inverse sixth power of the distance between the interacting protons. In the initial rate approximation, NOE cross-peak
194
G. A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
intensities are proportional to the cross-relaxation rate constants. Thus, if one interproton distance, rref is known (from covalent geometry), then another, unkown interproton distance r i ,is determined by the relationship : ri = rref (Sref/Si)l/6 (1) where Sref and Si are the integrated cross peak intensities. In practice, NOESY mixing times short enough to satisfy the initial-rate approximation have the dissandvantage that yield spectra with low signal-to-noise ratios. For longer mixing times, the intensities of NOESY cross-peaks are no longer directly proportional to the cross-relaxation rate constants between the interacting spins because the magnetization is transferred between spin in multiple steps via "spin diffusion". However, the use of the appropriate algorithms and complex calculations could precisely determine the 1H-1H distances from the NOE cross-peak intensities. NOE cross peaks are usually grouped into three (according to their intensities; strong, medium, and weak) or into five (according to the nature of proton-proton interactions; intraresidue, sequential, medium range, backbone long-range and longrange) categories. Each category, regardless the protocol used considering the above mentioned three or five proton categories, is associated with an upper-bound separation between the interacting spins. The cross-peak volume limits and the upper-bound distances for the categories are estimated from the NOE intensities observed for protons of known covalent geometry (for example geminal methylene protons, or vicinal protons in aromatic rings) or between protons in regions of regular secondary structure (for example sequential NH-Ha, cross-strand Ha-Ha and NH-Hot B-sheets, or NH-Ha medium range NOEs in helices). Conservative upper-bound distances compensate for cross-peaks affected by spin diffusion or partial overlap; otherwise failure to allow for such artifacts, results in structural models that have large violations of the input restraints, high energies and in some cases artificially high precision [14]. Useful angle restraints are extracted though measurements of scalar coupling constants [11,15] either from high resolution homonuclear phase sensitive COSY, TOCSY and NOESY experiments or from the appropriate 1H-15N HN-HA [16] spectra. As first demonstrated by Karplus [17], the magnitude of a 3J scalar coupling constant is a function of the dihedral angle, 0, formed by the three covalent bonds: V = A cos2 9 + B cos 9 + C (2) where the constants A, B and C depend on the particular nuclei involved in the covalent bonds. In general, 3 J H N H a constants measured in the range 3.0 to 6.0 Hz are indicative for helical secondary structure; while values equal or above 8.0 are typical for extended, ^-strand, conformations [11,18]. Values of coupling constants in the range 6.0 to 8.0 Hz are representative for an equilibrium between different possible conformations. As stated above, the majority of the restraints used to calculate the 3D structure of a polypeptide arise from many hundreds of NOE cross-peaks transformed into upper distance bounds (called also upper distance limits) representing interproton distances. It generally holds, that the lower distance limits for each proton pair is set equal to the sum of the van der Waals radii. Distance constraints could be supplemented with dihedral angle restraints, with upper and lower limits determined from the observed scalar coupling constants and hydrogen-bond distance restraints. There are various methods that allow the calculation of a protein structure using NMR-derrived restraints. The protocols implemented to this kind of software, aim to determine coordinates for the protein atoms that will satisfy the input distance and angular restraints in an unbiased fashion while avoiding local energy minima, and
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
195
exploring all regions of conformational space, compatible with the observed NMR parameters. NMR data do not uniquely define a 3D protein structure, because the restraints are included as ranges of allowed values, the data contain experimental uncertainties, and only a sparse subset of all possible distance, angle and hydrogen bonding constraints are obserevable. Instead, the structure calculation is repeated frequently to determine an ensemble of (low-energy) structures consistent with the input data. A high quality ensemble of structures should exhibit the minimum of violations (where no consisted violation - violation observed in all the calculated structures observed) of the input restraints while the RMSD (root-mean-square-deviation) among the members of the family should be below 1.0 A for the backbone and below 2.0 A for all heavy atoms. The two most common approaches for generation of structures are distance geometry (DG) [19] and restrained molecular dynamics (MD) using the simulating annealing (SA) protocol [20]. The former determines ensembles of 3D structures consistent with an incomplete (not all distances can be quantified since NOE is limited to distances up to 5.0-5.5 A) set of distance constraints. The latter is the alternative method to DG approach where the MD force fields are supplemented by square-well pseudoenergy terms based on NMR-derived restraints [20]. The generated structure is restrained according the experimental geometric restraints toward a conformation that satisfies in maximum degree the experimental data while reduces the arised violations (and consequently the overall energy of the system/molecule) imposed from the same experimental data during the control heatup-cooldown proceedure of the SA protocol. Precision, Accuracy and Quality Assesment of the Structures. The criteria for the precision and accuracy of the calculated structure could be summarized as follows : (i) large number and variety of distance and geometric constraints (NOEs, torsion angles, H-bonds, stereospecific assigned protons/methyls); (ii) high number of experimental NOE-derived constraints (>15 per residue) and tight bound limits; (iii) RMS deviation of backbone atoms from the mean structure should be below 0.5 A for the well defined regions of secondary structure: (iv) low energy (after energy minimization - Molecular Mechanics refinment) and small residual violations, and (v) favorable backbone conformation in terms of allowed values of backbone torsion angles
196
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
principal neuroregulator of the secretion of adrenocorticotropic hormone (ACTH), 0endorphin and other proopiomelanocortin products of the anterior pituitary gland considered now as the key-hormone controlling hypothalamic-pituitary-adrenal function [23]. The CRH acts at the nanomolar level by interacting with a membrane receptor belonging to the G protein-coupled family of receptors. CRH binds to membrane homogenates from the anterior pituitary, revealing a high-affinity component of 0.1 nM and a low-affinity binding site at 20nM [24]. The primary sequence of various organisms is presented below in Chart 2. DOG HUMAN RAT PIG SHEEP
5
10
15
SEEPP SEEPP SEEPP SEEPP SQEPP
ISLDL ISLDL ISLDL ISLDL ISLDL
TFHLL TFHLL TFHLL TFHLL TFHLL
20
REVLE REVLE REVLE REVLE REVLE
25
MPGAE MARAE MARAE MARAE MTKAD
30
QLAQQ QLAQQ QLAQQ QLAQQ QLAQQ
35
AHSNR AHSNR AHSNR AHSNR AHSNR
40
KLMEI KLMEI KLMEI KLMEI KLLDI
I I I I A
Chart 2. Sequence alignment of various Corticotropin Realing Hormones. Residues that are different in the sequences of other than human CRH are noted with different character format
The analogue of the human CRH studied is a synthetic polypeptide where in positions 12 and 15 of the sequence the native amino acids L-Phenylalanine and Leucine has been substituted by a D-Phenylalanine and by the a-aminoisobutyric acid (Aib), respectively. The CRH analogue was synthesized manually by solid phase Fmoc chemistry protocols [25] using 2-chlorotrityl resin [26] as solid support and 1-hydroxybenzotriazole (HOBt), N,N'-diisopropyl-carbodiimide (DIC) as activators of carboxylic residues [27]. The peptide was cleaved from the solid support by a TFA/DCM mixture using as ion scavengers anisol. 1,2- ethandithiol and water. The purification and characterization of peptide was achieved with an AKTA purifier RP-HPLC system and ESI-MS. Purity of CRH analogue was greater than 95%. Sample preparation. Deuterated trifluoroethanols, TFE-d2 and TFE-d3 (CORTEC, Paris, France), were used in mixtures with H2O and D2O, respectively. The peptide was dissolved to a concentration 2-2.5 mM in H2O/TFE-d2 (34%/66% v/v), while the pH was adjusted to 3.8. For amide exchange experiments, the peptide was lyophilized and redissolved in D2O/TFE-d3 (34%/66% v/v). NMR experiments carried out in 298K and 31 OK. NMR spectroscopy. NMR spectra were recorded on a Bruker 500, 600 and 700 MHz spectrometers. 1D spectra on 12-14 ppm spectral width were acquired using either the "presaturation" (presaturation during the relaxation delay and the mixing time) [28] or the "watergate" (gradient tailored excitation) [29] pulse sequence for the elimination of the H2O signal (Figure 4).
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
197
8.3O19/22" 25/22
9.00
8.50
8.00
a)
7.50
7.OO
5
ppm
Figure 4. Regions (A) 1H-1H 2D TOCSY 600 MHz NMR of the fingerprint region of NH-Ha protons. (B) 1H-1H 2D NOESY 600 MHz NMR of the NH-NH protons. Both spectra recorded on a 10.7 ppm spectral width (H2O/TFE-d2 34%/66% v/v, at pH=3.8, T= 310 K).
DQF-COSY [7] and TOCSY (using MLEV-17 spin-lock sequence, mixing time of 80-100 ms, 2K data points in F2 dimension, 16-32 transients and 1024 complex increments in the Fl dimension) [5] experiments were performed in order to facilitate the identification of the spin-systems of each individual aminoacid. TPPI NOESY [6] spectra were recorded using mixing times of 100 ms and 200 ms. The other acquisition parameters were the same as for the 1D spectra (pulse width - 90°- 11.28 us, relaxation delay 1.0-1.5 s and acquisition time 157.794 ms). The spectra consisted in 2K data points in the F2 dimension, 16-32 transients and 720-1024 complex increments in the Fl dimension. All 2D maps were acquired with a spectral width of 6410.26 Hz (10.7 ppm). Raw data were multiplied in both dimensions by a pure cosine-squared bell window function and Fourier-transformed to obtain 2048x1024 or 2048x2048 real data points. A polynomial base-line correction was applied in both directions. NMR data processing was performed using the standard Bruker software package and 2D maps were analyzed on a Silicon Graphics O2 workstation or on PentiumIII PC-Linux computers with the aid of the program XEASY (ETH, Zurich) [30].
198
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
Identification of slowly exchanging amide hydrogens. Slowly exchanging amides were identified after 4-5 hours in D2O/TFE-d3 and acquisition of NOESY where nonexchangeable amide protons are giving rise to detectable NH resonances after that period. 16 hydrogen bonds (13 in 20-41 region) involving non-exchangable NH amide protons, are occured to the final set of structures (with occurance > 70%) composing the family of 40 structures were used as structural constraints and introduced in the final structural calculation. Exchangeability of amide NHs is illustrated at the top of the Figure 5. NMR Constraints. The volumes of the 2D NOESY cross peaks between assigned resonances were obtained by manual integration. 1868 dipolar connectivities concerning sequential, short medium range connectivities Figure 6. The total number of unique NOESY cross peaks was 928 and the intensities were converted into upper limits of interatomic distances by following the methodology of the program CALIBA [31] and cross peaks of NOESY spectra were classified into five categories [11]. Appropriate pseudoatom corrections were applied to methylene and methyl hydrogens that were not stereospecifically assigned [32]. Determination of scalar coupling constants. 3JHNHa were determined using the method of D. S. Wishart [15] for measuring 3JHNHa coupling constants from NOESY spectra. 21 out of 28 measured coupling constants, were measured in the range of 4.8-5.6 Hz, equal or above 8.0 [33] and introduced to the calculations (Figure 5). Seven 3JHNHa were calculated in the range 6 to 7 Hz suggesting conformational avergaging and have not been used in structural calculations [11]. Structure Calculations and Refinement. A total of 1868 NOESY cross-peaks (90% derived from the NOESY map acquired with the "PRESATURATION" pulse sequence) were integrated and transformed into 928 unique upper distance limits, corresponding to almost 23.1 constraints per assigned residue (no resonances for Ser1 has been assigned) (Figure 6). 656 constraints (16,4 per assigned residue) were found meaningfull and used in structural calculations with DYANA program [34]. The Aib (a-aminoisobutyrilo) residue was generated from a Valine where the Ca. of the Aib corresponds to CB of a Valine and, CB1/CB2 of the Aib corresponds to Cy1/Cy2, respectively, of a Valine. DPhenylalanine is generated by modifying the coordinates in one axis of an LPhenylalanine. 14 pairs of diastereotopic protons (among them the two methyl groups of Aib15) have been stereospecifically assigned through the program GLOMSA [31]. The 40 best structures (out of 400 calculated) in terms of target function with NOE constraint violations below 0.2 A (larger violation 0.09 A) are included to the final familly. Each one of the 40 DYANA models is refined by Restrained Energy Minimization (REM) through programs included to AMBER 5.0 Package [35]. The whole proceedure is set up following protocols developed and applied on solution structure determination of larger biomolecules, such as metalloproteins [36]. A force constant of 133.76 kJ mol-1 A2 is applied for the distance constraints. Structural calculations have been performed on IBM RISC6000 or on PentiumIII PC-Linux computers.
199
G.A. Spyroulias et al. / Conformational Analysis ofBiomolecules in Solution
Quality Assessment of the Structure. The structure quality was analyzed in terms of backbone conformation through the PROCHECK program [22], which provides the elements of the secondary structure, Ramanchandran plot, and other statistical parameters. Secondary structure. Figure 5 shows the short and medium range NOEs observed for the backbone and ft protons in the NOESY maps. Sequential NH-NH connectivities for stretches up to two aminoacids were observed for residues 6-13, 14-19, 20-28, 29-31, and 31–40. Two prolines (in 4 and 5 position) are present in the sequence and interrupt the sequential connectivities. Strong NH-NH and medium range Ha-NH(i, i+3), Ha-Hp (i, i+3) and Ha-NH(i, i+4) NOEs, have been observed throughout the region from Pro5 to Ile40 indicating the extended a-helical structure of the CRH analogue. (Figure 4). 2
Figure 5. (A) Schematic representation of the sequential and medium-range NOE connectivities involving NH, Ha and H3 protons. Thickness of the bar indicates the intensity of NOEs. 3JHNHa coupling constants are illustrated by arrows ( for values in the range 4.2 -5.6, for values equal or above 8.0 Hz) or by filled circles (values in the range of 6.0-7.0 Hz) The average, over the 40 DYANA structures, secondary structure elements are also reported.
Valuable information for the secondary structure of a polypeptide chain could, also, be provided by the spin-spin coupling constants which in most cases are found typical for a- and 03/10 helical structures since experimental 3J values are measured in the range of 4.8 to 5.6 [33] somewhat larger than the ideal values of 3.9 and 4.2 Hz, respectively. In Figure 5 measured V values for 28 residues are illudstrated. From those, 18 3JHNHa are found to be equal or less than 5.6 Hz (range of 4.0 to 5.6 Hz) while 3 (Glu2, Glu3 and Val18) are found to be in the range of 8.0 to 8.5 Hz, one (DPhel2) is measured 6.9 Hz and 6 (Leu10, Thr11, Leu19, Gln26, Ile39 and Ile40) exhibited values in the range of 6.0 to 6.6 Hz. Tertiary structure. The resulting DYANA family of 40 structures has RMSD values (for residues 6-41) of 0.56 ± 0.24 A and 1.40 ± 0.20 A for backbone and heavy atoms, respectively. The target function lies in the range 0.10-0.12 A2 (average target function 0.11 ± 0.01 A 2 ). The final family resulting from REM refinment performed using both NOEs and Hydrogen bond constraints exhibits pairwise RMSD values for the 40
200
G.A. Spyroulias et al. / Confortnational Analysis of Biomolecules in Solution
structures 0.56 ± 0.23 A and 1.42 ± 0.19 A and 0.39 ± 0.16 A and 0.99 ± 0.13 A to the mean structure for backbone and heavy atoms, respectively. The RMSD per residue for the final family and the NOE-derived distance constraints are reported in Figure 6. Figure 7 shows the family of 40 structures for all heavy atoms (A) and for the backbone atoms (B). The polypeptide chains seems to fold to a rather linear helical structure. Features, in terms of NOEs, of the helix has been observed for the residue fragment from Ile6 to Ile41, while for the mobile, ill-defined N-terminus data (chemical shift index, 3J/HNHa, and 9 versus ^ plots) imply a conformational preference to extended, p sheet, secondary structure.
Figure 6. Number of all NOE constraints per residue (top) and number of meaningful NOE constraints (middle) per residue for [DPhe12, Aibl5]CRH used in the structural calculations. White, gray, and dark gray bars represent respectively intraresidue, sequential, and medium-range, connectivities. Long-range connectivities have not been observed. RMSD per residue (bottom) for the family of 40 structures for backbone (dark gray bars) and heavy atoms (light gray bars).
Beyond the structure : Biological Implications. A noteworthy characteristic of the [DPhe12, Aib15]CRH is the bend of the helical structure at the region 12-16 which is not observed for the native peptide [37]. This bend is supported by numerous NOEs at the 10-20 region and this distortion out of linearity calculated in the basis of helix dipole for
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
201
the peptide fragments 6 to 16 and 20 to 40 and found to be 34.2°. This is probably the result of the presence of a D- and an a,a-dialkyl aminoacids in close proximity in a helical polypeptide chain. Another interesting feature of this region of the threedimensional structure of the molecule, is that the chain at the region 12-15 folds in a helical structure in such a way that the turn formed comprised by three instead of four residues bringing the aromatic ring of the DPhel2 and the one methyl group of Aibl5 in close proximity while the subsequent four-residue turn in the region 15-19 allow the other methyl group of AiblS to approach the main- and side-chain of Leu 18 (Figure 7). Serl
Ile41
N terminus
terminus
Aibl5
Met21
Met38
Figure 7. The family of 40 structures calculated for the [DPhe12, Aib15]CRH analogue in line (all heavy atoms) and in saucage (backbone) representation. Figure was generated with MOLMOL [38].
The number, nature and density of charged residues in the surface of proteins or biomolecules in general, is of crucial importance, since electrostastic interactions possess a central role in numerous biological processes. The knoweledge of the nature and the strength of such intramolecular interactions requires the detailed analysis of the structural features of the biomolecules. In the synthetic CRH-analogue, as well as in the native polypeptide, there are eleven charged residues without taken into account the NH+3 group of the N-terminus. Seven, out of the eleven, residues are negatively charged while only four are positively charged resulting to a net negative charge of the molecule. Analysis of the charge distribution along the main axis of the almost linear and helical, peptide indicates that the 20-peptide of the N-terminus bears 5 negatively and 1 positively charged residues, while the second half of the molecule towards the C-
202
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
terminus exhibits a slight excess in positively charged residues (two Arg+ and one Lys+ versus two Glu-) giving the character of a dipole to the molecule with the negative pole at the N-terminus and a rather positive pole at the C-terminus. Calculation of the distribution of the electrostatic potential on the surface of the molecule reveals the amphipathic character of the helix and the non-equivalent distribution of the charges in each side of the helix. The five out of six negatively charged Glutamates existing in the sequence of CRH are oriented in a different way than the sixth, Glu20, residue. The three of them are clearly oriented in the opposite direction than that of Glu20 while the two others are found in the intermediate space between the two extreme orientations manifested by Glu20 on one side and by Glu3, 17 and 25 on the other, exhibiting though a tendecy to orient towards the side of the three Glutamates. This density of negative charges is slightly diminished by the presence of two consecutive, positively charged, residues at the C-terminus, Arg35 and Lys36, where the lysyl residue extent its long side chain bearing the amino group, to the same direction as the three Glutamates do. Arg35 exhibits rather the same orientation as that of its side chain which ends to a positively charged group and orients in direction that differs about 90° with that of the side chains of Glu3, 17, 25 and Lys36. On the other hand there are three additional residues that orients towards or similar to the Glu20. From these the two positevely charged arginines at positions 16 and 20 extent their side chains far from the helix axis, to the solvent, exactly on the opposite side form the position of the three glutamates (Glu3, 17, 25) and Lys36. Concluding Remarks The detailed conformational study of a represenative synthetic analogue of Corticotropin Releasing Hormone led to the solution structure determination of [D-Phe12 , Aib'5]CRH in high resolution. Detailed analysis of the structural features attempts to gain valuable insights of the impact of side-chains belong to residues foreseen to possess a key-role in molecular recognition process, binding and activity properties. Substitution of the natural amino acids at a close proximity to the peptide fragment 12-15 by non natural residues, seems to introduce a bend across the molecule which has not been observed in the solution structure of the native peptide. Although hard to quantitatively evaluate the outcome of the new structural features observed for each of the two new residues introduced to the polypeptide chain at positions 12 (DPhe) and 15 (Aib), the experimental results indicate that the DPhe seems to destabilize the helical secondary structury while Aib seems to lead to a more constrained helix with features between the a- and the 0 3 /10 helices. The distribution of the electrostatic potentials on the surface of the peptide could provide valuable information not only for the properties of the molecule and the intramolecular interaction with the CRH receptor, but also towards the design and the synthesis of new molecules with higher binding activity and possibly enhanced stability. Since it is reported that the side chains of the lipophilic part of the helix at the peptide fragment 6 to 20 are playing crucial role to binding and activation of the receptor, increase of the lipophilic character of this region would be designed with the extinction of the negative or positive charge density, or both. Rational design of new CRH analogues through substitution of natural residues by non natural is being carried out at our laboratory and synthesis of new peptides bearing residues that increase the action period of the molecule, and increase the binding and activity of the synthetic peptides are under way.
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
203
Acknowledgments P.C. acknowledges the financial support of General Secretariat of Research and Technology of Greece through the EKVAN (EPET II) Programme (Project 66, 19982001). G.A.S. wishes to acknowledge a European Union HP Fellowship (Marie Curie Research Training Grant, Contract No. HPMF-CT-1999-00344). We, also, wish to acknowledge European Community's Programme Access to Research Infrastructures Action of the Improving Human Potential Programme Contract No. HPRI-CT-199900009 and the NMR-PARABIO LSF at University of Florence for the provided access to NMR Instrumentation (500, 600 and 700 MHz), as well as for the access to powerful computational systems in order to run structural calculations. References [I] F.C. Bernstein, T.F. Koetzle, G.J.B. Williams, E.F. Meyer, M.D. Brice, J.R. Rodgers, O. Kennard, T. Simanoucghi, J. Mol. Biol, 112 (1977) 535. [2] G. Wider, K. Wiithrich, Curr. Opin. Struct. Biol., 9 (1999) 594. [3] R.R. Ernst, G. Bodenhausen, A. Wokaun, In: R. Breslow, J. Halpern Frs, J.S. Rowlinson Frs (eds.),Principles of Nuclear Magnetic Resonance in One and Two Dimensions. Clarendon Press, Oxford, 1987. [4] (a) J. Jeener, In: Ampere International Summer School. Basko Polje, Yugoslavia (1971); (b) W.P. Aue, E. Bartholdi, R. R. Ernst, J. Chen Phys. 64 (1975) 2229. [5] A. Bax, D.G. Davis, J Magn Reson 65 (1985) 355. [6] (a) D. Marion, K. Wiithrich, Biochem. Biophys. Res. Commun., 113 (1983) 967; (b) J. Jeener; B.H. Meier; P. Badimann; R. R. Ernst, J. Chem. Phys., 71 (1979) 4546. [7] U. Piantini; O. W. Sorensen; R. R. Ernst, J. Am. Chem. Soc., 104 (1982) 6800. [8] D. Wishart, B. Sykes, F. Richards, J. Mol. Biol., 222 (1991) 311. [9] D. Wishart, B. Sykes, F. Richards, Biochemistry, 31 (1992) 1647. [10] W. J. Chazin, P. E. Wright,J Mol. Biol., 202 (1988) 623. II1] K. Wuthrich, In: NMR of Proteins and Nucleic Acids. Wiley, New York, 1986. [12] K. Wuthrich, Biopolymers, 22 (1982) 131. [13] M. Billeter, W. Braun, K. Wuthrich, J. Mol. Biol, 155 (1982) 321. [14] T. Havel, In: V. Renugopalakrishnan, P.R. Carey, I.C.P. Smith, S. Huans and A.L. Storer (eds.), ESCOM Science Publishers, Leiden, Holland. 1991, pp. 110–115. [15] Y. Wang, A. M. Nip, D. S. Wishart, J. Biomol. NMR, 10 (1997) 373. [16] G.W. Vuister, A. Bax, J. Amer. Chem. Soc., 115 (1993) 7772. [17] M. Karplus, J. Phys. Chem. 30 (1959) 11. [18] L.J. Smith, K.A. Bolin, H. Schwalbe, M.W. MacArthur, J.M. Thorton, C.M. Dobson, J. Mol. Biol., 255(1996)494. [19] G.M. Crippen, T.F. Havel, In: Distance Geometry and Molecular Conformation. Research Studies Press, Taunton, UK, 1988. [20] (a) A.T. Brunger, G.M. Clore, A.M. Gronenborn, M. Karplus, Proc. Natl. Acad. Sci. (USA), 83 (1986) 3801; (b) G.M. Clore, A.T. Brunger, M. Karplus, A.M. Gronenborn, J. Mol. Biol, 191 (1986) 523. [21] B. Borgias, P.D. Thomas, T.L. James, Complete Relaxation Matrix Analysis (CORMA). U of California, San Francisco, USA 1989. [22] (a) R.A. Laskowski, M.W. MacArthur, D.S. Moss, J.M. Thornton, J. Appl. Crystallogr., 26 (1993) 283; (b) R.A. Laskowski, J.A.C. Rullmann, M.W. MacArthur, R. Kaptein, J.M. Thornton, J. Biomol. NMR, 8(1996)477. [23] (a) W. Vale, J. Spiess, C. Rivier, J. Rivier, Science, 213 (1981) 1394; (b) W. Vale, C. Rivier, M.R. Brown. J. Spiess, G. Koob, A. Swanson, Recent Prog. Horm. Res., 39 (1983) 245. [24] E.B. De Souza, T.R. Insel, M.H. Perrin, J. Rivier, W.W. Vale, M.J. Kuhar, Neurosci. Lett., 56 (1985) 121.
204
G.A. Spyroulias et al. / Conformational Analysis of Biomolecules in Solution
[25] (a) L.A. Carpino, G.Y. Han, J. Org. Chem., 37 (1972) 3404; (b) D. Sarantakis, J. Teichman, E.L. Lien, R.L. Fenichel, Biochem. Biophys. Res. Commun., 73 (1976) 366. [26] K. Barlos, D. Gatos, J. Kallitsis, G. Papaphotiou, P. Sotiriou, Y. Wenquing, W. Shafer, Tetraedron Lett., 30 (1989) 3943. [27] (a) D. Hudson, D. Kain, D. Ng, In: Y. Kiso (ed.), Peptide Chemistry. Protein Research Foundation, Osaka, 1986, pp. 4113; (b) W. Konig, R. Geiger, Chem. Ber., 103 (1970) 788; (c) Konig, R. Geiger, Chem. Ber., 103 (1970) 2024; (d) Konig, R. Geiger, Chem. Ber., 103 (1970) 2034. [28] S. Macura, K. Wuthrich, R.R. Ernst, J Magn. Reson., 47 (1982) 351. [29] M. Piotto, V. Saudek, V. Sklenar, J. Biomol. NMR. 2 (1992) 661. [30] C. Eccles, P. Guntert, M. Billeter, K. Wuthrich,J. Biomol. NMR, 1 (1991) 111. [31] P. Guntert, W. Braun, K. Wuthrich, J. Mol. BioL. 217 (1991) 517. [32] K. Wuthrich, M. Billeter, W. Brown, / Mol. Biol. 169 (1983) 949. [33] L.J. Smith, K.A. Bolin, H. Schwalbe, M.W. MacArthur, J.M. Thorton, C.M. Dobson, J. Mol. Biol., 255(1996)494. [34] P. Guntert, C. Mumenthaler, K. Wuthrich, J. Mol. Biol, 273 (1997) 283. [35] D.A. Pearlman, D.A. Case, J.W. Caldwell, W.S. Ross, T.E. Cheatham, D.M. Ferguson, G.L. Seibel, U.C. Singh, P.K. Weiner, P. A. Kollman, AMBER 5.0. U of California, San Francisco, USA. 1997. [36] L. Banci, I. Bertini, C. Luchinat, P. Turano, Solution structures of hemoproteins. In: K.M. Kadish, K.M. Smith, R. Guilard, (eds.), The Porphyrin Handbook. Academic Press, 2000, Volume 5, pp 323– 350. [37] C. Romier, J.-M. Bernassau, C. Cambillau, H. Darbon, Protein Eng., 6 (1993) 149. [38] R. Koradi, M. Billeter, K. Wuthrich, J. Mol. Graphics, 14 (1996) 51.
Drug Discovery and Design: Medical Aspects J, Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
205
Synthesis and Structural Investigation of a Synthetic LHRH Analogue in Solution A. A. Zompra, G. A. Spyroulias, V. Magafa and P. Cordopatis Department of Pharmacy, University of Patras, 26504 Patras, Greece
Abstract. The pivotal role that LHRH and its analogues play in the modulation of reproductive functions have attracted considerable scientific interest because of their usefulness in the treatment of endocrine-based diseases such as prostate cancer, breast cancer, endometriosis and precocious puberty. Several LHRH agonists, represented by Leuprolide ([DLeu6, desGly10]-LHRHethylamide), are currently used in the treatment of the above conditions. The goal of this research is the synthesis and conformational analysis of a LHRH analogue, such as [Aib6, desGly10]-LHRH-ethylamide in order to gain valuable insights on bioactive conformation and use these for the design of further analogues. This LHRH analogue was synthesized by the solid phase methology on a [3-((Ethyl-Fmoc-amino)-methyl)-l-indol-l-yl]-acetyl AM resin via Fmoc/tBu methology. ID and 2D NMR experiments were carried out in 2-2.5mM solution of [Aib6, desGly10]-LHRH ethylamide in DMSO-d6. The average structure of LHRH analogue was calculated from the family of the 20 models and refined using NOE-derived structural information.
Introduction The hypothalamic decapeptide luteinizing hormone-releasing hormone (LHRH) (pGluHis-Trp-Ser-Tyr-Gly-Leu-Arg-Pro-Gly-NH2) plays a key role in the control of mammalian reproduction. It is secreted in pulses from the hypothalamus and stimulates the anterior pituitary gland to release the gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Ultimately, both of these hormones elicit gonadal production of sex steroids and gametogenesis [1-4]. By modification of the molecular structure of this decapeptide, analogues were synthesized with agonistic or antagonistic effects on the gonadotrophic cells of the anterior pituitary gland. The agonists, after an initial stimulatory effect ('flare up'), lead
206
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
to desensitization of the gonadotrophic cells and a reduction in the number of LHRH receptors on the cell membrane ('down-regulation') [5], while the antagonists produce an immediate effect by competitive blockade of the LHRH receptors. Oncological uses of LHRH analogues are based on the inhibition of the pituitary gonadal function, the suppression of gonadal steroid hormone secretion leading to inhibition of the growth of sex hormone dependent tumors [6,7]. However, direct effects of LHRH analogues on tumor cells may also play a role [8]. There is evidence that specific membrane receptors for LHRH are present in various tumors, such as human prostate and breast cancer [811], ovarian [13, 14], endometrial [12-14], and pancreatic cancers [4, 15). In patients afflicted with these malignancies, conventional chemotherapy results in a varying degree of response and is associated with severe toxic side effects [6]. Agonistic analogues of LHRH, represented by Leuprolide ([DLeu6, desGly10]LHRH-ethylamide), have been widely used in oncology and gynecology for nearly two decades. Leuprolide (hCAS: 53714-56-0; Leuprolide or Leuprorelin) and Leuprolide Acetate (hCAS: 74381-53-6; Leuprorelin Acetate) are synthetic structural analogues of LHRH. Leuprolide presents a higher affinity for the LHRH receptors and a higher resistance to enzymatic degradation than the natural hormone, which increases its biological potential. It can thus be used for the treatment of a large number of diseases related with the regulation of sexual hormones, such as masculine and feminine infertility, uterine myomas and prostatic and mammalian tumors [16, 17]. It was revealed many years ago that substitution of the Gly amino acid residue at position 6 of LHRH by various D-amino acids results in very potent analogues of LHRH with high binding affinity [18]. In our early attempts to create agonistic analogue of LHRH, Aib (a-aminoisobutyric acid) residue, which is known to favor the formation of a turn when inserted in the peptide sequence [19-21], was incorporated at position 6 of LHRH. Key considerations in the synthesis of the above LHRH analogue were on the basis that the bioactive conformation of LHRH includes a type II ß-tum involving residues 5-8 {Tyr5-Gly6-Leu7-Arg8} [22]. Also, in previous studies, we had synthesized Leuprolide using the solid phase Fmoc/tBu methology on a Sieber Ethylamide resin (Ethylamino-xanthen-3-yloxy-Merrifield resin) in 30% overall yield. Conformational analysis of this molecule suggested the existence of a loop structure in the region of Tyr5-DLeu6 residues [23]. In this study, we report an improved synthesis (higher yield) and the conformational analysis in solution of the LHRH analogue namely [Aib6, desGly10]LHRH-ethylamide using NMR spectroscopy, in order to gain valuable insights on bioactive conformation and use these for the design of further synthetic analogues (agonists and/or antagonists) of LHRH. Experimental Section Materials: 9-Fluorenylmethoxycarbonyl-protected amino acids and peptide reagents were obtained from Bachem AG and Novabiochem. All solvents and reagents used for solid-phase synthesis were of analytical quality. Dimethylsulfoxide DMSO-d6 (MERCK) was used as deuterated solvent in NMR experiments and the peptides were dissolved to a final concentration of 2-2.5 mM in order to record ID and 2D NMR spectra.
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
207
Peptide Synthesis and Purification: [Aib6, desGly'°]-LHRH-ethylamide was assembled on a [3-((Ethyl-Fmoc-amino)-methyl)-l-indol-l-yl]-acetyl AM resin [24] containing 0.85 mequiv. of amino group/g with the protecting groups shown in parentheses (pyroglutamic acid-His(Trt)-Tip(Boc)~Ser(tBu)-Tyr(tBu)-Aib-Leu-Arg(Pbf)-Pro-NHEt). Chain extension was carried out using standard Fmoc (9fluorenylmethyloxycarbony]) protocols [25]. In summary, the Fmoc group was removed with 25% piperidine in N,N-dimethylformamide (DMF). Activation of each amino acid was performed in situ using diisopropylcarbodiimide/l-hydroxy-7-azabenzotriazol DIC/HOAt in DMF [26]. Completeness of the reaction was monitored by Kaiser test [27]. Treatment of the peptidyl resin with trifluoroacetic acid/ methylene chloride/1,2ethanedithiol/anisole/water (90:5:1:2:2, v/v) (15 ml/g peptide resin) for 4 hours afforded the desired product. The peptide was precipitated upon evaporation in vacuo and addition of ether. The crude peptide was purified by gel filtration chromatography on Sephadex G-15 using 15% acetic acid as the eluent. Final purification was achieved by preparative HPLC (Pharmacia LKB-2250) on re versed-phase support C-18 with a linear gradient from 10 to 80% acetonitrile (0.1% TFA) for 40 min at a flow rate 2.0 ml/min and UV detection at 220 and 254 nm. The appropriate fractions were pooled and lyophilized. Analytical HPLC (Pharmacia LKB-2210) equipped with a C-18 Phase Sep column S3 ODS2 produced single peak with at least 98% of the total peptide peak integrals. Electro-spray MS was in agreement with the expected results. The desired product was in 44.5% overall yield. NMR Spectroscopy: ID NMR and 2D NMR spectra were recorded at the temperature of 298K on a Bruker Avance 400 spectrometer operating at 400.13 MHz. ID spectra over the full spectral width (12 ppm) were acquired with and without presaturation of the H2O signal and a recycle delay of 0.8 sec or 1.0 sec. DQF-COSY [28] and TOCSY [29] experiments were performed in order to facilitate the identification of the spin-systems of each individual amino acid. TOCSY experiments were carried out by using the MLEV-17 spin-lock sequence and a mixing time of 80-100 ms in both temperatures, consisted by 2K data points in F2 dimension, 16-32 transients and 1024 complex increments in the Fl dimension. TPPI NOESY [30] spectra were recorded using the same spectral width as in the case of ID spectra (12 ppm). The mixing times varied from 200 ms to 800 ms. Spectra recorded with mixing time above 400 ms were used in order to facilitate the resonance assignment. The suppression of the residual water signal was carried out by its presaturation during the relaxation delay and the mixing time using the [31] "PRESATURATION" pulse sequence of the standard BRUKER library. The other acquisition parameters were the same as for the ID spectra. The spectra consisted of 2K data points in the F2 dimension and IK experiments in the Fl dimension. Raw data were multiplied in both dimensions by a pure cosine-squared bell window function and Fourier-transformed to obtain 2024x1024 real data points. A polynomial base-line correction was applied in both directions. NMR data processing was performed using the standard Bruker software package on a Silicon Graphics O2 workstation. The 2D maps were analyzed on a Silicon Graphics O2 workstation or on Pentium III PC-Linux computers with the aid of the program XEASY (ETH, Zurich) [32].
208
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
Results and Discussion NMR Constraints: The various protons are classified into 5 categories (according to classification by Wuthrich) [33] and their distance-to-volume correlation is calibrated according to the following scheme: intraresidue except HN, Ha, Hß- number of peaks 85 (8 methyl peaks) sequential and intraresidue HN, Ha, Hß - " 75 medium range " 14 long range backbone " 1 long range" 59 (10 methyl peaks) Total number of cross peaks 234 (18 methyl peaks). The above-mentioned upper distance limits were used as input data (structural constraints) for the structural calculations with program DYANA (Dynamics Algorithm for Nmr Applications) [34]. Structure Calculations and Refinement: A total of 456 NOESY cross-peaks (90% derived from the NOESY map acquired with the "PRESATURATION" pulse sequence) were integrated and transformed into 234 unique upper distance limits (Figure 1). From those, 149 constraints (14,9 per assigned residue) were found meaningful and used in structural calculations with DYANA program [35]. The Aib residue was generated from a Valine where the Ca of the Aib corresponds to Cß of a Valine and, Cß1/Cß2 of the Aib corresponds to Cyl/Cy2, respectively, of a Valine. The 20 best structures (out of 300 calculated) in terms of target function with NOE constraint violations below 0.2 A (largest violation 0.255 A) are included to the final family. Each one of 20 DYANA models, is refined by Restrained Energy Minimization (REM) through programs included to AMBER 5.0 Package [36]. A force constant of 133.76 kJxmol 1 A is applied for the distance constraints. Structural calculations have been performed on IBM RISC6000 or on Pentium ITJ PC-Linux computers.
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
209
Secondary Structure: Figure 1 shows the short and medium range NOEs observed for the backbone and ß protons in the NOESY spectrum.
4
6 Sequence
Figure 1: (A) Sequential dipolar connectivities involving NH, Ha, and Hß protons in [Aib6, desGly10]LHRH-ethylamide), (B) The number of observed experimental NOEs per residue. White bars indicate intraresidue NOEs, light gray bars, sequential NOEs, and dark gray and black, medium and long range NOEs, respectively.
Sequential NH-NH are observed throughout the region of residues 1 to 8. HaHN connectivities are also observed in the peptide fragment 1 to 6, while interruption of this kind of connectivities is due to the presence of Aib residue at position 6, which lacks a protons. Additional Ha-HN connectivities are observed for the terminal tripeptide between residues Leu7, Arg8 and Pro9 with the NH-Et. Hß-HN are detected for the peptides fragments 1 to 6 and between the protons of Pro9 and NH-Et terminus. HN-HN couplings for stretches up to two residues are detected for the fragment of residues Ser - Pro9, while coupling of the same range (stretches up to two residues) of the type Ha-HN are detected for the region 1 to 7. Only one Ha-HN type of coupling
210
A.A. Zompra et at. /A Synthetic LHRH Analogue in Solution
between residues that sited three positions apart at the peptide sequences is observed and concerns the residues Tyr5 and Arg8. Tertiary Structure: Numerous NOE signals are detected between main- and side- chain protons in the peptide fragments consisted by residues 3-6 and 5-8. The fact that the pairs of residues Trp3/Aib6 and Tyr5/Arg8 are found in short distance apart giving rise to NOE contacts, could be justified by the existence of bend occurred in the middle of the sequence of the peptide chain. Indeed, the skeleton of the peptide, as calculated using NMR data, found to form aß turn at the region covered by residues Ser4 up to Arg8. Table 1: Restraint violations, structural and energetic statistics for the solution structure of [Aib6, desGly'°]-LHRH-ethylamide)
2 (20 structures)
RMS Violations per experimental distance constraint (A) Intra-residue-34 0.0304 ± 0.0046 0.0334 Sequential-76 0.0192 ± 0.0017 0.0240 Medium range-37 0.0128 ± 0.0053 0.0231 Long range-2 0.0416 ± 0.0250 0.1068 Total-149 0.0219 ± 0.0021 0.0289 Average number of Violations per structure Intra-residue 1.10 ± 0.30 1.00 Sequential 3.95 ± 0,67 6.00 Medium 1.10± 0.54 3.00 Long range 0.75 ± 0.43 1.00 Total 6.90 ± 0.83 11.00 Average number of NOE 0.00 ± 0.0 00.0 violations larger than 0.3
_A
Average number of NOE 2.15± of violations between 0.1 -0.3 A Largest NOE violation 0.26
0.7
5.00 0.20
Average distance penalty 2.75 ± 0.4 4.25 function (A2) REM indicates the energy-minimized family of 20 structures. 2 is the energy-minimize! energy-minimized average structure obtained from the coordinates of the 20 individual REM structures. Moreover, detected signals between the amide protons of the Trp3 and the terminal Ethylamide clearly indicates that the two termini of the peptide are in close proximity. Thus, the whole molecule is characterized by a U-shape tertiary structure. The resulting calculated DYANA family of 20 structures are characterized by high resolution and precision due to the large number of NOE constraints used. From the 234 upper distance limits (23.4 constraints per residue) resulted from the volume-todistance conversion of the NOE intensities, the 149 were found meaningful and used in molecular dynamic calculation. This number of constraints correspond to 14.9 meaningful distance constrain per residue of the LHRH analogue. The pairwise RMSD
21
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
values are calculated for the region 2 - 9 are 0.31 ± 0.14 A for the backbone and 1.36 ± 0.59 A for the heavy atoms. The RMSD values for the mean structure compared to the 20 models of the family are 0.23 ± 0.07 A for the backbone and 1.02 ± 0.04 A for all the heavy atoms. At Table 1 statistical data concerning the calculation and the quality of the 20 models and the mean structure after the energy minimization procedure, are reported.
Leu7
NEt10"
NEtlO
Figure 2: Backbone of the best 20 structures of [Aib6, desGly'°]-LHRH-ethylamide. Side chains of Trp3, NEt10 (left) and Ser4, Leu7 (right) are also shown.
The backbone of the family, together with the side chains of the Trp3 and the ethylamide terminus, of 20 energy-minimized structures are presented at Figure 2. The conformation of the backbone and the orientation of the indole ring of Trp3 have been calculated with high resolution as indicated by the low RMSD values and illustrated at Figure 2. The bulky indole ring of Trp3 is found apart from the backbone of the peptide region 7-10, while the two termini of the peptide are in close distance. As shown at Figure 2 (left) the amide protons of Trp3 and NH-Et are found in distance that varies from 2.7 to 3.3 A among the family of 20 models. Additionally, the side chains of Ser4 and Leu7, which are sited at the ß turn region of the molecule, orient towards the same direction while approach one each other (Figure 2, right). Validation of the Structure: In order to validate our experimental results in terms of calculated structures based on NOE-derived distance constraints, we attempted to recalculate the structure in absence of two specific distance constraints. These constraints might play a fundamental role in the ß turn formation and the convergence of the two termini. In other words, we wish to elucidate whether the calculated structures are biased, or not, to form the characteristic ß-turn, by two structural constrains which are derived by NOE signals between protons of the two termini. More specifically, well-defined NOE cross-peaks are detected between the Hel imidazole 7 proton of His and the Ha backbone proton of Arg8 , in accordance with an intense NOE signal involving the two amide protons of Trp3 and NH-Et. Therefore, structural
212
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
calculations were performed with a set of distance constraints that lacks only these two distances that concerns the above-mentioned proton NOEs. The resulted structures are of slightly lower resolution (higher RMSD values) due to the higher degree of conformational/orientational freedom that possesses the two termini, in the absence of these two distance constraints. The RMSD values are calculated 0.77 ± 0.35 A for the backbone and 1.76± 0.55 A for all the heavy atoms. The final 20 best models (in terms of dynamic energy and residual distance violations) are divided in two subfamilies; one consisted by 9 models and the other that consisted by 11 models (Figure 3).
Trp3
NEt10 pGluI Figure 3: Backbone of the best 20 models calculated without considering structural data for the long range NOEs.
Although the backbone atoms of the first and last residues of the peptide sequence in the subfamily of the 11 models might be in a somewhat larger distance than in the second subfamily (see also Figure 4) as well as in the originally calculated structures (using all the experimental distances) the resulting tertiary structure of the peptide is rather similar. These findings are leading to the following conclusions: Concerning the relative conformation of the two terminal parts of the peptide, the fact that two different conformational preferences are calculated without the use of the NOE constraints between the His2-Arg8 and Trp3-NHEt, can not rule out a possible convergence of the two termini, as found at the originally calculated structures. The tertiary structure of the peptide does not differ considerably in the absence of these strategic NOEs, and the molecule does not adopt a more linear structure while the ß turn observed in the region 4-8 is not collapsed. Surprisingly, the two fragments of the ß turn structure are found in a distance even smaller than in the original structure (see also Figure 4). This is manifested by the distance between the amide nitrogens of the residues Ser4 and Arg8 monitored for the members of each one of the two families of the 20 calculated models (Figure 4A). In 19 out of 20 models the HN-HN distance is found shorter for the models calculated without the two distance constraints.
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
0
2
213
4 6 8 10 12 14 16 18 20 Structure number (20 best models DYANA) N H 3 - N H 1 0 — •— D Y A N A w i t h n o e HN3-HN10 NH3-NH10 D Y A N A w i t h o u t n o e HN3-HNH)
10,0 9,0 -
0
2
4 6 8 10 12 14 16 18 20 Structure number (20 best models DYANA)
Figure 4: The range of the distances between the nitrogens of the backbone for the Ser4 and Arg8 residues (above), and amide protons of the Trp3 and NEt10 (below) among the family of 20 models with or without the distance constraints derived from the NOE signals between residues 2/8 and 3/10.
The typical ß turn structure calculated for the peptide fragment 4-8 based on numerous interesidue NOEs is possibly the result of the introduction of Aib residue in the position 6 of the sequence. Additionally, it is noteworthy that this structure could also be stabilized by the existence of several hydrogen bonds found in the calculated structures, involving the amide HN protons of Leu7 and Arg8 with the carbonyl oxygen atoms of Ser4 and Aib6, respectively. Figure 5 presents the backbone of the mean, energy minimized, structure of the peptide [Aib6, desGly10]-LHRH-ethylamide, with the characteristic fi turn and the orientation of the side chains of all the residues.
214
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
.Arg8
(C) Figure 5: (A) Backbone of the mean energy minimized structure of the [Aib6, desGly10]-LHRHethylamide, (B) Mean structure and conformation of the side chains of residues pGlu1,Tip3, Ser4, NEt10.Van der Waals radius of heavy atoms are also shown for these residues, (C) Mean structure of [Aib6, desGly10]-LHRH-ethylamide with all atoms. Figures are generated with MOLMOL [37].
Biological implications: The molecule of [Aib6, desGly10]-LHRH-ethylamide belongs to the family of Leuprolide synthetic analogues, which have demonstrated noticeable biological activity in the treatment of sex hormone dependent tumours. Consequently, the profound study of its structure-activity relationship is of immense interest and the detailed analysis of the structure and its features/properties could significantly contribute to the design and synthesis of new synthetic analogues. Such molecules, peptides or not, should have high binding affinity towards the target receptors and form stable receptor-peptide molecular complexes. For this reason the knowledge of the distribution of the electrostatic potentials on the surface of the molecule is essential. The present synthetic analogue has no charged end-residues due to the presence of pyroglutamic acid to the position 1 of the sequence and NH-Et to the carboxyl terminus of the peptide. The only charged residue in the sequence is the Arg8; thus the overall charge of the molecule is +1. At Figure 6 the distribution of the electrostatic charges on the surface of the peptide is illustrated. The positive charge density arisen from guanidine group is rather localized at the side chain of the Arginine residue and to the backbone of the proximal residues. The side chain of Arginine is oriented towards the exterior of the molecule and due to its flexibility could play a certain role in intra-molecular, receptor-substrate, recognition processes. The rest of the molecule is characterized mainly by its hydrophobic character (white colour = no charge, blue colour = positive charge) due to the presence of the aromatic residues, Histidine, Tryptophane and Tyrosine at positions 2, 3 and 5, respectively.
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
215
Figure 6: (A) Mean, energy minimized, NMR structure and Van der Waals surfaces for His2, Trp3, Tyr5 and Arg8 residues, (B) Distribution of the electrostatic potentials on the surface of the mean structure of [Aib6, desGly'°]-LHRH-ethylamide. Figures are generated with MOLMOL [37].
Perspectives: [Aib6, desGly10]-LHRH-ethylamide has not been yet tested for the biological activity and binding affinity on the LHRH receptor, but having gone through design, synthesis and analysis of this molecule suitable model compounds that fulfil the structural requirements for a potent LHRH agonist or antagonist will be designed and synthesized. More specifically, the present conformational studies are expected to provide the structural basis for the development of new synthetic analogues (agonists and/or antagonists) through rational modification of the physical properties, such as hydrophobic/hydrophilic character, number and position of charged residues, while monitoring their binding affinity and biological activity. Synthesis of new analogues varying in positions 6, 8 and 10 are currently under way in our laboratory. References [1] H. Matsuo, Y. Baba, R.M.G. Nair, A. Arimura & A.V. Serially, Biochem. Biophys. Res. Commun., 43 (1971)1334. [2] Y. Baba, H. Matsuo & A.V. Schally, Biochem. Biophys. Res. Commun., 44 (1971) 459. [3] P.M. Conn, J.A. Janovick, D. Stanislaus, D. Kuphal & L. Jennes, In: Vitamins and Hormones, (G. Litwack, ed.). Academic Press, New York, 1995,Vol. 50, pp 151–214. [4] A.V. Schally & A.M. Comaru-Schally, Hypothalamic and other peptide hormones. In: Cancer Medicine, (J.R. Holland, E Frei III, R.R. Jr. Bast, D.E. Kufe, D.L. Morton & R.R. Weichselbaum, eds.), Williams and Wilkins, Baltimore, 1997, edn. 4, pp 1067–1086. [5] P.E. Belchetz, T.M. Plant, Y. Nakai, E.J. Keogh & E. Knobil, Science, 202 (1978) 631. [6] A.V. Schally, A. Nagy, K. Szepeshazi, J. Pinski, G. Halmos, P. Armatis, M. Miyazaki, A.M. ComaruSchally, T. Yano & G. Emons, In: Treatment with Gn-RH Analogs: Controversies and Perspectives, (M. Filicori & C. Flamigni eds.), Parthenon, Carnforth, U.K., 1996, pp. 33–44. [7] G. Emons & A.V. Schally, Human Reproduction, 9 (1994) 1364. [8J A. Qayum, W. Gullick, R.C. Clayton, K. Sikora & J. Waxman, fir. J. Cancer, 62 (1990) 96. [9] M. Fekete, T.W. Redding, A.M. Comaru-Schally, A.E. Pontes, R.W. Connelly, G. Srkalovic & A.V. Schally, Prostate, 14 (1989) 191. [10] A.V. Schally, A.M. Comaru-Schally & D. Gonzalez-Barcena. Biomed. Pharmacother., 46 (1992) 465. [11] M. Fekete, J.L. Wittliff. & A.V. Schally, J. din. Lab. Anal., 3 (1989) 137.
216
A.A. Zompra et al. /A Synthetic LHRH Analogue in Solution
[12] G. Emons & A.V. Schally, Hum. Reprod., 9 (1994) 1364. [13] G. Emons, O. Ortmann, M. Becker, G. Irmer, B. Springer, R. Laun, F. Holzel, K.D. Schulz & Schally, A.V., Cancer Res., 53 (1993) 5439. [14] G. Emons, B. Schroder, O. Ortmann, S. Westphalen, K.D. Schulz & A.V. Schally, J. Clin. Endocrinol. Metab., 77 (1993) 1458. [15] A.V. Schally, S. Radulovic & A.M. Comaru-Schally, In: Experimental and Clinical Studies in Hormone Dependent Cancers, (E. Mazzaferri & N. Samaan, eds.), Blackwell, Boston, 1993, pp. 4973. [16] Martindale, The Extra Pharmacopeia, 31th ed.. Royal Pharmaceutical Society, London, 1996. [ 17] G.L. Plosker &. R.N. Brogden, Drugs, 48 (1994) 930. [18] M.J. Karten & J.E. Rivier, Endocrine Reviews, 7 (1986) 44. [19] P. Cordopatis, E. Manessi-Zoupa, D. Theodoropoulos, R. Bosse, R. Bouley, S. Gagnon & E. Escher, Int. J. Pept. Prot. Res., 44 (1994) 320. [20] R.E. London, P.G. Schmidt, R.J. Vavrek & J.M. Stewart, Int. J. Pept. Protein. Res.. 4 (1982) 334. [21] T.S. Sudha & P. Balaram, Int. J. Pept. Protein. Res.. 21 (1983) 381. [22] F.A. Momany, J. Am. Chem. Soc.. 98 (1976), 2990. [23] (a) D. Benaki, E. Paxinou, V. Magafa, G.. Pairas, E. Manessi-Zoupa, P. Cordopatis & E. Mikros, 2nd Hellenic Forum on Bioactive Peptides, (P. Cordopatis, ed.), Typorama, 2001, p.p. 439–446, (b) D. Benaki, E. Paxinou, V. Magafa, G. Pairas, E. Manessi-Zoupa, P. Cordopatis & E. Mikros, Letters in Peptide Science, submitted. [24] K.G. Estep, C.E.E. Neipp, L.M. Stephens-Stramiello, M.D. Adam, M.P. Allen, S. Robinson & E.J. Roskamp, J. Org. Chem.. 63 (1998) 5300. [25] B.G. Fields & L.R. Noble, Int. J. Peptide Res., 35 (1990) 161. [26] L.A. Caprino, J. Am. Chem. Soc., 115 (1993) 4397. [27] E. Kaiser, R.L. Colescott, C.D. Bossinger & P.I. Cook, Anal. Biochem., 34 (1970) 595. [28] U. Piantini, O.W. Sorensen &. R.R Ernst, J. Am. Chem. Soc., 104 (1982) 6800. [29] A. Bax & D.G. Davis, J. Magn. Reson., 65 (1985) 355. [30] (a) D. Marion & K. Wuthrich, Biochem. Biophys. Res. Commun., 113 (1983) 967; (b) J. Jeener, B.H. Meier, P. Badimann & R.R. Ernst J. Chem. Phys., 71 (1979) 4546. [31] S. Macura, K. Wuthrich &. R.R. Ernst, J Magn. Reson., 47 (1982) 351. [32] C. Eccles, P. Guntert, M. Billeter & K. Wuthrich, J. Biomol. NMR, 1 (1991) 111. [33] K. Wuthrich, In: NMR of Proteins and Nucleic Acids, Wiley, New York, 1986. [34] P. Guntert, W. Braun & K. Wuthrich, J. Moi Biol., 217 (1991) 517. [35] P. Guntert, C. Mumenthaler & K. Wuthrich, J. Mol Biol., 273 (1997) 283. [36] D.A. Pearlman, D.A. Case, J.W. Caldwell, W.S. Ross, T.E. Cheatham, D.M. Ferguson, G.L. Seibel, U.C. Singh, P.K. Weiner & P.A. Kollman, AMBER 5.0. U of California, San Francisco, USA, 1997. [37] R. Koradi, M. Billeter & K. Wuthrich, J Mol. Graphics, 14 (1996) 51.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
217
Synthesis, Biological Evaluation & NMR Solution Structural Models of New Oxytocin Analogues M. Fragiadaki1, S. Koumentakos1, D. Raptis2, G. A. Spyroulias1, V. Magafa1, J. Slaninova3 and P. Cordopatis 'Department of Pharmacy, University of Patras, GR-265 04 Patras, Greece 2 Technological Education Institute, Egaleo, GR-12210 Athens, Greece Department of Peptide Biochemistry, Institute of Organic Chemistry and Biochemistry, Academy of Sciences of Czech Republic, Flemingovo square 2, Prague 6, CZ-166-10, Czech Republic Abstract. We report the solid-phase synthesis of four oxytocin analogues in which the residues in sequence position 8 or 9 were replaced by L-a-t-butylglycine [Gly(Bu')]: [Gly(Bu')8]OT (re-synthesized), [Gly(But)9]OT, [Mpa1, DTyr(Et) 2 , Gly(But)8]OT, [Mpa1, D-Tyr(Et)2, Gly(But)9]OT. The analogues were tested in the in vitro oxytocic assay. In addition, the rat pressor activity was also examined. The analogues [Gly(But)8]OT and [Gly(But)9]OT were found to be weak agonists, while the peptides [Mpa1, D-Tyr(Et)2, Gly(But)8]OT and [Mpa1, D-Tyr(Et)2, Gly(But)9]OT act as very potent antagonists of oxytocin in the rat uterus assay with in vitro pA2 value of 8.2. The solution conformations of the analogues were also studied by a combined use of ID NMR and 2D NMR spectroscopy in DMSO-d6 through restrained molecular dynamics (MD).
218
M. Fragiadaki et al./ New Oxytocin Analogues
Introduction Oxytocin, [cyclo (Cys1-Tyr2-Ile3-Gln4-Asn5-Cys6)-Pro7-Leu8-Gly9-NH2] (OT), is a neurohypophyseal hormone which has been mainly associated with milk ejection and uterine contractions in mammals. Oxytocin was the first neuropeptide whose primary structure was determined and proved by total synthesis [1]. Ever since in several laboratories all over the word, hundreds of oxytocin analogues have been synthesized and studied in an important effort to obtain selective and active analogues, and to elucidate the requirements for peptide-receptor recognition and signal transduction. The first model of solution conformation of OT was proposed by Urry and Walter (cooperative model) [2] and consequently, a broad variety of models have been suggested. Most of them are in agreement that a conformation with a ß-turn at positions 3 and 4, and with a reverse turn at positions 7 and 8, might be a pre-requisite for the uterotonic activity of OT and its analogues [3], whereas the Tyr2 and Asn5 positions are thought to be important for initiation of the biological response [4]. Furthermore, the importance of conformational flexibility for agonist activity and relative conformational rigidity and steric constraints for antagonism has been emphasized (dynamic model) [5]. The oxytocin receptor (OTR) is a classical membrane receptor with seven transmembrane domains linked through a G protein complex to a phospholipase C/protein kinase C signal transduction system [6]. Using a series of OT antagonists, Chan [7] suggested the presence of two distinct subtypes of oxytocin receptors but, to date, there has been no evidence supporting the existence of more than one OTR gene or transcript [8]. The design of OT antagonists has been mainly based on data from structureactivity studies. Structural modifications of OT were studied for the changes in activity as determined by several in vitro and in vivo tests. These studies of oxytocin analogues have revealed that OT agonists and antagonists have different receptor-binding conformations [9]. With the cloning of the OT receptor the design of new analogues is based on structural modeling of the OT receptor complex [10]. Furthermore, recent studies establish that a small 12-residue segment in the distal portion of the N-terminus of the human OTR is required for agonist binding but it does not contribute to the binding site for antagonists [11]. Oxytocin antagonists, in contrast with currently used tocolytics, afford greater specificity and can be expected to exhibit improved efficacy and risk profiles. Such compounds will allow a more effective treatment of preterm labor, with a lower risk of side-effects. To date, although the structures of many oxytocin analogues have been reported, very few have been tested in animal models of parturition, probably because of their low receptor specificity or low receptor affinity and only one, atosiban [Mpa1, D-Tyr(Et)2, Thr4, Om8] has been administered to pregnant women [12]. Generally, inhibitors of the uterotonic activity of OT have been traditionally produced by introduction of bulky ß-carbon substituents into position 1 and/or by substitution of L-tyrosine in position 2 of OT with an aromatic D-amino acid. On the other hand previous structure-activity studies in our laboratory revealed that minimal structural changes of the oxytocin molecule provide weak antagonists [13]. Moreover, synthesis of analogues of biologically active peptides in which structural modifications should enhance resistance toward enzymatic cleavage of peptide bonds resulting in prolonged time course of action is of great importance. It has
M. Fragiadaki et al. /New Oxytocin Analogues
219
been reported that modification of the C-terminal tripeptide side chain influences the chymotryptic cleavage of Tyr-Ile peptide bond in the oxytocin molecule [14]. On the basis of these findings, we set out the synthesis of three new oxytocin analogues and one re-synthesized [15] which contain the conformation constraining amino acid residue L-a-t-butylglycine [Gly (But)] in positions 8 or 9. For these analogues were performed in vitro pharmacological tests and conformational analysis.
Experimental Section Materials: 9-Fluorenylmethoxycarbonyl-protected amino acids and peptide reagents were obtained from Bachem AG and Nova Biochern. The derivative Mpa(Trt) was prepared according to literature [16]. All solvents and reagents used for solid-phase synthesis were of analytical quality. Dimethylsulfoxide DMSO-d6 (MERCK) was used as deuterated solvent in NMR experiments and the peptides were dissolved to a final concentration of 2-2.5 mM in order to record ID and 2D NMR spectra. Peptide Synthesis and Purification: The analogues were synthesized by Fmoc solid phase methodology [17] utilizing a 2-chlorotrityl chloride resin [18] as solid support bearing a Rink-Bernatowitz linker to provide the peptidic amide [19]. Stepwise synthesis of the peptide analogue was achieved with diisopropylcarbodiimide/1hydroxy-benzotriazol (DIC/HOBt) in N,N-dimethylformamide (DMF) as coupling agent [20]. Couplings were performed with 3 molar excess of Fmoc-amino acid, 3.3 molar excess DIG and 4.5 molar excess HOBt as coupling agents in DMF for 2 h at room temperature. Completeness of the reaction was monitored by the Kaiser test [21]. The Fmoc groups were removed by treatment with 20% piperidine in DMF for 30 minutes. Cleavage of peptide-linker bond and removal of the protecting Trt-group from Cys, Asn, Gln and Mpa was accomplished in 4 h at room temperature using a solution of trifluoroacetic acid/ methylene chloride/1,2-ethanedithiol/anisole/water (80:10:5:3:2, v/v) (15 ml/g peptide resin). The obtained peptide was precipitated upon concentration of solvent and addition of ether. The formation of the disulfide bridge was performed in 25% DMSO/H2O for 48 h at room temperature [22]. The solution was frozen and lyophilized to give the final crude oxidized product. All the analogues were purified by gel filtration chromatography on Sephadex G-15 using 25% acetic acid as the eluent. Final purification was achieved by preparative HPLC (Pharmacia LKB-2250) on reversed-phase support C-18 with a linear gradient from 10 to 60% acetonitrile (0.1% TFA) for 30 min and 60 to 100% acetonitrile (0.1% TFA) for 10 min at a flow rate 1.5 ml/min and UV detection at 220 and 254 nm. The appropriate fractions were pooled and lyophilized. All products gave single spots on TLC (Merck precoated silica gel plates, type G60-F254) in the solvent systems: (A) 1butanol-acetic acid-water (4:1:5 upper phase) and (B) 1-butanol-acetic acid-waterpyridine (15:3:10:6). Analytical HPLC (Pharmacia LKB-2210) equipped with a C-18 Phase Sep column S3 ODS2 produced single peaks with at least 98% of the total peptide peak integrals. Electro-sprays MS were in agreement with the expected results.
220
M. Fragiadaki et al. /New Oxytocin Analogues
Biological Assay Methods: The uterotonic activity was determined in vitro on an isolated strip of rat uterus in the absence of magnesium [23]. In principle, cumulative dosing was applied in most experiments, i.e. doses of standard (in the presence or absence of analogues) or of the analogue were added successively to the uterus in the organ bath in doubling concentrations and in 1 min intervals without the fluid being changed until the maximal response was obtained. The agonistic activity was determined by comparing the threshold doses of oxytocin and the analogue. In the case that the analogues were not able to reach the same maximal response as oxytocin, the single dosing procedure was employed. The inhibitory potencies are expressed as their pA2 values. The pA2 values represent the negative logarithm to the base 10 of the average molar concentration [M] of an antagonist which will reduce the response of 2 x units of the agonist to the response with x units of the agonist [24]. Each analogue was tested on uteri from 4-8 different rats. Pressor activity was determined on phenoxybenzamine treated male rats [25]. NMR Spectroscopy: ID NMR and 2D NMR spectra were recorded at the temperature of 298K on a BRUKER A VANCE 400 spectrometer operating at 400.13 MHz. ID spectra over the full spectral width (12 ppm) were acquired with and without presaturation of the H2O signal and a recycle delay of 0.8 s or 1.0 s. DQF-COSY [26] and TOCSY [27] experiments were performed in order to facilitate the identification of the spin-systems of each individual amino acid. TOCSY experiments were carried out by using the MLEV-17 spin-lock sequence and a mixing time of 80-100 ms in both temperatures, consisted by 2K data points in F2 dimension, 16-32 transients and 1024 complex increments in the Fl dimension. TPPI [28] NOESY spectra were recorded using the same as for ID spectra, spectral width. The mixing times varied from 200 ms to 800 ms. Spectra recorded with mixing times 400 ms or longer, were used in order to facilitate the resonance assignment. The NOE intensity volume-to-distance conversion was performed for the NMR data derived by NOE maps acquired with mixing times of 200 or 400 ms. The suppression of the water signal was carried out by its presaturation during the relaxation delay and the mixing time using the "PRESATURATION" [29] pulse sequence of the standard BRUKER library. The other acquisition parameters were the same as for the ID spectra. The spectra consisted of 2K data points in the F2 dimension and IK experiments in the Fl dimension. Raw data were multiplied in both dimensions by a pure cosine-squared bell window function and Fourier-transformed to obtain 2024x1024 real data points. A polynomial base-line correction was applied in both directions. The assignment of TOCSY and NOESY spectra and the identification of dipolar intraproton connectivity for the analogues were performed with the use of sequence specific resonance assignment methodology [30]. NMR data processing was performed using the standard BRUKER software package on a Silicon Graphics O2 workstation. The 2D maps were analyzed on a Silicon Graphics O2 workstation or on Pentium III PC-Linux computers with the aid of the program XEASY (ETH, Zurich) [31]. Results and Discussion The new analogues [Gly(But)9]OT, [Mpa1, D-Tyr(Et)2, Gly(But)8]OT, [Mpa1, DTyr(Et)2, Gly(But)9]OT and the re-synthesized analogue [Gly(But)8]OT were prepared
M. Fragiadaki et al. /New Oxytocin Analogues
221
by Fmoc solid-phase procedure and tested for their biological potency in two pharmacological tests. The results are shown in Table 1 along with the activities of the parent hormone oxytocin, the deaminooxytocin and the oxytocin antagonist analogue Atosiban. As can be seen, replacement of Leu8 (analogue III) or Gly9 (analogue IV) by t Gly(Bu ) decreased biological activities significantly. The analogue [Gly(But)8]OT (III), with a shortened lipophilic bulky side-chain in position 8, was found to be a weak agonist in the uterotonic assay with a potency approximately 8% that of the native hormone. In addition, low potency was observed in the rat pressor assay. For comparison, previous reports concerning the biological activity of this analogue are in very good agreement with our results [15]. The analogue modified in position 9 (IV) with L-a-t-butylglycine (conformationally constrained and lipophilic residue) was found to be a partial agonist, weaker than the analogue III in the uterotonic assay (0.7% that of the oxytocin) and more selective due to the almost complete elimination of pressor activity. These findings are consistent with earlier observations in bibliography, which suggested that the C-terminal tripeptide sequence and especially the proper orientation of the C-terminal glycine carboxamide appear to be critical for obtaining high potency in oxytocin analogues [32, 33]. Therefore, structural modifications of the side-chain moieties in the C-terminal tripeptide oxytocin might lead to analogues with variable biological properties at different oxytocin target tissues. Such variations in biological activity could result from conformational changes influencing interactions of the analogues with the receptor, from steric demands, or from changed susceptibility of the molecules to enzymic cleavage [34]. Furthermore, the greater decrease in the agonistic potency of the analogue [Gly(But)9]OT comparing with the analogue [Gly(But)8]OT indicates that a lipophilic and bulky residue in position 9 induces major restrictions at the conformational freedom of the C-terminal part of the molecule than an identical residue in position 8. Most of the oxytocin antagonists described to date contain a residue without Nterminal amino group, i.e. ß-mercaptopropionic acid (Mpa) or ß-mercapto-ß, ßcyclopenta-methylenepropionic acid (Mca). This approach is the result of thorough study of data which indicate that vasopressin analogues lacking a terminal amino group are inactivated quite slowly [35] and also that the first amino acid has a decisive role in the receptor binding [36]. Furthermore, it has been reported that the configuration and the hydrophobicity of the aromatic amino acid in position 2 of oxytocin are important for the antagonistic activity [37]. In accordance with these observations we synthesized the analogues V and VI. Upon additional substitution of the residue in position 1 by ß-mercaptopropionic acid and the residue in position 2 by 0-ethyl-D-tyrosine, however, the weak agonist analogues (HI and IV) were converted to highly potent antagonist analogues in the in vitro uterine assay, having a pA2 value of 8.1. In comparison to [D-Tyr(OEt)2] OT it is almost 5 times higher inhibitory potency (pA2 = 7.36) [38]. Both analogues were completely inactive in the pressure test as was also the [D-Tyr(OEt)2] OT. The identical high antagonistic activity confirms the hypothesis that the basic character of the amino acid in position 8 is not important for the antagonism [12] of the uterotonic activity.
222
M. Fragiadaki et al. / New Oxytocin Analogues
Table 1. Biological Activities of Oxytocin and its Analogues Uterotonic activity in vitro (lU/mg)
Compound
Uterotonic activity in vitro pA2
Pressor activity lU/mg
I
Oxytocin
546
3.1
11
[Mpa1] oxytocin (deaminooxytocin) "
803
1.44
III
[Gly(Bul)8]OT
46.8 ± 7.2
1.1
IV
[Gly(But)9]OT
3.7+0.9
<0.04
V
[Mpa1, D-Tyr(Et)2, Gly(But)8]OT
8.1+0.2
0
VI
[Mpa1, D-Tyr(Et)2,Gly(But)9]OT
8.110.2
0
8.29
0.02+ 0.02
1
VII
2
4
8
[Mpa , D-Tyr(Et) , Thr , Arg ]OT (Atosiban)
The biological assays for the oxytocin analogues III, IV, V & VI were performed as outlined in the text. The biological activities of the other analogues reported here are taken from the literature: for oxytocin and deaminooxytocin see ref. [5] and for atosiban see ref. [12]. *If tested using cumulative dose-response procedure, the analogue is not able to reach the same maximal response as oxytocin; if tested using single dose procedure the response is reaching the same maximal contraction as oxytocin. Inactive up to the dose of 0.4 mg/kg of exp. animal.
The solution conformations of the analogues IV, V and VI were also studied by a combined use of ID NMR and 2D NMR spectroscopy in DMSO-d6. Upon completion of the identification and assignment of the NOE connectivities, the elements of secondary as well as the tertiary structure of the peptide have been identified. Structural information was extracted from signal intensities, (NOE[Hi-Hj]-l/r [Hi-Hj]6, where Hi, Hj are two protons, and r[Hi-Hj] is the distance between the two protons) and implemented to the structural calculations in order to determine the folding and the tertiary structure of each one of the analogues, in solution (Figure 1 shows representative spectra for analogue IV). Finally, it should be reported that 332, 210 and 243 unique NOE signals were detected for the analogues IV, V and VI respectively. The signal intensity was transformed into upper distance limits and used as structural constraints for the calculation of the 3D structure. From the NOEs observed, and reported above, 276, 154 and 187, respectively, are found meaningful and used in the calculations. After the end of the structural calculations through DYANA (Dynamics Algorithm for NMR Applications) program [39], the 20 best structures (in terms of minimum dynamic energy and smallest deviation of the experimental NOE distances with the same distances as found in the final set of calculated structures) are collected. The family of the 20 models is characterized by high resolution and accuracy as indicated by the low RMSD values for all analogues.
223
M. Fragiadaki et al. / New Oxytocin Analogues
8.50
8.00
3.80
3.80
4.00
4.00
4.20
4.20
4.40
4.40
4.60;
4.60
4.80
4.80
5.00
540
3.80
3.80
4.00
440
4.20
4.20
4.40
4.40
4.60
4.60
4.80
4.80
5.00
540
7.20 ',
7.20
7.40
c
4T
7.40
7.60
7.60
7.80
7.80
8.00
840
8.20
8,20
8.40
8.40
8.60
8.60 8.50
8.00
7.50
Figure 1: 2D !H NMR spectra of the analogue IV. At the A and B it is presented the characteristic area of Ha-NH conjugations as it is concerned the TOCSY and NOESY spectra respectively, while at the C it is presented the characteristic area of NH conjugations, as long as NH-NH NOE signals between the Cys1 and Asn5.
As can be seen, differences in the conformation of the final tripeptide are observed among the three analogues. This fact can be due to the expected high flexibility and mobility of this part of the molecule. High mobility of protein and peptide fragments results in a loss of an important number of NOE signals (constraints), and low precision of the calculated 3D structures. As far as the analogue IV is concerned the cyclic part of the molecule comprised by the amino acids 1-6 (which results by the disulfide bridge between the sulfur atoms of the residues at the positions 1 and 6) is well defined, as well as the aromatic ring of the Tyr at the position 2. In analogue VI the conformation of the C-terminal tripeptide is also well determined and it seems that this fragment folds in such a way that it adopts a ß-turn structure. This is manifested by the numerous NOE signals between the backbone and/or side chain protons of amino acid residues from which terminal of the molecule is consisted. In contrast, for analogue V with L-a-tbutylglycine in position 8, the final tripeptide fragment seems to adopt a different orientation in space but without loosing the characteristics of ß-turn structure observed in the case of analogue VI (L-a-r-butylglycine in position 9) [Figure 2A]. In addition, the tyrosine rings of the analogues IV and VI [Figure 2B] seem to have different orientation (the latter has Oethyl-D-tyrosine instead of the native tyrosine residue). The different conformation of the aromatic ring of the D-amino acid in position 2 could be discussed in conjunction with the antagonistic activity of the analogue VI, compared with the analogue bearing the natural L-amino acid at position 2 (IV).
224
M. Fragiadaki et al. / New Oxytocin Analogues
Figure 2: (A) Superposition of the mean structures of the analogues V and VI and (B) Superposition of the mean structures (as it concerns the part 1-6) of the analogues IV and VI
The combined approach including pharmacological tests, ID NMR, 2D NMR spectroscopy and 3D structures determined through molecular dynamics (MD) protocols allow us to conclude, that the C-terminal tripeptide has very different conformations and thus its conformation is relatively unimportant for the antagonistic character of the analogue in the oxytocin series and that the basic amino group in position 8 is also not a pre-requisite for the potency. On the other hand the position 2 plays a decisive role. References [ 1 ] V. du Vigneaud, C. Ressler, J. M. Swan, P. G. Katsoyannis and S. Gordon J. Am. Chem. Soc., 75 (1953) 4879; V. du Vigneaud, C. Ressler, J. M. Swan, C. W. Roberts and P. G. Katsoyannis, ibid., 76 (1954) 3115. [2] D. W. Urry and R. Walter, Proc. Natl. Acad. Sci. USA, 68 (1971) 956. [3] V. J. Hruby, M. Lebl, In CRC Handbook of Neurohypophyseal Hormone Analogs, K. Jost, M. Lebl, F. Brtnik (Eds), CRC Press, Boca Ralton, FL, Vol. 1 (1987) 105–155; P. Malon, ibid. Vol. 1(1987)211. [4] R. Walter, G. Stahl, Th. Caplaneris, P. Cordopatis and D. Theodoropoulos, J. Med. Chem., 22 (1979) 890; P. S. Hill. W.Y. Chan and V. J. Hruby, Int. J. Pept. Prot. Res., 38 (1991) 32. [5] P. S. Hill, D. D. Smith, J. Slaninova and V. J. Hruby, J Am. Chem. Soc., 112 (1990) 3110. [6] S. Phaneuf, N. Europe-Firmer, M. Varney, I. Z. Mackenzie, F. I. Watson and A. Lopez-Bernal, 7. Endocrinol, 136 (1993) 497. [7] W.Y. Chan, J. Pharmacol. Exp. Ther., 213 (1980) 575. [8] B. F. Mitchell, X. Fang and S. Wong, Reviews of Reproduction, 3 (1998) 113. [9] M. D. Shenderovich, K. E. Kover, S. Wilke, N. Collins and V. J. Hruby, J. Am. Chem. Soc.. 119 (1997)5833. [10]T. Kimura, O. Tanizawa, K. Mori, M. J. Brownstein and H. Okayama, Nature, 356 (1992) 526. [ 11]S. R. Hawtin, H. C. Howard and M. Wheatley, Biochem. J., 354 (2001) 465. [12]P. Melin, J. Trojnar, B. Johansson, H. Vilhardt and M. Akerlund, J. Endocrinol, 111 (1986) 125. [13]M. Lebl, In CRC Handbook of Neurohypophyseal Hormone Analogs, K Jost, M. Lebl, F. Brtnik (Eds), CRC Press, Boca Ralton, FL, Vol 2 (1987) pp. 17-74; N. Assimomytis, E. Manessi-Zoupa and P. Cordopatis, Collect. Czech. Chem. Commun., 59 (1994) 718; N. Assimomytis, V. Magafa. D. Theodoropoulos, P. Cordopatis and J. Slaninova, Lett. Pept. Sci, 3 (1996) 217. [14]J. Hlavacek and 1. Fric. Collect. Czech. Chem. Commun., 54 (1989) 2261. [15]M. Lebl, J. Pospisek. J. Hlavaek, T. Barth, P. Malon, L. Servitova, K. Hauzer and K. Jost. Collect. Czech. Chem. Commun., 47 (1982) 689.
M. Fragiadaki et al. /New Oxytocin Analogues
[16] I. Photaki,, C. Tsougraki, and C. Kotsira-Engonopoulos, Int. J. Pept. Prot. Res., 13 (1979) 426. [17]B. G Fields and L. R. Noble, Int. J. Pept. Prot Res., 35 (1990) 161. [18]K. Barlos, O. Chatzi, D. Gatos and G. Stavropoulos, Int. J Pept. Prot. Res., 37 (1991) 513. [19]M. S. Bernatowitz, S. B. Daniels and H. Koster, Tetrahedron Lett., 30 (1989) 4645. [20] D. Sarantakis, J. Teichnan, E. L. Lien and R.L. Fenichel, Biochem. Biophys. Res. Commun., 73 (1976) 336; W. Koning and R. Geiger, Chem. Ber., 103 (1970) 788. [21]E. Kaiser, R.L. Colescott, C.D. Bossinger. and P.I. Cook, Anal. Biochem., 34 (1970) 595. [22] J. P Tarn, C.-R. Wu, W Liu, and J.-W. Zhang, J. Am. Chem. Soc., 113 (1991) 6657. [23] P. Holton, Brit. J. PharmacoL, 3 (1948) 328-334; R. A. Munsick, Endocrinology, 66 (1960) 451. [24] J. Slaninova, In Handbook of Neurohypophysial Hormone Analogs, K. Jost, M. Lebl & F. Brtnik (Eds), CRC Press, Boca Raton, Vol I (1987) 83. [25] J. Dekanski, Br. J. PharmacoL, 1 (1952) 567. [26] U. Piantini, O.W. Sorensen &. R.R Ernst, J. Am. Chem. Soc., 104 (1982) 6800. [27]Bax & D.G. Davis, J. Magn. Reson., 65 (1985) 355. [28]D. Marion & K. Wuthrich, Biochem. Biophys. Res. Commun., 113 (1983) 967; J. Jeener, B.H. Meier, P. Badimann & R.R. Ernst, J. Chem. Phys., 71 (1979) 4546. [29] S. Macura, K. Wuthrich &. R.R. Ernst, J. Magn. Reson., 47 (1982) 351. [30] K. Wuthrich, In NMR of proteins and nucleic acids, Wiley, New York, 1986. [31]C. Eccles, P. Guntert, M. Billeter & K. Wuthrich, J. Biomol. NMR, 1 (1991) 111. [32] V. J Hruby, In the book: Biochemical Actions of Hormones, G. Litwack (Ed.), Academic Press, New York, (1986) 191. [33] Y. F. Ting, C. Smith, G. Stahl, R. Walter, P. Cordopatis and D. Theodoropoulos, J. Med. Chem., 23 (1980) 693; M. Lebl, J. Slaninova and R. L. Johnson, Int. J. Pept. Prot. Res., 33 (1989) 16. [34] J. Hlavacek, J. Pospisek, J. Slaninova, W.Y. Chan and V. J. Hruby, Collect. Czech. Chem. Commun., 52(1987)2317. [35] Z. Grzonka, F. Kasprzykowski, L. Lubkowska, K. Darlak, T. A. Halm and A. F. Spatola, Peptides Res., 4 (1991) 270. [36]G. K. Toth, K. Bakos, B. Penke, I. Pavo, C Varga, G. Torok, A. Peter and F. Fulop, Bioorg. & Med. Chem. Lett., 9 (1999) 667. [37] G. Flouret, W. Brieher, K. Mahan, J. Med. Chem., 34 (1991) 642; M. Lebl, G. Toth, J. Slaninova and V. J. Hruby. Int. J. Pept. Prot. Res., 40 (1992) 148. [38]R. Jezek, M. Zertova, J. Slaninova, P. Majer, Z. Prochazka, Coll. Czech. Chem. Commun., 59 (1994) 1430. [39] P. Giintert, C. Munenthaler and K. Wuthrich, J. Mol. Biol., 273 (1997) 289.
225
This page intentionally left blank
Part IV.
Molecular Targets and Drug Design
This page intentionally left blank
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
229
Molecular Aspects of Human Abdominal Aortic Aneurysm: Differential Expression of Genes Coding for Extracellular Matrix Proteoglycans A. D. Theocharis and N. K. Karamanos Department of Chemistry, Section of Organic Chemistry, Biochemistry and Natural Products, University of Patras, 261 10 Patras, Greece Abstract. Abdominal aortic aneurysm (AAA) is a common disease of human aorta with increased incidence the last decades. It is a complication to atherosclerosis and is closely associated with alterations in matrix macromolecules. Proteoglycans (PGs) are quantitatively minor components of aortic wall but are of importance for matrix assembly and the biomechanical properties of the tissue. The expression of the extracellular matrix arterial PGs (versican, biglycan and decorin) is reported. Analysis was performed by reverse-transcriptase polymerase chain reaction. The mRNAs coding for versican isoforms V0 and V1 were identified in both tissues, whereas V2 was absent. The expression of V0 was decreased 40% in aneurysmal vessel wall, whereas that for V1 remained constant. The level for the mRNA coding for biglycan was found to decreased 60% in AAA, while the biosynthesis of decorin remained constant in both tissues. It is suggested that the modulated biosynthesis of PGs in combination with the failure of elastin fibers are crucial factors contributing to the altered viscoelastic and compressive properties of the tissue and thereby to the deformity and dilatation of aorta.
Atherosclerosis and Abdominal Aortic Aneurysm: Atherosclerosis is the most common disease of blood vessels and the prime disorder leading to death and serious morbidity in the Western world [1]. The disease affects primarily the intima layer of both muscular and elastic arteries. The earliest type of atherosclerotic lesion, called fatty streak, is an inflammatory lesion consisting only of monocyte-derived macrophages and T-lymphocytes. Although different theories have been proposed for the atherogenesis, the hypothesis of the response to injury dominates. This theory proposes that endothelial denudation is the first
230
A.D. Theocharis andN.K. Karamanos / Human Abdominal Aortic Aneurysm
step of the disease, but the most recent version of this hypothesis emphasizes endothelial dysfunction rather than endothelial denudation. Several causes suspected to be responsible for endothelial dysfunction including elevated and modified low density lipoproteins (LDL), free radicals caused by cigarette smoking, elevated plasma homocysteine concentrations, hypertension, infections by microorganisms such as herpes virus or Chlamydia pneumoniae and other factors [2]. The chronicity of the inflammation results in emigration of numerous inflammatory cells from the blood within the lesion. Activation of these cells leads to release of several proteases, cytokines, chemokines and growth factors, which induce further damage and eventually lead to focal necrosis. The repeated cycles of emigration and proliferation of smooth muscle cells (SMCs) and accumulation of fibrous tissue and inflammatory cells lead to further enlargement and formation of advanced complicated lesion [3]. In this case intima layer is markedly thickened and consisted by a raised focal plaque, which has a lipid core covered by a fibrous cap and surrounded by increased number of SMCs, inflammatory cells and depositions of extracellular matrix components. The atherosclerotic lesion occludes the lumen of the vessel leading to various complications of the disease, such as ischemia of the heart, brain, or extremities and further, in infarction [2]. In these areas, where atherosclerotic occlusive disease (AOD) developed, aneurysmal dilatation often occurs with increasing age [4]. The most common type of aneurysm in human is the abdominal aortic aneurysm (AAA), which is constantly accompanied by severe atherosclerosis and often is referred as atherosclerotic aneurysm [5]. Zarins et al. [4] found that primates fed an atherogenic diet developed only occlusive atherosclerotic plaque during the first year, but aneurysmal dilatation occurred thereafter at sites of previous occlusive lesions. It is suggested that the chronicity of the disease is important for the development of the atherosclerotic aneurysm. This suggestion is consistent with the clinical observation that patients with AAA are approximately a decade older than those with AOD. Aneurysmal aorta is characterized by alterations of aortic wall architecture, which is mainly due to the destruction of media layer and the failure of elastic fibers, whereas the collagen fibers seems to affected only in ruptured aneurysms [6–8]. The decrease of the number of smooth muscle cells (SMCs) in aneurysmal aortic wall due to the induction of their apoptosis suggested that significantly contribute to the development of the disease [9]. Recent studies have also demonstrated that PGs and glycosaminoglycans (GAGs) present in aneurysmal aortas undergo marked and specific alteration in their contents and structures [10–12]. The overall GAG content significantly decreased (60%) in AAA as compared to normal aortas. A 90% decrease in heparan sulfate (HS) content and 65% and 73% in chondroitin sulfate (CS) and hyaluronan (HA), respectively was recorded. Dermatan sulfate (DS) was slightly decreased (8%) in AAA. Fine structural alterations in respect to disaccharide composition of DS and HS were also detected in AAA [10]. The differences in the composition of matrix molecules occurred in the diseased tissue may in part be attributed to the increased degradation of these molecules by several matrix metalloproteases released by SMCs and inflammatory cells [13–15]. Functional Roles of Proteoglycans in Aortic Wall:Although PGs / GAGs are quantitatively minor structural components of the aortic wall, they influence its physicochemical properties and participate in several physiological events. Versican, the large molecular size chondroitin sulfate proteoglycan (CSPG), is the most abundant PG of the arterial wall and is mainly localized in the arterial intima. This PG is synthesized predominantly by vascular SMCs and it is composed of a core protein, carrying several covalently bound CS chains. Versican, through its CS chains, is implicated in the entrapment of LDL in the arterial wall during the atherogenesis [16]. There are at least four possible splice variants of mammalian
A.D. Theocharis and N.K. Karamanos / Human Abdominal Aortic Aneurysm
231
versican - V0, V1, V2 and V3 - varying in length of their glycosaminoglycan attachment domain. Only the largest isoforms - V0 and V1 - have been identified in the arterial wall. Vo includes two such domains (GAG-a and GAG-J3), which provide attachment sites for GAG chains, while V1 include only the GAG-a domain [17]. Interactions of versican with hyaluronan and link proteins lead to formation of large aggregates that trap water creating viscoelastic and turgor pressures within the arterial wall. These aggregates are considered to create reversibly compressive structures necessary to avoid the deformity of blood vessels by the pulsatile pressures of the circulatory system [16]. Biglycan and decorin belong to a class of small leucine-rich proteoglycans (SLRPs), bearing two or one DS/CS side chains, respectively. They are usually found in close association with collagen type I and III and are thought to play important roles in stabilization and proper arrangement of collagen fibers [17]. SLRPs can also bind via their core proteins the transforming growth factor beta (TGF-P), regulating its bioavailability in this tissue. The DS chains of biglycan and decorin exhibit antithrombotic properties since can bind heparin cofactor II (HC II) accelerating the inhibition of thrombin by this complex [18]. Furthermore, several reports have demonstrated that chondroitin sulfated at the C-4 of galactosamine (C4S) and DS exhibit antiatherogenic properties, since protect LDL and high density lipoproteins (HDL) against copper induced oxidation [19, 20]. Heparan sulfate proteoglycans (HSPGs) is another class of arterial PGs present in basement membranes of endothelial cells playing important roles as tissue barrier. The HSPGs is present also on cell membrane of arterial cells and can act as low affinity receptors for basic fibroblast growth factor (bFGF) facilitating the internalisation of this growth factor. HSPGs are also powerful anticoagulant molecules and the basis for this activity lies in the ability of their GAG chains to bind serine protease inhibitors such as antithrombin III (AT III) and HC II [21]. Expression of Extracellular Matrix Proteoglycans: Aneurysmal abdominal aortic tissues (n = 12, males aged 55-80 years) were derived at aortic reconstruction surgery, performed in the University Hospital of Patras, Patras, Greece. Macroscopically normal abdominal aortas (n - 4 males aged 30-45) were obtained from normal organ transplant donors and were free of macroscopically observable disease. Outer adventitia and mural thrombi were removed from the remaining portions of aneurysmal aortas. Outer adventitia was also removed from normal aortas. Total RNA was isolated from aortic tissue samples by the quanidine thiocyanate / phenol / chloroform method [22] and the DNA impurities were removed following digestion with DNase I [12]. The RNA was isolated following addition of phenol / chloroform and consequently centrifugation at 10 000 x g for 20 min at 4 °C. The aqueous phase, containing the RNA, was precipitated by the addition of an equal volume of isopropanol. The yield and the purity of the RNA preparations were determined by measuring the absorbance at 260 nm and estimating the ratio A 260 nm/ A 280 nm , respectively. The expression of extracellular matrix proteoglycans was analysed by RT-PCR of the total RNA preparations. Equal amounts (2 u,g) of total RNA from each tissue samples were used for first strand cDNA. The entire reverse transcription products served as the template to PCR amplification of PGs transcripts using specific primers for human extracellular matrix PGs (versican isoforms, biglycan and decorin) and the stable expressed gene of GAPDH. PCR amplifications were performed for 40 cycles (Table 1), except of those of GAPDH, which performed for 28 cycles. PCR reaction includes 1 min denaturation at 94°C, 1 min in annealing temperature and 2 min at 72°C for extension. Aliquots (16 u.L) of the amplification products were electrophoresed in 2 % (w/v) agarose gels containing 1 |4,g/mL ethidium bromide, using low molecular weight DNA markers as standards. Bands were visualized on a UV lamp and gels were photographed with a Kodak
232
A.D. Theocharis and N.K. Karamanos /Human Abdominal Aortic Aneurysm
camera. The amounts of PCR products obtained were determined by measuring the fluorescence of bands, using the digital image analysis program [12]. Table 1: Sequence of oligonucleotide primer and PCR conditions.
PGs Versican V0 Versican
v,
Versican V2 Decorin Biglycan GAPDH
Upstream primer GACCTCAGGCG CTTTC GCGCCACCCTG TGAC GACCTCAGGCG CTTTC GGGGCAAAAC ACCAGTCC TGCAGAACAAC GACATCTCCG ACATCATCCCT GCCTCTACTGG
Downstream primer CAGTGGTAACG AGATGCTTC CAGTGGTAACG AGATGCTTC TAGCACTGCCC TTGGA TATTTCCTAAG CCCAGCCTCTC GGAGGAGCTTG AGGTCTGGGA AGTGGGTGTCG CTGTTGAAGTC
4174–4524
Anneal. Temp. (°C) 50
Length (bp) 351
1178–1563
50
386
4174–4547
52
373
417-834
60
413
409-1027
60
618
608-868
-
261
Region
In both normal and aneurysmal aortas mRNA encoding for Vo and V1 versican isoforms was detected, whereas V2 isoform was not expressed in any of the preparations. As estimated by the semi-quantitative RT-PCR the cells from AAA expressed much less Vo isoform (40 %), as compared with normal aortas (fluorescence 0.58 ± 0.06 and 0.35 ± 0.04, respectively) (Fig 1). On contrary, V1 isoform was similarly expressed in both normal (0.24 ± 0.03) and aneurysmal aortas (0.25 ± 0.03) (Figs 1 and 2). The lower amounts of versican found in aneurysmal aortas thus correlate with a specifically lower expression of the V0 isoform.
Figure 1. Screening of gene expression coding for the extracellular matrix PGs at the mRNA level studied by RT-PCR. Lanes 1, 2 indicate two out of four representative cases of normal aortas, and lanes 3-7 five out of twelve cases of aneurismal aortas. The amplification of GAPDH reference gene was the internal standard for the method.
The analysis of both SLRPs (biglycan and decorin) in both tissues revealed that these two molecules were differentially regulated in AAA. Analysis showed that arterial cells in AAA
A.D. Theocharis and N.K. Karamanos /Human Abdominal Aortic Aneurysm
233
transcribed substantially less biglycan (fluorescence 0.71 ± 0.075) than those in normal aortas (fluorescence 1.78 ± 0.15) (Fig 2). In contrast, the biosynthesis of decorin was not affected in AAA, since decorin was found to express constantly in both tissues (fluorescence 0.31 ± 0.04 in normal aortas and 0.30 ± 0.04 in aneurysmal aortas) (Fig 2).
Figure 2. Variation of mRNA coding for matrix PGs estimated by RT-PCR analysis and determined by measuring the fluorescence in normal (n = 4) and aneurysmal aortas (n = 12). Asterisks indicate significantly different values (P < 0.001) as compared to normal aortic tissue.
Discussion - Concluding Remarks The architecture of the aortic wall is responsible for the biomechanical properties of the tissue. The extracellular matrix components elastin collagen and PGs provide the aortic wall with mechanical strength and elasticity, properties which needed to accommodate the pulsatile pressures of the circulatory system. Analysis for the mRNA encoding for versican by RT-PCR revealed that normal and aneurysmal aortas express only V0 and V1 versican isoforms, whereas V2 isoform was not detected in any case. A specific down regulation for the largest isoform V0 (40%) demonstrated in AAA in comparison to normal tissue. In contrast, the V1 isoform was constantly expressed in both tissues. The V0 includes both GAG-a and GAG-P domains providing attachment sites for up to 30 CS/DS chains, whereas the V1 isoform includes only the GAG-P domain with the fully glycanated form to contain up to 15 CS/DS chains [17]. Decreased concentration of versican with increased proportion of the V1 isoform, as seen in AAA would create an extracellular matrix with lower swelling pressure and decreased viscoelastic properties. It has been also proposed that the different isoforms of versican expressed in distinct and temporal patterns are related to specific properties of the tissues. The decreased expression of versican in AAA may be a response to various cytokines and growth factors present in diseased tissue. It is well known that several growth factors and cytokines influence the expression of versican and may also regulate the transcription of versican isoforms. For example, TGF-P and PDGF stimulate the expression of versican mRNA by SMCs in vitro [23], whereas interleukine-lbeta (IL-1P) and insulin growth factor (IGF-1) decrease the synthesis of this molecule [24, 25].
234
A. D. Theocharis and N. K. Karamanos / Human Abdominal Aortic Aneurysm
Significant decrease (60%) in the expression of biglycan mRNA was also observed in AAA. Our results are in agreement with a previous study [26], which demonstrated a more marked decrease in the expression of biglycan in AAA, whereas the expression of decorin remained unchanged. Biglycan binds a variety of matrix molecules, such as collagen types I, III and V, participating in the matrix assembly [27]. The decreased expression of biglycan in AAA may result in anomalous arrangement of collagen fibrils and in alterations in the mechanical properties of the fibrils. Furthermore, significant decrease in the antithrombotic properties in AAA is strongly suggested due to the limited expression of biglycan, since it is known that biglycan and deorin either bound to collagen type V or free in solution accelerate the HC II - thrombin inhibition reaction [18]. The elimination of the expression of biglycan in AAA may be regulated by the cytokines and growth factors found in the tissue. The maintenance of the expression of mRNA encoding for decorin in AAA in similar levels to that of normal aortas is in agreement with other investigators [26]. Decorin is directly implicated in collagen type I fibriilogenesis, assisting in the correct positioning of the collagen molecule in the staggered conformation of the fibril [17], The molar ratio of decorin to collagen is important for the proper formation of the matrix architecture. The elevated collagen amounts in aneurysmal aortas taking together to the unchanged biosynthesis of decorin suggest imbalance in this ratio, which probably results in disordered collagen deposition. The latter is also suggested by other studies, which demonstrate increased extractability of collagen and decorin in AAA in comparison to normal aortas [28, 29]. The alteration in conformation of collagen fibrils may further influence the mechanical properties of the aneurysmal aortic wall. In conclusion, in this article we demonstrate that the expression of the genes of matrix PGs is significantly affected in AAA. This modulated biosynthesis of PGs in combination with the failure of elastin fibbers, results to functional alterations and reduced mechanical resistance of the aortic wall, contributing in the development of aneurysmal dilatation.
References [1]TN Wight, Arteriosclerosis 9 (1989) 1. [2] R Ross, New Engl J Med 340 (1999) 15. [3] R Ross, Nature 362 (1993) 801. [4] CK Zarins, S Glagov, D Vesselinovitch and RW Wissler, J Vasc Surg 12 (1990) 246. [5] CB Ernst, N Engl J Med 328 (1993) 1167. [6] JM Reilly and MD Tilson, Incidence and etiology of abdominal aortic aneurysms. In GE Pierce (ed.) The surgical clinics of North America: Abdominal aortic aneurysms. W.B. Saunders Company, Vol 69 (4) 1989. [7] JS Campa, RM Greenhalgh and Powell JT, Atherosclerosis 65 (1987) 13. [8] S Menashi, JS Campa, RM Greenhalgh and JT Powell, J Vasc Surg 6(6) (1987) 578. [9] A Lopez Candales, DR Holmes, S Liao, MJ Scott, SA Wickline and RW Thompson, Am J Pathol 150 (1997)993. [ 10] AD Theocharis, I Tsolakis, T Tsegenidis and NK Karamanos, Atherosclerosis 145 (1999) 359. [11] F Lamari, AD Theocharis, A Hjerpe and NK Karamanos, J Chromatogr B 730 (1999) 129. [ 12] AD Theocharis, I Tsolakis, A Hjerpe and Karamanos NK, Atherosclerosis 154 (2001) 367. [ 13] RW Thompson, DR Holmes, RA Mertens, S Liao. MD Botney, RP Mecham, HG Welgus and WC Parks, J Clin Invest % (1995) 318. [14] I Halpert, UI Sires, JD Roby, S Potter-Perigo. TN Wight, SD Shapiro, HG Welgus, SA Wickhne and WC Parks, Proc Natl Acad Sci 93 (1996) 9748. [15] T Freestone, RJ Turner, A Coady. DJ Higman, RM Greenhalgh and JT Powell. Arterioscler Thromb Vasc Biol 15 (1995) 1145. [ 16] TN Wight, Arterial wall. In WD Comper (ed.) Extracellular Matrix. Tissue Function. OPA. Vol. 1. 1996, 175-202.
A.D. Theocharis and N.K. Karamanos /Human Abdominal Aortic Aneurysm
235
[ 17J RV lozzo, Ann Rev Biochem 61 (1998) 609. [ 18) RA Shirk, N Parthasarthy, JD San Antonio, FC Church and WD Wagner, J Biol Chem 275 (2000) 18085. [ 19] R Albertini, P Ramos, A Giessauf, A Passi, G De Luca and H Esterbauer, FEBS Letters 403 (1997) 154. (20] R Albertini, G De Luca, A Passi, M Moratti M and PM Abuja, Arch Biochem Biophys 365 (1999) 143 [21] L Kjellen and U Lindahl, Ann Rev Biochem 60 (1991) 443. [22] P Chomczynski and N Sacchi, Anal Biochem 162 (1987) 156. [23] E Schonherr, HT Jarvelainen, LJ Sandell and TN Wight, J Biol Chem 266 (1991) 17640. [24] EE Owarnstrom, HT Jarvelainen, MG Kinsella, CO Ostberg, LJ Sandell, RC Page and TN Wight, Biochem J 294 (1993) 613. [25] PY Davis, CR Frazier, JR Shapiro and NS Fedarko, Biochem J 324 (1997) 753. [26] NA Tamarina, MA Grassi, DA Johnson and WH Pearce, J Surg Res 74 (1998) 76. [27] E Schonherr, P Witsch-Prehm, B Harrach, H Robenek, J Rauterberg and H Kresse, J Biol Chem 270 (1995)2776. [28] M Spina, AM Degani, A Stella, M Gargiulo, G Guiducci and G Genacchi, Biology and Physiology of the Extracellular Matrix. In C Balduini, P Cherubino, G De Luca (eds.) La Goliardica Pavese, Pavia, 1996, pp. 219-224. [29] AD Theocharis, A Hjerpe, G De Luca and NK Karamanos, Submitted (2001).
236
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Advances in Botulinum Neurotoxin Inhibitors based on the SNARE Motif G. J. Moore, D. Moore, S. Roy, L. J. Hayden and G. Murray Hamilton. Pepmetics Inc., 1762-1st Avenue N.W., Calgary, Alberta, Canada T2NOB1 and Therapy Group, Defence Research Establishment Suffield, Medicine Hat, Alberta, Canada T1A 8K6 Abstract. Botulinus neurotoxins are zinc proteases which cleave the SNARE proteins of synaptic vesicles thereby preventing neurotransmitter release. The SNARE complex is a four helix bundle made up of three proteins which each contain the repeating SNARE motif AAxxA, where A is Asp or Glu and x is any amino acid. In the present report 'hinge' peptide libraries were constructed based on the SNARE motif and shown to inhibit Botulinum neurotoxin.
Introduction Botulinum neurotoxin A (Botox A) is one of several protein toxins from clostridium botulinum which cause paralytic syndromes resulting from blockade of neurotransmitter release. These toxins are all zinc endopeptidases acting in the neuronal cytosol: Botox B, D, F and G as well as tetanus toxin attack specifically VAMP (also called synaptobrevin) - a protein of synaptic vesicles, whereas Botox A and E cleave SNAP-25 and Botox C acts on syntaxin - both proteins of the presynaptic membrane SNAP-25 is one component of the so-called SNARE complex which is responsible for docking synaptic vesicles and fusion to the cell membrane as the immediate precursor event to transmitter release. The SNARE complex is a four-helix bundle made up of two small proteins, namely VAMP and syntaxin, and a larger protein SNAP-25 which provides two of the four threads of the helix bundle. SNAP-25 also contains a lipid anchor region (amino acids 85-92) between the two helical threads, which lines up with transmembrane domains at the C-termini of both VAMP and syntaxin [1]. The botulinus neurotoxins each cleave selectively a different peptide bond within one of the three target proteins which comprise the SNARE complex.. Whereas Botox A selectively cleaves the Glnl87-Arg203 bond near the C-terminus of SNAP-25, Botox B cleaves Q -F77 of VAMP, Botox C cleaves K253-A254 of syntaxin, Botox D cleaves K59-L60 of VAMP. Botox E cleaves R180I181 of SNAP-25, Botox F cleaves Q58-K59 of VAMP, and Botox F cleaves A81-A82 of VAMP.
G.J. Moore et al. /Advances in Botulinum Neurotoxin Inhibitors
237
A repeating motif exists within the sequences of all three of the proteins of the SNARE complex which, when introduced in the form of synthetic peptides of about ten residues in length, inhibits the actions of botulinus toxins on the SNARE complex [2]. For example one version of the repeating motif present in VAMP, designated V2 and having the sequence E62LDDRADALQ71, blocks the neurotoxic actions of Botox A and B when the peptide is injected into cultured Aplysia neurons. The significance of the repeating SNARE motif, which appears twice on each of the four threads of the SNARE helix bundle upstream of the cleavage sites, is not well understood, although it presumably acts as a recognition site for binding of some other biomolecule(s), and may also be used by Botox as a binding-recognition element. In agreement with this, cross recognition of the target proteins by the various toxins occurs: Botox A inhibits VAMP proteolysis by Botox B, and Botox B and tetanus inhibit the cleavage of SNAP-25 by Botox A. Moreover serum albumin, which contains SNARE motifs within helical regions of its secondary structure (D183ELRD187 and D255DRAD259), inhibits the cleavage of synthetic substrate by Botox A (present report).. Thus although the mechanism of action of V2 (and other variants of the SNARE motif) is not proven, it could involve binding to a complementary recognition site on Botox resulting in inhibition of productive binding of Botox to the SNARE complex. Interestingly, possible complementary SNARE motifs (K592KVNK596 and K701RNEK705 in the heavy chain, and K335LKFDK340 and K359VLNRK364 in the light chain) exist in Botox A. It has been suggested that the distance between the SNARE motif and the cleavage site may be a determining factor for recognition/cleavage by the various Botox enzymes [3]. Strategy: Several possible strategies for inhibiting Botox exist: block the transmembrane domain of Botox and prevent cell entry, e.g. complementary peptides, antibodies; block the proteolytic site of Botox, e.g. substrate inhibitors, selective chelation of zinc; block the interaction of Botox with binding-recognition motif, e.g. mimics of SNARE motif which bind to complementary site(s) on Botox. The first strategy has potential application prior to exposure to toxin (e.g. immunization), as well as for the deactivation of toxin in body fluids after exposure. The latter strategies, on the other hand, have the potential for providing total body treatment after toxin exposure - assuming that the therapeutic agent is able to cross cell membranes and inactivate toxin which has already entered the cell. Whereas agents based on active site inhibitors will have to be tailored to individual variants of Botox, therapies based on the SNARE motif have the potential to treat poisoning by all forms of Botox as well as tetanus. Regardless of mechanism, one approach to producing improved inhibitors of Botox is to identify the structural elements of V2 (and other SNARE motifs) which make it an effective inhibitor and then reconstruct these components into a smaller and preferably nonpeptidic molecule which would be able to traverse membanes, thereby providing not only access to the inside of cells but also the potential for oral activity. A streamlined and efficient approach [4,5] to this goal is to create a library of small semimimetic peptides containing the essential elements of the repeating motif in the SNARE complex, identify the most active peptide in the mixture by iterative deconvolution of the library, and then restructure the best semimimetic peptide identified into a fully fledged nonpeptide mimetic using computer molecular modelling techniques based on structural details derived from Xray crystallography studies of Botox bound to SNARE motif. The important structural feature of the SNARE motif is comprised of an amino acid sequence made up of residues: A-A-x-x-A-x-x where A=acidic residue and x=nonpolar or polar residue.
238
G.J. Moore et al /Advances in Botulinum Neurotoxin Inhibitors
In the helical arrangement of these groups within the secondary structure of the SNARE protein complex from which they are derived, the end result is a cluster of three neighbouring negative charges juxtaposed by a nonpolar moiety. In other words, the required motif could be envisaged as three negatively charged groups mounted in close proximity on a hydrophobic template. A number of commercially available compounds which fit this general scheme are being investigated for inhibitory activity in our laboratory, together with several peptide libraries which have been designed and synthesized with these considerations in mind. On the other hand, recent evidence basd on X-ray crystallography studies on Botox B bound to its substrate VAMP/synaptobrevin, suggest that Botox enzymes attack their substrates, not when they are present in the four helix bundle formation of the SNARE complex, but rather when they are in random coil or extended structures [3]. Accordingly the triad of negative charges which comprise the SNARE motif would not necessarily exist in close proximity, and in this regard it is significant that our "hinge" peptide libraries are constructed so that they can assume extended as well as folded structures, capable of mimicking any of the possible relevant conformational scenarios for the SNARE motif. Design and Synthesis of Peptide Libraries: For example, the following library has been synthesized which is designed to mimic the SNARE motif: SN-1 = Ac -X1-X2-LINKER-X3-X4-NH2, where Xi, X2, X3 and X4 are mixtures of Asp, Glu, Gln and Arg, and the LINKER group is 4-aminobutyric acid The rationale for the design of the peptide minilibrary SN-1 is based on the preponderance of acidic residues in SNARE motifs, together with the occurrence of other residues in SNARE motifs which can provide solubility properties to the libraries - hence the inclusion of Arg and Gln in SN-1. The multiple negative charges represented by the Asp and Glu residues, when mounted around the flexible linker group 4-aminobutyric acid (Abu), should engender structural and conformational variations on the SNARE motif which will inhibit Botox Coincidentally the Gln and Arg residues in SN-1 are also representative of the scissile bond (Q-R) in the Botox A substrate, so that the peptide minilibrary SN-1 doubles in a rudimentary way as a substrate mimic. This parallels the classical approach to inhibitor discovery, which is usually done by minor modification of substrate structure leading to compounds which bind to the active site but are not cleaved by the enzyme. Accordingly, incorporating residues of the scissile bond (Gln-Arg) into peptide libraries such as SN-1 should provide structural and conformational characteristics which might result in substrate-based inhibitors. Clearly SN-1 contains the potential for containing inhibitors based not only on the SNARE motif but also on substrate structure. Another consideration for the design of peptide libraries with inhibitory potency is the inclusion of residues which would be expected to coordinate to the active site zinc atom of Botox, namely Cys and His as well as acidic residues. The approach here is to create a peptide library containing an array of zinc binding elements in a variety of different formats, wherein there should be individual peptides which recognise not only the zinc atom but also unique aspects of the active site of Botox A in which the zinc atom resides. The intent is to produce zinc targeted inhibitors which show selectivity for Botox over other zinc metalloenzymes. An example of a library targeted to produce active site zinc inhibitors is:SN-3 : Ac-X 1 -X 2 -Abu-X 3 -X 4 -NH2 where Ac = acetyl, Abu = 4-aminobutyric acid, and X1, X2, X3 and X4 are mixtures of the amino acids Asp, Glu, His and Cys.
G.J. Moore et al. /Advances in Botulinum Neurotoxin Inhibitors
239
This library contains both active site zinc directed probes and variations on the SNARE motif within the same basis set (library) of peptides. Inhibitors of Botox A: In our studies a number of compounds (Table 1) were found to inhibit the Botox A mediated cleavage of the 17-amino acid synthetic peptide substrate Ac-SNKTRIDQANQRATKML-NH2, which derives from the C-terminal part (residues 187-203) of SNAP-25 and contains the scissile Q-R peptide bond targeted by Botox A. Several thiol-containing compounds, namely dithiothreitol (widely used to reduce disulphide bonds in proteins), DMPS (a potent chelator of heavy metals) and Captopril (a clinical inhibitor of the zinc dipeptidase Angiotensin Converting Enzyme) block the cleavage of this substrate. However the non-thiol ACE inhibitor Lisinopril was ineffective, whereas the non-thiol prodrug ACE inhibitor enalapril was an effective inhibitor. The thiol compounds DMPS, captopril and DTT presumably act by a mechanism involving sequestration of the zinc atom at the active site of Botox A. The inhibitory activity of enalapril was unexpected but could suggest an affinity for the active site of Botox A which is not shared by lisinopril. Table 1. Inhibition of the Botox A cleavage of synthetic heptadecapeptide substrate
Inhibitor Dithiothreitol
Concentration (mM) % Inhibition I 26
-SH Concentration (mM)
DMPS*
0.1
72
0.2
Captopril
1
76
1
Lysinopril
5
7
0
Enalapril
5
40
0
V2(DDRADALQ)
5
40
0
Glu-Glu-Glu
5
22
0
Glycyrrizic acid
5
55
0
LibrarySN-1
5 (20uM each)
16
0
Library SN-3
0.5 (2uM each)
51
0.125
2
The synthetic peptide V2 was also able to inhibit Botox A, but at a higher concentration (5mM) than has been observed previously in vivo when SNAP-25 was the substrate (l00uM, see ref [2]). This may reflect the absence of SNARE motif recognition site in the short synthetic substrate used for the present studies. Interestingly serum albumin (l0uM), which contains the SNARE motif in duplicate, inhibited cleavage of SNAP-25(187–203) under our assay conditions which had been optimized for zinc (1 uM) and DTT (0.5 mM) concentrations. In contrast Schmidt & Bostian [7] observed that in the presence of 250uM zinc and 5mM DTT cleavage of this substrate by Botox A only occurred in the presence of BSA; our findings would suggest that this was probably due to sequestration of inhibitory levels of zinc/DTT by BSA. Investigations of SN-1 on the Botox A cleavage of substrate have shown that this peptide minilibrary does inhibit the enzyme activity (Table 1). This suggests that variations on the SNARE motif present in the library mixture may interfere with binding of Botox to the substrate. However a series of peptides which were selected for their potential to represent simple variations on the SNARE motif, i.e. Glu-Glu, Glu-Glu-Leu, Glu-Glu-Glu and GluPro-Glu-Thr, were generally inactive (data not shown), with the notable exception of Glu-
240
G.J. Moore et al. /Advances in Botulinum Neurotoxin Inhibitors
Glu-Glu. A number of peptides with potential complementary sequences of the SNARE motif, i.e. Lys-Arg, Lys-Lys, Orn-Om, Lys-Lys-Lys, Om-Orn-Om and Lys-Phe-Gly-Lys, were inactive as expected because the synthetic 17mer peptide substrate used for the assay lacks the repeating SNARE motifs that are present in the longer SNAP-25 natural substrate. However these basic peptides would be expected to inhibit the cleavage of SNARE proteins by Botox enzymes in general, if they bind well to the SNARE recognition motifs in these proteins. Interestingly glycyrrhizic acid, which is a steroid glycoside containing 3 carboxylate groups, was able to inhibit Botox (Table 1). This finding, as with Glu-Glu-Glu, suggests that certain configurations of negative charges are able to approximate the SNARE motif and inhibit Botox A. However in the final analysis it would appear that the most potent inhibitor of Botox containing a SNARE motif variation is likely to be found in the peptide library SN-1 because each peptide in the mixture was present at 20uM concentration (Table 1). Finally the present experimental findings also indicate that effective inhibitors of Botox A are likely to be derived from thiol containing compounds (the inhibitory activity of DMPS is particularly remarkable). In agreement with this, the peptide minilibrary SN-3 turned out to be an exceptionally potent inhibitor of Botox A (Table 1). When the overall thiol concentrations of the inhibitors are compared (Table 1) it is apparent that the peptide library SN-3 is the most potent thiol inhibitor of Botox. Furthermore it is very likely that SN-3 contains a peptide(s) which is a more potent inhibitor than DMPS. More recently we have found that an uncapped "hinge" peptide minilibrary XI-X2Abu-X3, where XI, X2 and X3 are mixtures of Asp, Glu and Arg, contains very potent inhibitors based on the SNARE motif (7). We are currently evaluating these minilibraries for inhibition of other botulinum neurotoxins, to determine if generic therapies based on the SNARE motif are present. In a much larger study, we are undertaking the synthesis of minilibrary subsets deriving from a more complete hinge peptide minilibrary comprising more than 20,000 hinge peptides, in order to ensure that optimal peptides are not overlooked. References [ 1 ] MA Poirier et al. Nature Struct Biol 5 (1998) 765. [2] O Rossetto et al, Nature 372 (1994) 416. [3] MA Hanson and RC Stevens, Nature Struct Biol 7 (2000) 687. [4] GJ Moore, Drug Devel Res 42 (1997) 157. [5] GJ Moore, Trends Pharmacol Sci 15 (1994)124. [6] JJ Schmidt & KA Bostian.,7 Protein Chem 16 (1997) 19. [7] LJ Hayden et al, J Appl Toxicol (2001) in press.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
241
Effect of Linear and Cyclic Peptide Analogues of Myelin Basic Protein Epitope MBP72-85 on Human T-cell Activation S. Thymianou1, K. Chatzantoni1, M. Karakantza1, T. Tselios2, P. Papathanassopoulos3, J. Matsoukas2, A. Mouzaki1 'Laboratory Haematology and Transfusion Medicine, Medical School, University of Patras, GR-26110 Patras, 2 Department of Chemistry, University of Patras, GR-26500 Patras, 3 Neurology Clinic, Medical School, University of Patras, GR-26110 Patras, Greece Abstract. Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) mediated by helper (CD4) T-cells. We have previously shown that two linear and two cyclic peptide analogues of the guinea pig epitope 72–85 of the myelin basic protein (MBP) induce (agonists) or treat (antagonists) an animalmodel version of MS. In this report we studied whether these MBP72-85 are recognized as auto-antigens by normal peripheral blood (PB) T-cells. To this end we cultured PB T-cells in the presence of the petides at various concentrations and assessed their effect on cellular proliferation, phenotype and cytokine synthesis. The peptides, when administered separately, induced apoptosis in exvivo cells and acted synergistically with mitogens to stimulate the cells, acting through the T Cell Receptor (TCR). When administered together (agonist+antagonist), the peptides blocked T-cell proliferation completely without rendering the cells anergic. Cytokine secretion experiments showed that none of the peptides targets a specific Thl or Th2 type of T-cells. Overall, the peptides studied in this work show a promising pharmacological profile and a putative therapeutic use in the treatment of MS.
242
5. Thymianou et al. / Effect of Linear and Cyclic Peptide Analogues
Introduction Multiple sclerosis is a chronic CD4 T-cell mediated disease of the central nervous system characterized by local-cell and macrophage infiltrates, demyelination and loss of neurologic function [1,2]. MS is widely believed to be an autoimmune disease and to be triggered by CNS-specific CD4 T lymphocytes [3–6]. Candidate autoantigens include constituents of the myelin sheath such as myelin basic protein and proteolipid protein (PLP) [7]. Modem approaches towards the therapeutical management of MS involve the design and use of peptide analogues of disease-associated myelin epitopes to induce peripheral T cell tolerance [8]. Experimental autoimmune or allergic encephalomyelitis (EAE), one of the best studied experimental animal models of MS [1,9], is a useful in vivo system for the evaluation of such therapeutic approaches. In Lewis rats immunized with guinea pig MBP protein, encephalitogenic T cells which recognise the 72-85 amino acid sequence MBP772-85, dominate the immune response [10]. The linear analogue Gln-Lys-Ser-Gln-Arg-Ser-GlnAsp-Glu-Asn-Pro-Val (MBP72-85) has been found to induce EAE while substitution of the Asp residue at position 81 with Ala resulted in an analogue (Ala8lMBP72-85) which prevented the induction of EAE by its parent peptide [11,12]. Peptides that interfere with the formation of the trimolecular complex MHC-Peptide-TCR can actively inhibit disease through the activation of antigen-specific regulatory T cells [13,14]. Peptide therapy, however, is hindered due to the sensitivity of peptides to proteolytic enzymes. Continuous infusions and therefore prohibitive amounts of peptides are necessary to elicit the necessary biological response. Cyclization is known to restrict the number of possible conformations allowing to determine which of the conformations observed in solution best approximates the receptor bound conformation. To this end, we synthesized cyclic analogues [15,16] to address the need for more stable molecules, which could maintain or suppress the biological function of the original peptide and yet be able to elicit a response in pharmacological quantities. We recently showed that the cyclic MBP72-85 (c-MBP72-85) analogue has comparable potency to linear MBP72-85 in inducing EAE in Lewis rats and that the clinical and histopathological manifestations of disease induced by C-MBP72-85 are prevented by a linear antagonist (Ala8lMBP72-85) [16]. Furthermore, it was demonstrated that the encephalitogenic activity of MBP72-85 can be completely prevented by the co-injection with c-Ala8lMBP72-85 [17]. In the present study, the linear (MBP72-85 and Ala8lMBP72-85) and cyclic analogues (cMBP72-85 and c-Ala8lMBP72-85) were assessed for their biological activities in cultured human PB T-cells in order to investigate whether any of these peptides can be eventually used as an immunomodifying agent in MS.
Materials and Methods Peptides: In order to assess the biological activity of the MBP72-85 peptide analogues, the following peptides were used: Pl=Glu-Lys-Ser-Glu-Arg-Ser-Glu-Asp8l-Glu-Asn-Pro-Val Linear agonist MBP72-85. [16]; P2=Glu-Lys-Ser-Glu-Arg-Ser-Glu-Ala8l-Glu-Asn-Pro-Val Linear antagonist Ala8lMBP7285. [16]; P4=Glu-Lys-Ser-Glu-Arg-Ser-Glu-Asp8l-Glu-Asn-Pro-Val-NH2 Cyclic agonist cMBP72-85 [16]; I 1 P5= Glu-Lys-Ser-Glu-Arg-Ser-Glu-Ala81-Glu-Asn-Pro-Val Cyclic antagonist cAla81MBP72 85. [17].
I
1
As a negative control we used the peptide P3=Cyc[(D-F)LLReKDap]
S. Thymianou et al. /Effect of Linear and Cyclic Peptide Analogues
243
Cell cultivation: Heparinized venous blood was collected from three randomly selected blood donors (Blood Transfusion Center, Patras University Hospital). Mononuclear cells were prepared by centrifugation over a Ficoll-Paque gradient (Pharmacia, Sweden). The cells were cultured in RPMI1640 culture medium (CM, GIBCO BRL, Gaithersburg, MD, USA) containing 10% Fetal calf serum (PCS) (GIBCO BRL, Gaithersburg, MD, USA) and other supplements as described [18], in the presence or absence of 5 ug/ml/2xlO^ cells phytohaemaglutinine (PHA) (Sigma, St-Louis, MI, USA). When indicated, the cells were cultured in the presence of 1 ug/ml/2x!06 cells of anti-CD28 monoclonal antibody (mAb) (CALTAG Labs, Burlingame, CA, USA). When indicated the cells were cultured with varying doses of linear peptides MBP72-85 or Ala81MBP72-85 or the cyclic peptides C-MBP7285 or c-Ala81MBP72-85 or both linear or cyclic agonist and antagonist peptides at a ratio of 1:100 (see Figure legends). The number of cells was counted using a Sysmex NE-8000 counter (Japan) and their viability was estimated by the trypan blue exclusion method as described [18]. Cell surface immunophenotyping and FACS analysis: Cells obtained from 24 hours cultures were concentrated to 5xl06 cells/ml PBS (GIBCO BRL, Gaithersburg, MD, USA) and 100 ul of the cells were incubated with the following mAb: CD3-FITC, CD4-FTTC, CD8-FITC, CD4-FITC/CD25-PE, CD8-FITC/CD25-PE [19], for 30 min at 4°C. All mAbs were used at saturating conditions as instructed by the manufacturer (all mAb were from Daco A/S Denmark; the CD25-PE mAb was from Serotec ltd., Oxford, UK). Subsequently the cells were fixed using the Immunoprep reagent system of Coulter EPICS and the automated CoulterQ-Prep EPICS Immunology Workstation (Maiami, FL). Negative controls consisted of the sequential incubations as above, substituting specific antibodies with isotype-matched irrelevant controls at equivalent concentrations. The cells were analyzed using a Coulter EPICS XL-MCL. Cell debris were excluded from the analysis. For each sample 20,000 viable cells were acquired in list mode and data on these cells were processed using the EPICS-XL2 software. All the positive results were obtained by substracting the percentage of negative controls. ELISA assays: These were performed as instructed by the manufacturers. The following ELISA kits were used: Human (h) DL-2 ELISA from Boerhinger (Mannheim, Germany), hlFN-y ELISA from R&D Systems Quantikine™ (Minneapolis, USA), hIL-4 and hIL-10 from Endogen Inc. (Woburn, MA, USA). Results Effect of the peptides on human peripheral blood T-cells: The immunogenicity of the linear and cyclic peptides were tested in human adult peripheral blood mononuclear cell (PBMC) cultures in the presence or absence of the mitogen PHA. Dose response curves depicting the number of viable cells versus peptide concentration were constructed 48h later and the results are shown in Figures 1A and 1B. When the peptides were added to the cells without co-stimulation, the cells died reaching minimum numbers at 7nM concentration of any peptide (Fig. 1A and 1B, panels A and B). In contrast, when the peptides were added to the cells together with PHA they increased cellular proliferation rates reaching maximum numbers also at 7nM peptide concentration (Fig. 1A and 1B, panels D and E). These observations indicated that (i) both linear and cyclic analogues had a very similar to almost identical effect when administered to human PBMC and that (ii) all the peptides engage the TCR [20]. To show that this is the case, the cells were cultured in plain culture medium
244
5. Thymianou et al. /Effect of Linear and Cyclic Peptide Analogues
with the peptides in the presence of anti-CD28 mAb and the number of viable cells was estimated 48h later. In this case the number of cells remained constant (Fig. 1A and 1B, panels A and B). Thus, all the peptides tested can influence T cells with various Ag specificities and CD28 co-stimulation prevents cell death upon peptide administration to T cells [21]. Interestingly, when the peptides were added to the cell cultures together at a ratio of agonist:antagonist of 1:100, they had no effect on cellular proliferation regardless whether the cells were stimulated with PHA or not (Fig. 1A and 1B, panels C and F). In effect, the results are similar to those obtained when the cells were cultured in plain culture medium with a peptide alone in the presence of anti-CD28 mAb (Fig. 1A and 1B, panels A and B). To understand how this canceling-out effect was achieved, the following experiments were performed:
•Peptide concentration (nM)
Figure 1A: 1.5xl06 PBMC were cultured for 48h in plain culture medium (CM) (A, B, C) or with 5 ug/ml PHA (D, E, F) and various nM concentrations (as indicated on the X-axes) of the linear agonist peptide MBP72-85 (=P1) (A, D) or the linear antagonist peptide A1a81MBP72-85 (=P2) (B, E). In A and B, identical cultures were set up in the presence of 1 ug/ml anti-CD28 mAb. In C and F, the agonist (PI) and antagonist (P2) peptides were added together at a ratio of 1:100; on the X-axes the concentrations of PI are recorded. At the end of the culture period, the cells were harvested and counted by a Sysmex NE-8000 counter, their viability was estimated by the trypan blue exclusion method and the number of viable cells was calculated and plotted on the Y-axes.
5. Thymianou et al. /Effect of Linear and Cyclic Peptide Analogues
245
Peptide concentration (nM)
Resting PB T-cells
Activated PB T-cells Cyclic MBP72-85
Figure IB: Exactly as in Fig. 1A, with the difference that the cyclic forms of the agonist (c-MBP72-85;:::P4) and antagonist (c-Ala81MBP72-85=P5) peptides were used. A, B, C: Culture in plain CM. D, E, F: Culture in CM(PHA). In A and B identical cultures were set up with anti-CD28 mAb. In C and F the agonist (P4) and antagonist (P5) peptides were added together at a ratio of 1:100.
Effect of peptides on T-cell phenotypes: Cells were cultured with 7nM peptide concentration for 24h and phenotyped using mAb against the CDS, CD4, CDS, CD4/CD25 and CD8/CD25 markers. FACS analysis was performed and the percentage of the viable cells that carry these markers was calculated. The results are summarized in Table 1. Overall, the effect of the peptides on the percentage of the above cell types, compared to the control groups, was slight. An exeption was observed with the cyclic antagonist which increased significantly the CD25 [19] expression on unstimulated CD4 cells. When the agonist and antagonist were administered together, CD25 expression on CD4 cells was diminished. From these results it can be concluded that the canceling-out effect (cf. Fig. 1) observed when the agonist and antagonist peptides are added to the cultures together, is not
246
S. Thymianou et al. /Effect of Linear and Cyclic Peptidc Analogues
due to an alteration of the activity phase of the cells i.e. the resting cells remain so but do not die apoptotically and the activated cells remain so also but do not proliferate. Table 1: Effect of cyclic peptides on peripheral blood T cell phenotype cMBP72-85 cAIa81MBP72-85 Cellular Control group phenotype CM (%) + M ( % ) CM(%) +M(%) CM(%) +M(%) 76,7 82,4 86,7 CD3 67,5 68,8 85,3 CD4 52,7 42,5 58,8 53,2 60,3 43,1 25,7 CD8 24 25 23,6 25 32,8 22,6 10,8 23,6 CD4/CD25 0 2,5 21 10,7 13,2 CD8/CD25 0 0,5 0 11,1
c[Agonist+ Antagonist] CM(%) +M(%) 85,9 65 58,4 40 26,6 25 22,4 3,2
0
8
CM=Culture medium; +M=CM+5 ug/ml PHA; [cMBP72-85] and [cAla8lMBP72-85]=7 nM; c[Agonist+Anatgonist]=[ cMBP7285 + cAla8lMBP72-85]=7 nM + 700 nM
Effect of peptides on cytokine production by PB T-cells: The cells were cultured with or without PHA either alone or in the presence of 7nM of the agonist peptides MBP72-85 cMBP72-85 or 7nM of the antagonist peptides AIa8lMBP72-85, c-Ala8lMBP72-85 or with both agonist + antagonist peptides in linear and cyclic form, at a ratio of 1:100 respectively, for 48h. At the end of this period, supernatants were collected and were measured by ELISA for the Thl-type cytokines IL-2 and IFN-y and the Th2-type cytokines IL-4 and IL-10. The results are summarized in Figure 2 and show that: (i) The agonist or the antagonist peptides (linear or cyclic), when added to the cell cultures separately or together, do not induce the production of any of the above cytokines by the cells, (ii) The agonist and antagonist peptides when administered separately or together to PHA-stimulated cells, induce the production of both the Thl- and Th2-type cytokines tested, though in varying quantities. In all cases, the cyclic antagonist (P4) when administered alone induced the production of by far the highest amounts of IL-2, INF-y as well as IL-4 and IL-10. Interestingly, the simultaneous presence of cyclic agonist and antagonist did not inhibit the production of any of these cytokines by the cells but resulted in a reduction of the quantities of all the cytokines tested to levels similar or lower to the ones obtained when the cyclic agonist was added to the cells alone. Under the same culture conditions, the linear agonist and antagonist inhibited IL-2 production significantly, EL-4 and IL-10 production to a lesser degree, but had no effect on EFN-y production. In conclusion, both agonist and antagonist peptides do not seem to target any particular Th-type in healthy individuals and although cytokines are been produced by stimulated normal T-cells co-cultured with peptides, the cells do not proliferate (cf. Fig. 1A,F). It can be therefore deduced that, when administered together in activated cells, the peptides and especially the cyclic antagonist do not render the cells anergic, because they can still produce cytokines, but they block cellular proliferation.
5. Thymianou et al. /Effect of Linear and Cyclic Peptide Analogues
247
Figure 2: 2X100 PBMC/ml were cultured in CM (white columns) or with 5ug/ml PHA (black columns) for 48h. At the end of the culture period cellular supernatants were analyzed for their content of the cytokines IL2, IFN-y, IL-4 and IL-10 by ELISA. The results, plotted on the Y-axes, are [cytokine] in pg/ml/2x!06 cells. Placebo=cells cultured without peptides. Pl-P5=see M&M.
Discussion EAE is an inflammatory CD4 T cell-mediated disease that can be induced by immunisation with MBP and PLP proteins as well as with peptide epitopes such as the 72–85 amino acid sequence of the guinea pig MBP. Altered peptide ligands, which differ from their parent encephalitogenic peptides by individual amino acid substitutions, have been described which are capable of inhibiting autoimmune-mediated disease. The binding of such altered ligands to the TCR can prevent disease through one of several different mechanisms including TCR antagonism [22], induction of anergy in responding cells [23], or the stimulation of immunoregulatory T cells which can actively modulate disease outcome through the secretion of Th2- and Th0- type cytokines [14,24]. However, the development of alternative molecules that will mimic the immunomodulatory activity of MBP epitope peptides and will maintain an advantage over regular peptides in terms of stability is a necessary step before these molecules can be used for therapeutic purposes. There are two approaches in the development of such molecules. One is based on the knowledge of the immunodominant amino acid residues of the MBP72-85 epitope and involves the construction of a chemical moiety in which the important pharmacophoric groups will be incorporated. Such approach may lead to nonpeptide mimetics or semimimetic peptides with immunomodulatory activity [15]. The other approach is the design and synthesis of potent cyclic MBP72-85 analogues which offer several advantages such as increased resistance to metabolic degradation and consequently
248
5. Thymianou et al. /Effect of Linear and Cyclic Peptide Analogues
longer duration of action. Due to restriction of conformational flexibility, constraint peptide analogues of high potency offer important structural information towards the bioactive conformation assumed by the peptide and could be very useful in drug design for the development of non-peptide mimetics [25,26]. Such approaches have been successful in the design and synthesis of non-peptide mimetics for peptide hormones such as Angiotensin II which is implicated in cardiovascular diseases [27-31] or for motifs of proteins such as Thrombin implicated in haemostasis and angiogenesis [32–34,25,26]. This computer-aided novel technology may eventually be applied in generating mimetic molecules of peptides with immunomodulatory ability. The guinea pig analogue of MBP72–85 peptide that induces transient EAE in Lewis rats was used as a reference peptide to synthesize the cyclic analogues which is an important intermediate step towards non-peptide mimetics. We have recently shown that a cyclic analogue of MBP72-85 (c-MBP72-85) is a potent inducer of EAE and that this cyclic molecule has a comparable potency to that of the linear analogue MBP72-85 [16]. These findings were extended to show that a cyclic analogue of the linear antagonist, c-Ala81MBP72-85, is a potent inhibitor of EAE when co-injected with MBP72-85 85 in Lewis rats: EAE induced by linear MBP72-85 was completely suppressed when it was co-injected with the cyclic antagonist c-Ala8lMBP72-85 [17]. EAE induced by cyclic analogue c-MBP72-85 was also completely suppressed when it was co-injected with the linear analogue Ala8lMBP72-85 [16] or the cyclic analogue c-Ala8lMBP72-85 [17]. To begin to assess the possibility that the cyclic antagonist c-Ala8lMBP72-85 can be eventually used in human MS patients, the effect of linear MBP72-85 and Ala8lMBP72-85 and cyclic c-MBP72-85 and c-Ala8lMBP72-85 peptides was analysed separately or in combination in human T-cell cultures. The rationale behind using peripheral blood cells obtained from randomly selected healthy individuals, was that first we had to establish whether these peptides work in the human system and then to deduce whether there is any point to test them in cells derived from MS patients. In addition, MS patients express a variety of TCRspecificities other than the autoreactive T-cell clones, and therefore it is important to know whether these peptides affect their survival and function. Furthermore, if the peptides cause a deleterious effect in normal T-cells their use becomes unlikely for therapeutic purposes in patients with an already established autoimmune disease with a Thl bias [35]. In MS patients, the majority of myelin Ag-specific T cells isolated during active disease secrete Thl-type cytokines while during remission the balance shifts to DL-4 and IL-10 producing cells [35]. The results shown here raise the following major points: (i) Both agonist and antagonist peptides act through the TCR. (ii) If the peptides under study are given to unstimulated cells they force them to die of apoptosis. (iii) If the peptides are administered together to cell cultures from PBMC without an additional stimulus, apoptosis is prevented completely but the cells are not stimulated. This fact can be explained by the hypotheses that the peptides either compete with each other for the same binding site and none can bind to the TCR or, otherwise, they render the cells anergic. This is to be clarified in future experiments, (iv) When the cells are PHA-activated, the simultaneous administration of the agonist and antagonist peptides to the cells seems to result to a qualitatively different activation of T cells so that cytokine production is induced without the concomitant proliferation of the cells. The above observations are reminiscent of previous studies performed with altered peptide ligands that have been shown to engage the TCR and induce cytokine production or cytolytic activity without the proliferation of the T cells induced by the wild-type agonist peptide [36,37]. An alternative explanation for the failure of activated T cells to proliferate in the presence of both agonist and antagonist peptides is that the simultaneous secretion of IL-2, IFN-y, IL-4 and IL-10 cytokines is in such quantities that apparently neutralize each other's action. Experiments are in progress aiming to show whether the peptides induce secretion of different cytokines from different PB T-cells or whether they alter the pattern
S. Thymianou et al. /Effect of Linear and Cyclic Peptide Analogues
249
of cytokine production in the same cells [38–40]. Finally, another parameter to be studied is the fact that this canceling out effect of the agonist and antagonist peptides on T-cell proliferation and also in the EAE animal model takes place when the ratio between agonist:antagonist is 1:100 (found to be the optimal ratio in preliminary experiments-data not shown). This could mean that indeed this is the required ratio for the antagonist to counteract the effect of the agonist that causes the disease (ie. the antagonist has a much lower affinity than the agonist). But it could also mean that it is the overall high peptide dose that causes a high dose supression effect or high zone tolerance observed also in other studies of EAE [41]. Acknowledgments This work was supported by the Greek Ministry of Energy and Technology (EPET II, PENED 99–115). T. Tselios is a recipient of a grant by the Leonidas Zervas foundation for MS. References [1] R. Martin, H. McFarland and D. McFarlin, Ann. Rev. Immunol. 10 (1992) 153. [2] L. Steinman, Cell 85 (1996) 299. [3] M. Pette, K. Fujita, D. Wilkinson, D. Altmann, J. Trowsdale, G. Giegerich, A. Hinkkanen, J. Epplen, L. Kappos and H. Wekerle, Proc. Natl. Acad. Sci. USA 87 (1990) 7968. [4] D. Hafler and H. Weiner, Immunol. Rev. 144 (1995) 75. [5] K. Ota, M. Matsui, E. Milford, G. Mackin, H. Weiner and D. Hafler, Nature (Lond) 346 (1990) 183. [6] R. Martin, D. Jaraquemada, M. Flerlage, J. Richert, J. Whitaker, E. Long, D. McFarlin and H. McFarland, J. Immunol. 145 (1990) 540. [7] J. Greer, P. Csurhes, K. Cameron, P. McCombe, M. Good and M. Pender, Brain 8 (1997) 1447. [8] R. Hohfeld, Brain 120 (1997) 865. [9] S. Zamvil and L. Steinman, Ann. Rev. Immunol. 8 (1990) 579. [10] Y. Chou, A. Vandenbark, R, Jones, G. Hashim and H. Offner, J. Neuros. Res. 22 (1989) 181. [11] M. Wauben, C. Boog, R. Zee, I. Joosten, A. Scihlief and W. Eden, J. Exp. Med. 176 (1992) 667. [ 12] R. Smeitz, M. Wauben, N. Wolf, and R. Swanborg, J. Immunol. 162(2) (1999) 829. [13]M. Vergelli, B. Hemmer, U. Utz, A. Vogt, M. Kalbus, L. Tranquill, P. Conlon, N. Ling, L. Steinman, H. H. McFarland and R. Martin, Eur. J. Immunol. 26 (1996) 2624. [14]S. Brocke, K. Gijbels, M. Allegretta, I. Ferber, C. Piercy, T. Blankenstein, R. Martin, U. Utz, N. Karin, D. Mitchell, T. Veroma, A. Waisman, A. Gaur, P. Conlon, N. Ling, P. Fairchild, D. Wraith, A. O'Carra, G. Farhman and L. Steinman, Nature 379 (1996) 343. [15]T. Tselios, L. Probert, G. Kollias, E. Matsoukas, P. Roumelioti, K. Alexopoulos, G. Moore and J. Matsoukas, Amino Acids 14 (1998) 333. [16]T. Tselios, L. Probert, I. Daliani, E. Matsoukas, A. Troganis, P. Gerothanassis, T. Mavromoustakos, G. Moore and J. Matsoukas, J. Med. Chem. 42 (1999) 1170. [17]T. Tselios, I. Daliani, S. Deraos, S. Thymianou, E. Matsoukas, A. Troganis, I. Gerothanassis, A. Mouzaki, T. Mavromoustakos, L. Probert and J. Matsoukas, Bioorganic & Medicinal Chemistry Letters 10 (2000) 2713. [18] Mouzaki, D. Rungger, A. D. Tucci, A. Doucet, R. H. Zubler, Eur. J. Immunol. 23 (1993) 1469. [19] Barclay, M. Brown, S. Law, A. McKnight, M. Tomlinson and P. van der Merwe, (Eds.), Leukocyte antigen Facts book. Academic Press, Harcoun Brace and Co, (1997) pp.486–489. [20]M. Alderson, T. Tough, T. Davis-smith, S. Braddy. B. Falk, K. Schooley, R. Goodwin, C. Smith. F. Ramsdell and D. Lynch, J. Exp. Med. 181 (1995) 71. [21] P. Noel, L. Boise, J. Green and C. Thompson, J. Immunol. 157 (1996) 636. [22] V. Kuchroo, J. Greer, D. Kaul, G. Ishioka, A. Franko, A. Sette, R. Sobel and M. Lees, J. Immunol. 153(1994)3326. [23] J. Sloan-Lancaster, B. Evavold and P. Allen, Nature 363 (1993) 156. [24] L. Nicholson. J. Greer, R, Sobel, M. Lees and V. Kuchroo, Immunity 3 (1995) 396. [25] K. Alexopoulos. P. Fatseas, E. Melissari. D. Vlahakos, J. Smith, T. Mavromoustakos, M. Saifeddine. G. Moore, M. Hollenberg and J. Matsoukas, Bioorg. Med. Chem. 7 (1999) 1.
250
S. Thymianou et al. / Effect of Linear and Cyclic Peptide Analogues
[26] K. Alexopoulos, J. Matsoukas, T. Tselios, P. Roumelioti, T. Mavromoustakos and K. Holada, Amino Acids 15 (1998) 211. [27] J. Matsoukas, J. Hondrelis, G. Agelis, K. Barlos, D. Gates, R. Ganter, D. Moore and G. Moore, J. Med. Chem. 37 (1994) 2958. [28] J. Matsoukas, R. Yamdagni and G. Moore, Peptides 11 (1990) 367. [29] J. Matsoukas, G. Bigam, N. Zhou and G. Moore, Peptides 11 (1990) 359. [30]J. Matsoukas, J. Hondrelis, M. Keramida, T. Mavromoustakos, A. Makriyiannis. R. Yamdagni, Q. Wu and G. Moore, J. Biol. Chem. 269 (1994) 5303. [31]D. Vlahakos, J. Matsoukas, J. Ancans, G. Moore, E. Iliodromitis, K. Marathia and D. Kremastinos, Let. Pept. Sci. (LiPS) 3 (1996) 191. [32] J. Matsoukas, J. Hondrelis, G. Agelis, K. Barlos, D. Gatos, R. Ganter, D. Moore and G. Moore, J. Med. Chem. 37 (1994) 2958. [33] D. Panagiotopoulos, J. Matsoukas, K. Alexopoulos, A. Zebeki, T. Mavromoustakos, M. Saifeddine and M. Hollenberg, Let. Pept. Sci. (LiPS) 3 (1996) 233. [34]J. Matsoukas, M. Hollenberg, T. Mavromoustakos, D. Panagiotopoulos, K. Alexopoulos, R. Yamdagni, Q. Wu and G. Moore, J. Protein Chem. 16 (1997) 113. [35] J. Correale, W. Gilmore, M. McMillan, S. Li, K. McCarty, T. Le and LP. Weiner, J. Immunol. 154 (1995)2959. [36]Evavold, J. Sloan-Lancaster and P. Allen, Immunol. Today 14 (1993) 602. [37] S. Valitutti and A. Lanzavecchia, Immunol. Today 18 (1997) 299. [38] V. Apostolopoulos, G. Pietersz and I. McKenzie, Vaccine 9 (1995) 930. [39] S. Lofthouse, V. Apostolopoulos, G. Pietersz, W. Li and I. McKenzie, Vaccine 14 (1997) 1586. [40] V. Apostolopoulos, G. Pietersz, B. Loveland, M. Sandrin and I. McKenzie, Proc. Natl. Acad. Sci. USA 92(1995)10128. [41]J. Critchfield, M. Racke, J. Zuniga-Pflucker, B. Cannella, C. Raine, J. Goverman and M. Lenardo, Science 263 (1994) 1139.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
251
Use of Synthetic Peptides for the Identification of the Biologically Active Domains of the Growth Factor HARP E. Papadimitriou1, A. Polykratis2, E. Karestou1, J. Courty3, and P. Katsoris2 !
Lab of Molecular Pharmacology, Department of Pharmacy, University ofPatras, Greece, Lab of Cell Biology, Department of Biology, University ofPatras, Greece and 3 Lab de Recherche sur la Croissance Cellulaire, la Reparation et la Regeneration Tissulaires, Universite Paris XII, France.
Abstract. HARP (Heparin Affin Regulatory Peptide) is a growth/differentiation secreted protein with distinct lysine-rich clusters within both the NH2- and COOH-terminal domains and apparent molecular weight 18 kDa. It was initially purified from bovine uterus and neonatal rat brain and found to be highly conserved among species. It has a high affinity for heparin and is localized in the extracellular matrix through interactions with glycosaminoglycans. It is expressed in developing tissues and displays important function in the growth and differentiation processes. The purpose of the studies presented here was to define the effect of HARP on endothelial cell functions. We studied the effect of human recombinant HARP expressed in a bacterial expression system, as well as two HARP structurally related synthetic peptides (HARP residues 1–21 and residues 121–139), on different endothelial cell types derived from human umbilical vein (HUVEC), rat adrenal medulla (RAME), bovine retina capillaries (BREC) and bovine brain capillaries (BBC). We also tested the angiogenic effect of HARP and peptides on the in vivo system of the chicken embryo chorioallantoic membrane (CAM). Both peptides had a dose-response mitogenic effect whether added to the culture medium or adsorbed onto the tissue culture plate. The effect was at least two times more intense in the latter case. HARP seems to be mitogenic only when coated onto the cell culture plate, the only exception being the RAME cells. It is worth noticing that BBC cells, which are the most responsive to HARP, release the higher amounts of this growth factor into the culture medium, compared to the other types of cells. Both HUVEC and BBC endothelial cells express HARP mRNA and the expression changes at different levels of cell confluency. HARP, as well as both peptides, induce angiogenesis in vivo, exhibit chemotactic action and induce capillaries formation on collagen and fibrin gels, as well as on matrigel. These actions are dose dependent and different among studied cells, elucidating to a degree the implication of HARP terminal regions on endothelial cell biology and angiogenesis.
252
E. Papadimitriou et al. / Use of Synthetic Peptides
Introduction HARP (Heparin Affin Regulatory Peptide), also called pleiotrophin or HB-GAM (Heparin Binding - Growth Associated Molecule) is a new growth factor with high affinity to heparin [1, 2]. Along with Midkine, it belongs to a new family of heparin binding growth factors [3]. It consists of 168 amino acids -the first 30 corresponding to a signal peptide indicating a secretive protein- and has an approximate molecular weight of 18 kDa. There are no indications of post-translational modifications of the molecule. The mature form of the protein consists of 135 or 136 amino acids, depending on the species. It is highly basic, with 26 lysines forming two clusters at both termini of the molecule [1]. These amino acids contribute to the basicity of HARP and match with a consensus sequence involved in heparin-binding and angiogenic activity [4]. The presence of 10 cysteines at the central region of the molecule is characteristic, highly conserved and contributes to the formation of the tridimensional structure of the molecule through five intrachain disulphide bonds. HARP contains two p-sheet domains, homologous to the thrombospondin type I repeat (TSR), which are connected by a flexible linker. Each one of these domains contains three antiparallel p-strands (Figure I) [5].
Figure 1. The HARP molecule. The picture shows the two lysine-rich terminal domains, the two B -sheet domains and the five intrachain disulflde bonds.
HARP was originally purified from uterus [6], neonatal rat brain [1] and placental tissue [7]. It seems to be implicated in growth and differentiation processes and is expressed in several developing tissues [1, 8–10]. In adults, HARP is expressed in neuronal tissues, heart, uterus, cartilage and bone [3, 6, 9, 10], indicating that it also has important physiological role(s) during adulthood. This is further strengthened by the fact that HARP is highly conserved among species, with an overall homology reaching 98%. Although differences can be found at the signal peptide, no significant differences are observed in the mature molecule [11]. HARP regulation remains poorly documented. HARP mRNA is increased by the platelet-derived growth factor and basic fibroblast growth factor [7], retinoic acid [12], progesterone [13] dihydrotestosterone, testosterone and estrogen [14]. HARP may also be considered as a marker of neuronal injury [15]. Endothelial cells express HARP mRNA (Figure 2) and secrete the protein both in their conditioned medium and in the extracellular matrix [16]. Human recombinant HARP produced in a bacterial expression system downregulates the expression of HARP mRNA in endothelial cells (Figure 2). This is a strong indication that the molecule produced in bacteria is recognized by specific molecule(s) on the surface of endothelial cells and thus exerts its biolosical functions.
E. Papadimitriou et al. / Use of Synthetic Peptides
253
Figure 2. (A) RT-PCR reactions for human GAPDH and HARP mRNA levels in human umbilical vein endothelial cells (HUVEC) at several time points after stimulation with human recombinant HARP produced in a bacterial expression system. A representative image of 5 independent experiments is shown. (B) Expression of HARP mRNA as % ratio of HARP/GAPDH electrophoretic band values compared to controls (unstimulated cells). Results are mean ± S.E.M. from 5 independent experiments. * P<0,05.
Initially, HARP was described as a molecule that induces neurite outgrowth of neuronal cells [1] and its ability to promote neurite outgrowth is indisputable [8]. Furthermore, it is mitogenic for a variety of cells, although results concerning its mitogenic activity are controversial, depending on the cell type and the experimental conditions used [16–19]. Recombinant HARP expressed in eucaryotic cells is mitogenic, whereas HARP expressed in prokaryotic expression system, although promotes neurite outgrowth, was considered not to have any mitogenic effect [3]. We have recently shown that human recombinant HARP expressed in bacteria is mitogenic for endothelial cells only when presented to them as a substrate, while it does not influence endothelial cell proliferation when it is added to the medium of the cells [16]. The endothelial cells used are BBC (bovine brain capillary cells), RAME (rat adrenal medulla endothelial cells) and HUVEC (human umbilical vein endothelial cells).
Figure 3. Effect of human recombinant HARP expressed in bacteria and of two peptides, corresponding to its COOHand NH 2 -termini, on the proliferation of BBC cells. Different concentrations of the test agents were added to the cell culture medium of the cells (A) or coated onto the corresponding wells of the cell culture plates (B). Cells were further incubated for 48 h before counted, using the MTT assay [16). *P< 0.05, "P<0.0! and ***P<0.001 [16].
254
E. Papadimitriou et al. / Use of Synthetic Peptides
Two peptides that correspond to the NH2 and COOH termini of the protein (HARP residues 1-21 and 121-139 respectively) are mitogenic for all the types of endothelial cells used, in a concentration-dependent manner and the effect is more intense when the substances are coated onto the plate (Figures 3, 4 and 5). These two regions represent the lysine-rich domains of the molecule and are thought to be involved in multiple biological actions, since they are also required for successful transformation of NTH 3T3 cells by HARP residues 41–64 [20]. Recently, it was shown that the residues 111-136 of the lysine-rich COOHterminal domain are involved in the mitogenic and tumour formation activities of HARP and play a role in the binding of HARP to its high affinity receptor. The same residues are not implicated in the neurite-outgrowth activity [21], supporting the notion that the domains involved in HARP mitogenic and neurite outgrowth activities are different. The amino acids 111-128 seem to play a key role, since a mutant that lacks residues 129–136 is still mitogenic for Chinese hamster ovary cells [21].
icentrabon (ng/ml)
Figure 4. Effect of human recombinant HARP expressed in bacteria and of two peptides, corresponding to its COOHand NH2 -termini, on the proliferation of RAME cells. Different concentrations of the test agents were added to the cell culture medium of the cells (A) or coated onto the corresponding wells of the cell culture plates (B). Cells were further incubated for 48 h before counted, using the MTT assay [16]. *P<0.05, **P<0.01 and ***P
HARP may also be an important regulator of tumour transformation. It is detected in various carcinomas, such as human breast and prostate cancer, neuroblastomasr benign meningiomas, small cell lung cancer and rat mammary tumours [3, 22] and is involved in tumour growth and metastasis [22–24]. Many of the HARP expressing tumours become more invasive when HARP is overexpressed and reduction of HARP levels reduces the number of blood vessels in primary tumours, indicating a possible role of HARP in tumour angiogenesis [23, 25].
E. Papadimitriou et al. / Use of Synthetic Peptides
10
100
concentration (ng/well)
1000
255
Figure 5. Effect of human recombinant HARP expressed in bacteria and of two peptides, corresponding to its COOHand NH2 -termini, on the proliferation of HUVEC cells. Different concentrations of the test agents were added to the cell culture medium of the cells (A) or coated onto the corresponding wells of the cell culture plates (B). Cells were further incubated for 48 h before counted, using the MTT assay [16]. *P< 0.05 and ***P<0.001 [16].
Angiogenesis implicates many different actions, such as cell adhesion, migration, proliferation, differentiation and degradation of the basal lamina. HARP affects many endothelial cell processes involved in angiogenesis [26]. The two peptides that correspond to the NH2- and COOH- termini of the protein, have a similar effect in most of the systems studied. HARP stimulates the formation of cord-like structures by BBC and bovine retinal endothelial cells on collagen gels, while neither peptide has any significant effect. On matrigel, where both cell migration and differentiation take place, the two terminal peptides have a stimulating effect on tube formation by BBC and RAME and no effect on HUVEC, similarly to the effect of the whole molecule of HARP [26]. In the same line, HARP and both peptides stimulate migration of BBC and RAME cells, but have no significant effect on HUVEC (Figure 6). In vivo, in the chicken embryo chorioallantoic membrane (CAM) model, both HARP terminal peptides have an angiogenic effect, however, only in the case of the COOH peptide this effect is dose-dependent. The NH2 peptide has a weaker angiogenic effect, which was statistically significant only at one dose (Figure 7). The mechanism of action of HARP or its terminal peptides is not yet clarified and it is not known if it is a direct effect through binding to cell surface receptors or an indirect effect, potentiating the angiogenic effect of other growth factors.
256
E. Papadimitriou et al. / Use of Synthetic Peptides
Figure 6. Effect of human recombinant HARP and two peptides (loOng/mI), corresponding to its COOH- and NH2 - termini, on migration of HUVEC (A), R A M E (B) and BBC cells (C), using the Boyden chamber assay. Results are expressed as percent of the values obtained without stimulation. Data are the mean f SEM of at least three independent experiments. *Pc0.05,**P
In conclusion, HARP synthetic peptides representing native molecule residues 1-21 and 121-139 seem to be involved in multiple biological actions, since they stimulate endothelial cell proliferation 116, 211 and angiogenesis [26] and are required for successful transformation of cells by a central region of HARP [20]. These results suggest that minimal structures could be sufficient to trigger endothelial cell activation, if they are suitably presented to the cell. Whether these or similar peptides exist physiologically (e.g. after proteolysis of HARP) is not known at present and is under investigation.
Figure 7. Effect of human recombinant HARP and two peptides, corresponding to its COOH- and NH2- termini, on angiogenesis in the in vivo model of chicken embryo CAM. Data are the mean f SEM of three independent experiments, each one with 20 eggs tested for each concentration used. *P<0.05, **PcO.Oland ***P
E. Papadimitriou et al. / Use of Synthetic Peptides
257
References [ 1] H. Rauvala, EMBO J. 8 (1989) 2933. [2] F. Vacherot et al, J. Biol. Chem. 274 (1999) 7741. [3] N. Zhang and T. F. Deuel, Curr. Opin. Hematology 1 (1999) 44. [4] J. Courty et al., Heparin-Affin Regulatory Peptide, HARP. In: Comprehensive Vascular Biology and Pathology - An Encyclopedic Reference, Springer-Verlag, 2000, pp. 145-152. [5] I. Kilpelainen et al., J. Biol. Chem. 275 (2000) 13564. [61 P. G. Milner et al., Biochem. Biophys. Res. Commun. 165 (1989) 1096. [7] Y.-S. Li et al., J. Biol. Chem. 267 (1992) 26011. [8] Y.-S. Li et al., Science 250 (1990) 1690. [9] J. M. Vanderwinden et al.,Anat. Embryol 186 (1992) 387. [10] I. Silos-Santiago et al., J. Neurobiol. 32 (1996) 283. [ 11 ] B. S. Hampton et al, Mol. Biol Cell 3 (1992) 85. [12] P. Bohlen et al, Growth Factors 4 (1991) 97. [13] P. E. Milhieteral,J. Endocrinol. 158(1998) 389. [14] F. Vacherot et al, In vitro Cell Dev. Biol Anim. 31 (1995) 647. [15] A. Takeda et al, Neuroscience 68 (1995) 57. [16] E. Papadimitriou et al, Biochem. Biophys. Res. Commun. 274 (2000) 242. [17] J. Courty et al, Biochem. Bioph. Res. Commun. 180 (1991) 145. [ 18] W. Fang et al.J. Biol. Chem. 267 (1992) 25889. [19] E. Szabat and H. Rauvala, Dev. Biol. 178 (1996) 77. [20] N. Zhang et al, J. Biol. Chem. 274 (1999) 12959. [21] I. Bernard-Pierrot etal.J. Biol. Chem. 276(2001) 12228. [221 A. K. Chauhan et al, Proc. Natl. Acad. Sci. USA 90 (1993) 679. [23] F. Czubayko et al, Proc. Natl. Acad. Sci. USA 93 (1996) 14753. [24] N. Zhang et al, J. Biol Chem. 272 (1997) 16733. [25] R. Choudhuri et al.. Cancer Res. 57 (1997) 1814. [26] E. Papadimitriou et al, Biochem. Biophys. Res. Commun. 282 (2001) 306.
258
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Unusual Interactions between MHC, peptide and the T cell Receptor V. Apostolopoulos1 and M. Plebanski2 'Head, Immunology and Vaccine Laboratory, Head, Vaccine Development and Infectious Disease Laboratory, The Austin Research Institute, Studley Road, Heidelberg, VIC 3084, Australia.
2
Abstract. The major histocompatibility complex on the surface of antigen presenting cells presents peptides to the T cell receptor. Recognition of peptide-MHC by T cells activates them as well as initiating a sequence of signals in T cells which enables a T cell dependent/specific immune response. For vaccine design, it is important to understand how peptides bind to MHC class I molecules. In this review, we will discuss, some unusual features of peptides binding to MHC class I, crystal structures of low and high affinity peptides lacking canonical anchor motifs in complex with H-2Kb, peptide mimics in cancer and the role of altered peptide ligands in the immune response in particular in cancer and in infectious diseases.
Role of the MHC in the cellular immune response The T cell receptor (TCR) interaction with major histocompatibility complex (MHC) molecules is a central event in T cell-mediated responses. During thymic development, MHC molecules educate T cells, guiding their positive and negative selection. Outside the thymus, protein antigens from viral or bacterial sources are presented in the context of MHC molecules to T cells [1] after intracellular processing to peptides [2]. TCRs on lymphocytes recognize foreign antigens bound to the MHC and can produce cytokines, proliferate or cause cytotoxic cell lysis in the periphery in response to these stimuli. The MHC loci encode Class I and Class II MHC molecules. Class I MHC are found on the surface of nucleated cells [3] where they form non-covalently linked heterodimers between an MHC-encoded, membranespanning variable heavy chain (44 kDa) and a non MHCencoded, invariant light chain, (52 microglobulin (P2M,
Peptide
Figure 1: Structure of MHC class (H-2Kb in complex with MUC1recently determined in our laboratory.
V. Apostolopoulos and M. Plebanski / Unusual Interactions
259
12 kDa) (Figure 1). Class I molecules bind short peptides (usually -8-10 amino acids), mostly derived from endogenously synthesised proteins but also from exogenous antigens, in the endoplasmic reticulum and present them to cytotoxic T lymphocytes (CTLs) that express the CDS co-receptor. Since an individual expresses only a limited number of MHC molecules, these antigen-presenting molecules must be able to interact with a large diversity of peptide sequences. Nevertheless, the binding of peptides cannot be overly "promiscuous" since individual MHC molecules have their own particular peptide-binding motifs [4]. Specificity of MHC class I: The peptide binding groove of MHC class I can be subdivided into various pockets (A-F) [5]. For MHC class I, peptide length varies from 8-9-mers (H2Kb, HLA-A2) and up to 13-mers (HLA-Aw68) [6]. Early biochemical studies indicated the presence of conserved (consensus) sequences in high affinity binding peptides eg. for H-2Kb, P2, P6, P9 for 9mers or P2, P5, P8 for Smers it was found that the preferred anchors are Phe/Tyr for the central P5/6 residues, Leu/Met at the C-terminus (P8/9), and, in some instances, Tyr at P3 (these were found by acid elution from purified Class I molecules). In this process, low affinity peptides were lost and do not feature in the considerations of Rammensee and colleagues who have set the initial set of useful "rules" for peptide binding for Class I based on the sequence findings [6]. Crystallographic studies indicated that the conserved region were hydrophobic "anchors" - which bound in "hydrophobic" pockets. The non-conserved (ie. variable region) of peptides were likely to be presented to T cells. Although, these anchor residues are required for stabilisation and high affinity binding of peptide to MHC, it has been shown that some peptides which do not contain the respective canonical anchor residues can still bind and be presented by MHC class I and cell lysis still occurs [7–9]. Until now, it has not been clear how such non-standard or low affinity peptides are presented by MHC class I molecules in order to serve as targets for CTL. We have recently determined the crystal structures of two non-canonical anchor motif peptides (one with low affinity and the other with high affinity binding) in complex with H-2K . Low affinity peptides binding to MHC class I: their use in immune responses: High affinity peptides which bind to MHC class I molecules usually induce high avidity CTL. However, the use of high affinity peptides from tumour antigens may not be effective since most tumour antigens are self antigens, and their specific CTL repertoire would most likely be deleted. Thus, for immunotherapy it is conceivable that the best candidate peptides for immunisation are the low affinity binding peptides. A major problem to the use of low affinity peptide epitopes for immunotherapy is the difficulty of their identification due to their poor immunogenicity. It has been shown that the affinity of peptide binding to MHC and the stability of the peptide-MHC complex can correlate with overall immunogenicity [10, 11]. Furthermore, low affinity peptides cannot be detected by elution studies or by prediction algorithms. Thus, peptide identification by systematic binding studies and CTL assay is required for their identification. It is likely that enhancement of the immunogenicity of low affinity MHC binding peptides, is possible by mutations of the peptide anchor side chains. Low affinity MUC1 peptides presented by H-2Kb (SAPDTRPAP) and H-2Db (APGSTAPPA) were not able to stabilise RMA-S cells as determined by FACS, however they were recognised by peptide specific CTL [9]. Mutations of SAPDTRPAP to SAPDTYPAL and APGSTAPPA to APGSNAPPA were now able to stabilise class I on RMA-S cells [9]. Furthermore, the H-2Kb low affinity peptide, MUT1 (a peptide from Lewis lung carcinoma), when mutated at P3, P5 and P8, has increased stability and affinity of peptide for RMA-S cells [12]. In addition, the low affinity MUC1 peptide
260
V. Apostolopoulos and M. Plebanski / Unusual Interactions
(SAPDTRPAP) that binds to HLA-A2 has been converted by mutation to high affinity and are currently being used in a clinical setting [8]. Until recently, no structural information was available on the binding of nonstandard peptides to MHC class I molecules, and whether they differed substantially in their interaction with the MHC and, therefore, with the T cell receptor. We recently determined the crystal structure of two peptides lacking canonical anchor motifs bound to MHC class I, H-2Kb; an 8-mer MUC1 peptide (MUC1-8) and the other a 9-mer yeast peptide [YEA9]. Crystal structure of a low affinity peptide in complex with MHC class I, H-2ff : MUC1-8 (SAPDTRPA) derived from Mucinl does not contain Phe/Tyr at P5 or Leu/Met at P8 which are usually associated with high affinity binding to H-2Kb [4, 6]. MUC1-8 has small residues at P2 (Ala), P5 (Thr) and P8 (Ala) [9] and has a relatively low affinity, compared to high affinity H-2Kb binding peptides. We recently determined the crystal structure of MUC1-8 in complex with H-2K at 1.6A resolution [13, 14]. The side chains of residues that point into the C and F pockets are small; hence, these anchor pockets are not completely filled and a large cavity is formed, as compared to high affinity OVA8 or VSV8 peptides. [13, 14]. Thus, this relatively low affinity peptide binds with the same overall features to MHC class I as high affinity peptides, in that, the peptide starts and finishes at the same position at the N- and C- termini; deviations occur within the central region of the peptide. The small peptide anchoring side chains at P2, P5 and P8, and a cavity at the side of the C pocket, appear to contribute to the lower affinity and of MUC1-8 with H-2Kb. Crystal structure of a relatively high affinity peptide lacking canonical anchor motifs bound to H-2K b : During expression of MHC class I, H-2Kb, in Drosophila melanogaster fly cells, yeast peptides added by the manufacturers to the culture medium, stabilise H-2Kb and aid in its purification. The peptide found in the peptide-binding groove is SRDHSRTPM, or YEA9 [15]. YEA9 contains large residues at Arg-P2, Arg-P6 and Met-P9 at the anchor sites as compared to the preferred Ala/Gly/Ile-P2, Phe/Tyr-P6 and Val/Met-P9. We recently determined the crystal structure of YEA9 in complex with H-2Kb at 1.5A resolution [13, 14]. The non-canonical anchor motif peptide, YEA9, bound with relatively high affinity by insertion of long non-canonical anchor residues into the B and E pockets. This is the first description of the use of alternative anchors for high affinity peptide binding. It is clear that peptides lacking canonical anchor motifs can bind to MHC class I with either low or relatively high affinity, and that all P2, P6 and P9 residues contribute in anchoring the peptide into the peptide-binding groove. These studies are important as they show that binding of peptide with high affinity can occur in two ways: (i) by canonical hydrophobic anchors binding in the C and F pockets and (ii) by non-canonical anchors binding in the B, E and F pockets. Other features of peptides binding to MHC class I Binding of longer peptides to MHC class I: MHC class I molecules preferentially bind peptides 8–10 amino acids long. MHC class I, H-2Kb binds peptides 8 or 9 amino acids long, and the mechanism of binding involves a bulge at the center of the peptide, leaving. the N and C termini unchanged [15, 16]. However, studies have indicated that longer peptides can be accommodated by MHC class I [11, 17]. It has been implicated that the mechanism of binding of longer peptide involves creating a bulge at the center of the peptide, leaving the N and C termini interactions unchanged. However, longer peptides may be accommodated by MHC class I by protrusion, primarily at the C-terminus. The
V. Apostolopoulos and M. Plebanski / Unusual Interactions
261
crystal structure of a 10-mer peptide bound to HLA-A2 showed a single glycine C-terminal extension extending from the binding groove [18]. Peptides with the addition, at the N- or C-termini, of two amino acids of the VSV8 and OVA8 peptides bind to H-2Kb [19]; four residues could be added at the C- but not at the N-terminus of a VSV8 peptide bound to H2Kb [17]. A peptide extended at the C-terminus by 10 amino acids (an 18-mer) binds to H2Kk as it could be eluted from the MHC class I molecule [20]. Recently, it was demonstrated that extensions could occur at either the N- or C-terminus for H-2Kk, H-2Kb and H-2Db molecules [11]. T cells were able to identify the extension; thus, Reptide extensions may play a role in the specificity of the T cells [11]. We have demonstrated that H-2Kb can bind SAPDTRPAP peptides that are extended by up to 5 residues at the Cterminus (SAPDTRPAPGSTAP; MUC1-14). How long peptides are presented by MHC and recognised by the TCR is of importance for the design of alternative peptides in vaccination protocols. In addition, it will be important to determine whether such peptides "loop" in the middle (as recently seen with the rat MHC) or "hang" out from the MHC at either the N- or C-terminal ends. Binding of shorter peptides to MHC class I: It has been demonstrated that immunisation of mice with mannan-MUCl generates CTL that recognise both peptide-pulsed and MUC1transfected target cells that protect mice against a MUC1+ tumour challenge [21]. MUC1-9 presented by H-2Kb, can also be presented and recognised by CTL as a 5-mer, 6-mer, 7-mer and 8-mer (deletion at the C-terminus) when pulsed on RMA-S target cells [Apostolopoulos et al., 2001, submitted, J. Molecular Biology]. The 4-mer, SAPD, is not presented nor lysed by CTL. Other short immunogenic peptides for class I have been demonstrated to bind to H-2Ld and to be recognized by CTL, including, a 3-mer (QNH), 4mers (QNHR, ALDL, PFDL) and 5-mers (RALDL, HFMPT) [22]. These observations are clearly outside the normal structural guidelines for tight binding of class I peptides, deduced from crystal structures of many peptide-MHC complexes. How short peptides are presented by MHC class I molecules and recognised by the TCR is currently unknown. Peptides looping out at the mid- and C- terminus when bound to MHC class I: We have demonstrated that H-2Kb molecules can present the 9-mer peptide SAPDTRPAP (MUC19Kb) which does not contain the appropriate anchors [8, 9, 23] and binds with very low affinity. Furthermore, the peptide APGSTAPPA (MUCl-9Db) was found to be presented by H-2Db. This peptide also does not contain the canonical anchor of Asn-P5. MUC1 VNTR peptides (MUCl-9Kb and MUCl-9Db) have other unusual features exemplified by antibody binding and modelling studies. It was previously demonstrated that MUCl-9Kb could loop out sufficiently at the mid and C-terminus to be detected by anti-MUCl peptide monoclonal antibodies when bound to H-2Kb [23]. The same antibodies to the mid and C terminus were also able to block CTL killing. In addition, monoclonal antibodies to the mid-region of APGSTAPPA peptide presented by H-2Db were able to bind to the peptide whilst bound to the groove. Molecular modelling demonstrated that the mid (in H-2Db) and C- (in H-2Kb) terminus of the peptide looped out and above the peptide binding groove (Figure 2). This is the first demonstration of a peptide bound to MHC class I, with the mid and C-terminus looping out of the peptide binding groove. A structure would give further new insights of peptides binding to MHC class I molecules.
262
V. Apostolopoulos and M. Plcbanski / Unusual Interactions
Figure 2. Molecular model of high affinity canonical anchor motif peptide (SEV9, black) and low affinity non-canonical motif peptide (MUC1-9, grey) when in complex with MHC class I, H-2Kb
Binding of glycopeptides to MHC class I: The H-2Kb presented peptide, VSV8, with P6 linked to galabiose disaccharide is able to activate T cells and the crystal structure of H2Kb-VSV8 glycopeptide has been determined [24]. The crystal structure of a synthetic OGlcNAc bearing peptide in complex with MHC class I H-2Db was also determined at the same time. In both these structures the sugar points towards the TCR. MUC1 is highly glycosylated and, as result of aberrant glycosylation during cancer, MUC1 exposes certain carbohydrate, peptide and possibly glycopeptides that are not normally exposed on the normal mucin. In addition, due to the overexpression of MUC1 on cancer cells as compared to the normal cells, it is possible that such glycopeptides are presented by MHC class I or class II molecules for recognition by T cells. A number of studies have demonstrated potential glycosylation sites within the MUC1 VNTR region, reviewd in, [13] and studies are being pursued to identify specific glycopeptides presented by MHC class I or class II molecules. The identification of glycopeptides capable of stimulating an immune response, may be of potential value for immunotherapy studies in cancer and in human vaccination protocols [25]. Peptide mimetics for cancer The concept of molecular mimicry has been popular for many years, particularly in autoimmune disease where there is good evidence that mimicking epitopes present on viruses or bacteria to those on normal human tissue, give rise to autoimmune disease. Such concepts have been examined through the years and more recently the evidence is stronger in such diseases as multiple sclerosis, diabetes, rheumatoid arthritis and others. Mimics of
V. Apostolopoulos and M. Plebanski / Unusual Interactions
263
carbohydrates by proteins have also been described, wherein the shape of the protein and carbohydrate is sufficiently similar for them to be recognised by the Abs or CTLs. While molecular mimicry has mostly been discussed with regard to autoimmunity, there are now examples of such mimics occurring in cancer. Immunising humans (or mice) with an unrelated mimic peptide may have a therapeutic benefit to humans. A key question is whether a mimic, being different from the self molecule, will be more immunogenic and therefore more potent, but more likely to give rise to autoimmunity. The real answer to this, is by appropriate clinical trials and it would be of interest to see if mimics are more potent than attempting to induce immune responses to self cancer antigens. Peptide mimetics to mouse lymphoma and human melanoma: The identification of CTL epitopes of tumour antigens is a crucial first step for active tumour immunotherapy. One strategy is to use defined amino acids in two MHC anchor positions to search for epitopes that are recognized by CTL. One study used H2Db and H2K CTL for the mouse lymphoma EL4. Peptide libraries were screened and individual peptides were tested in an RMA-S cell assay. One H2Db and one H2Kb peptide mimics were deduced from the original library that contained more than 2xl0 8 potential peptides. Mice immunised with the synthetic peptide mimic induced specific EL4 CTL as well as inhibiting established EL4 tumours in mice [26]. In another study, an amino acid substitution in the MART-l(27–35) (AAGIGELTV) sequence led to related sequences which could sensitise cells for anti-MART-1 CTL lysis; no tumour studies were performed [27]. These studies mainly involved conventional amino acid substitutions in the basic framework of a peptide epitope. We have demonstrated one of the first examples of a completely different and unrelated peptide that elicits specific CTL and tumour protection to a cancer antigen. These methods represent an approach of identifying MHC class I epitopes and epitope mimics that can serve as immunogens for tumour immunotherapeutic vaccines. Peptide mimetic to human adenocarcinoma (MUC1 antigen): A peptide (DAHWESWL), isolated from a phage peptide library, mimics Gala(l,3)Gal expressed on pig endothelial cells [28]. In addition, DAHWESWL peptide also mimicked MUC1 peptides [29, 30], in particular, SAPDTRPAP within the MUC1 VNTR region (WESW mimicked PDTR). MUC1, a glycoprotein overexpressed on adenocarcinomas. However, could DAHWESWL mimic MUC1 peptides in association with MHC Class I molecules for recognition by T cells?. T cells recognise peptides presented in association with MHC-Class I molecules. To find a peptide mimic of MUC1, the mimic must be presented by the appropriate MHC molecule and the amino acids recognised by the T cell receptor must mimic that of the original peptide. DBA/2 (H2d) mice were injected with DAHWESWL peptide coupled to KLH. Mice generated Abs, and CTL responses to DAHWESWL peptide which also recognised MUC1 peptides. Mice were also protected against a MUC1+ tumour challenge [29]. C57BL/6 (H2b) and HLA-A*0201 mice could not generate CTLs. Using overlapping 9mer peptides spanning the 20 amino acid VNTR region of MUC1, the MUC1 epitope which was recognized by the anti-DAHWESWL CTLs was determined, which were SAPDTRPAP (H2Dd) and APDTRPAPG (H2Ld) from the MUC1 VNTR [29]. Using molecular modeling it was demonstrated that R5/R6 in SAPDTRPAP and APDTRPAPG and E5 in DAHWESWL projected downwards to the base of the MHC cleft; D4T5 from SAPDTRPAP, T4P6 from APDTRPAPG and W4 are located in the gap of the TCR. At the C-terminus of the peptide, A7P8 of APDTRPAPG and W7 of DAHWESWL also occupied similar space [29]. Although the amino acid composition of the peptide varied at the interface of the TCR, the shape and volume occupied by the mimic and MUC1 peptides is similar. Because of the lack of CTLs in HLA-A2 transgenic mice, the mimic DAHWESWL
264
V. Apostolopoulos and M. Plebanski / Unusual Interactions
peptide was mutated to a HLA-A2 compatible peptide (DLHWASWV) and CTLs were now induced [29]. Altered peptide ligands Recent evidence suggests that T cell epitopes with unusual properties can be maintained by populations of infectious pathogens to actively turn off protective T cell responses. The clearest example is malaria. Plasmodium falciparum malaria is a major cause of death in tropical regions [31]. Interferon gamma has emerged as one of the key mediators of protection [32]. Variability in specific parasite T cell epitopes can suppress IFNY secretion and cytotoxic activity by altered peptide ligand (APL) antagonism [33, 34]. Moreover, our recent findings suggest that the malaria parasite may actively switch Trl cells from IFNy to IL10 production by APL antagonism [34]. Trl cells are found frequently in malaria exposed individuals [35]. Thl cell secretion of pro-inflammatory lymphokines (IFNy and TNFa) can mediate malaria liver stage protection and early bloodstage immunity by nitrate based mechanisms, whereas Th2 and anti-inflammatory responses may promote antibody mediated protection and benefit the host by dampening potentially damaging TNFa responses [32, 36]. Interleukin 10 is a pleiotropic lymphokine which can directly or indirectly (via inhibition of accessory cell function) suppress T cell proliferation and interferon gamma (IFNY), tumour necrosis factor alpha (TNFa), granulocyte and monocyte colony stimulating factor (GM-CSF) and lymphotoxin (LT) production by T cells [37, 38]. Thus, although it can suppress proliferation of T cell in general, including T helper type 2 (Th2: cells secreting lymphokines like IL4, IL5, DL10 and IL13), it is mostly thought of as a specific inhibitor of T helper type 1 responses (Thl: cells secreting lymphokines like IL2. IFNY and TNFa) [39]. Interferon gamma, by contrast, up-regulates cellular immunity by a variety of mechanisms, for example by increasing the cytotoxic activity of macrophages, natural killer (NK) cells and T cells, the antigen presenting potential of macrophages and dendritic cells, the expression of MHC class I and II and adhesion molecules on somatic cells and by stimulating the production of other activating soluble pro-inflammatory factors such as TNFa [32, 39, 40]. Although it is now accepted that a spectrum of T cells with different lymphokine patterns may exist outside Thl/Th2, the existence of T cell clones capable of specifically secreting both IL10 and IFNY, which we will denominate T regulatory cells typel (Trl), is surprising. These cells are distinct from Th0, since they fail to secrete other Th2 type lymphokines such as IL4 [34, 35, 38, 41]. The fact that these cells can switch from IFNy to IL10 production suggests a potential for self-regulation [34]. This would radically alter current understanding of immune regulation as being mediated by distinct T cell types, since a cell of the same antigen specificity could perform a variety of opposed effector functions during its lifetime. It will be therefore important to characterise this novel T cell subset, both in homeostasis and in disease. Crystal structure ofMHC-agonist/antagonist peptide-TCR: We have determined the crystal structure of superagonist, agonist and antagonist peptides in complex with MHC and TCR. From these studies, it was evident that peptides which are involved in the trimolecular complex, (MHC-peptide-TCR), and cause an antagonistic effect (ie. loss of T cell activation) have a loss of hydrogen bond contacts of peptide side chains to the CDR3 loops of the TCR [42]. In such an event this loss of hydrogen bond contact causes an agonist peptide to become an antagonist peptide [42]. Mutation of large side chains of the peptide which interact with the TCR to small side chain amino acids (such as, Ala. Ser or Gly) can
V. Apostolopoulos and M. Plebanski / Unusual Interactions
265
cause antagonism, and thus, inhibit disease. These findings are important in designing antagonistic effects of an agonist peptide in infectious diseases, cancer and in autoimmunity. Future Prospects MHC on the surface of APCs to T cell receptors presents peptides. Recognition of peptide-MHC by T cells initiates a cascade of signals in T cells which maintains a T cell dependent immune response. An understanding of the how peptides bind to MHC class I molecules is important to determine the physiology of immune responses and how to manipulate peptides to affect an immune response. Thus far, only the crystal structures of high affinity binding peptides to MHC have been determined. We have determined structurally, features of non-canonical anchor motif peptides, one which binds with low affinity and the other with high affinity to MHC class I. Further structural studies in determining how long peptides (14-mer), short peptides (5-mer), glycopeptides and altered peptide ligands bind to the MHC and hence to the TCR are fundamental in defining additional immunogenic peptides from primary protein sequences and aid in the design of alternative approaches for vaccirtes. Acknowledgments This work was supported in part by, NH&MRC CJ Martin Fellowship, New Idea Breast Cancer Foundation and Hellenic Funds (V.A.), Howard Hughes Fellowship (M.P.) and The Austin Research Institute (V.A. and M.P.). References [I] Zinkernagel, RM and Doherty, PC Nature 248 (1974) 701. [2] Germain, RN Cell 1994; 76: 287–299,Townsend, AR, Gotch, FM and Davey, J Cell 42 (1985) 457. [3] Zinkernagel, RM and Doherty, PC Adv Immunol 27 (1979) 51. [4] Falk, K, Rotzschke, O, Stevanovic, S, Jung, G and Rammensee, HG Nature 351 (1991) 290. [5] Saper, MA, Bjorkman, PJ and Wiley, DC J Mol Biol 219 (1991) 277. [6] Rammensee, HG, Friede, T and Stevanoviic, S Immunogenetics 41 (1995) 178. [7] Mandelboim, O, Bar-Haim, E, Vadai, E, Fridkin, M and Eisenbach, L J Immunol 159 (1997) 6030, Gao, L, Walter, J, Travers, P, Stauss, H and Chain, BM J Immunol 155 (1995) 5519. [8] Apostolopoulos, V, Karanikas, V, Haurum, JS and McKenzie, IF J Immunol 159 (1997) 5211. [9] Apostolopoulos, V, Haurum, JS and McKenzie, IF Eur J Immunol 27 (1997) 2579. [10] Sette, A, Vitiello, A, Reherman, B, Fowler, P, Nayersina, R, Kast, WM, Melief, CJ, Oseroff, C, Yuan, L, Ruppert, J and et al. J Immunol 153 (1994) 5586. [ I I ] Stryhn, A, Pedersen, LO, Holm, A and Buus, S Eur J Immunol 30 (2000) 3089. [12] Tirosh, B, el-Shami, K, Vaisman, N, Carmon, L, Bar-Haim, E, Vadai, E, Feldman, M, Fridkin, M and Eisenbach, L Immunol Lett 70(1999) 21. [13] Apostolopoulos, V, McKenzie, IF and Wilson, IA Front Biosci 6 (2001) D1311. [14] Apostolopoulos, V, Yu, M, McKenzie, IF and Wilson, IA Curr Opin Mol Ther 2 (2000) 29. [15] Fremont, DH, Stura, EA, Matsumura, M, Peterson, PA and Wilson, IA Proc Natl Acad Sci U S A 92 (1995)2479. [16] Matsumura, M, Fremont, DH, Peterson, PA and Wilson, IA Science 257 (1992) 927, Fremont, DH, Matsumura, M. Stura, EA, Peterson, PA and Wilson, IA Science 257 (1992) 919. [17] Horig, H. Young, AC, Papadopoulos, NJ, DiLorenzo, TP and Nathenson, SG J Immunol 163 (1999) 4434. [18] Collins, EJ, Garboczi, DN and Wiley, DC Nature 371 (1994) 626 [ 19] Matsumura. M, Saito, Y, Jackson, MR, Song, ES and Peterson. PA J Biol Chem 267 (1992) 23589.
266
V. Apostolopoulos and M. Plebanski / Unusual Interactions
[20] Olsen, AC, Pedersen, LO, Hansen, AS, Nissen, MH, Olsen, M, Hansen, PR, Holm, A and Buus, S Ear J Immunol 24 (1994) 3K5. [21] Apostolopoulos, V, Pietersz, GA, Loveland, BE, Sandrin, MS and McKenzie, IF Proc Nail Acad Sci U S A 1995; 92: 10128-10132,Apostolopoulos, V, Pietersz, GA and McKenzie, IF Vaccine 14 (1996) 930. [22] Reddehase, MJ, Rothbard, JB and Koszinowski, UH Nature 337 (1989) 651, Eisen, HN, Sykulev, Y and Tsomides, TJ Adv Protein Chem 1996; 49: l-56,Gillanders, WE. Hanson. HL, Rubocki, RJ, Hansen. TH and Connolly, JM Int Immunol 9 (1997) 81. [23] Apostolopoulos, V, Chelvanayagam, G, Xing, PX and McKenzie. IF J Immunol 161 (1998) 767. [24] Speir, JA, Abdel-Motal, UM, Jondal, M and Wilson, IA Immunity 10 (1999) 51. [25]Muller, S, Goletz, S, Packer, N, Gooley, A, Lawson, AM and Hanisch, FG J Biol Chem 272 (1997) 24780. [26] Blake, J, Johnston, JV, Hellstrom, KE, Marquardt, H and Chen, L J Exp Med 184 (1996) 121. [27] Loftus, DJ, Castelli, C, Clay, TM, Squarcina, P, Marincola, FM, Nishimura, MI, Parmiani, G, Appella. E and Rivoltini, L J Exp Med 184 (1996) 647. [28] Vaughan, HA, Loveland, BE and Sandrin, MS Transplantation 58 (1994) 879. [29] Apostolopoulos, V, Lofthouse, SA, Popovski, V, Chelvanayagam, G, Sandrin, MS and McKenzie, IF Nat Biotechnol 16 (1998) 276. [30] Apostolopoulos, V, Osinski, C and McKenzie, IF Nat Med 4(1998) 315–320,Sandrin, MS, Vaughan, HA. Xing, PX and McKenzie, IF Glycoconj J 14 (1997) 97. [31] Snow, RW, Craig, M, Deichmann, U and Marsh, K Bull World Health Organ 77 (1999) 624. [32] Good, MF and Dooian, DL Curr Opin Immunol 11 (1999) 412. [33] Gilbert, SC, Plebanski, M, Gupta, S, Morris, J, Cox, M, Aidoo, M, Kwiatkowski, D, Greenwood, BM, Whittle, HC and Hill, AV Science 279 (1998) 1173,Plebanski, M, Lee, EA and Hill, AV Parasitology 115 (1997) S55,Plebanski, M, Lee, EA, Hannan, CM, Flanagan, KL, Gilbert, SC, Gravenor, MB and Hill. AV Nat Med 5 (1999) 565. [34] Plebanski, M, Flanagan, KL, Lee, EA, Reece, WH, Hart, K, Gelder. C. Gillespie, G, Pinder, M and Hill, AV Immunity 10 (1999) 651. [35] Winkler, S, Willheim, M, Baier, K, Schmid, D, Aichelburg, A, Graninger, W and Kremsner, PG Journal of Infectious Diseases 179 (1999) 209. [36] Stevenson, MM and Tarn, MF Clin Exp Immunol 92 (1993) 77, Taylor-Robinson, AW, Phillips. RS, Severn, A, Moncada, S and Liew, FY Science 260 (1993) 1931, von der Weid, T and Langhorne, J Immunobiology 189 (1993) 397, Kwiatkowski, D, Bate, CA, Scragg, IG, Beattie, P, Udalova, I and Knight, JC Ann Trap Med Parasitol 91 (1997) 533. [37] Cobbold, S and Waldmann, H Curr. Opin. Immunol. 1998; 10: 518–524,Moore, KW, O'Garra, A, de Waal Malefyt, R, Vieira, P and Mosmann, TR Annu Rev Immunol 11 (1993) 165. [38] Groux, H and Powrie, F Immunol Today 20 (1999) 442. [39] Mosmann, TR and Moore, KW Immunol Today 12 (1991) A49. [40] Boehm, U, Klamp, T, Groot, M and Howard, JC Annu Rev Immunol 15 (1997) 749. [41] Allen, JE and Maizels, RM Immunol Today 18 (1997) 387,Groux, H, O'Garra, A, Bigler, M, Rouleau, M, Antonenko, S, de Vries, JE and Roncarolo, MG Nature 389 (1997) 737, Asseman, C and Powrie, F Gut 42 (1998)157. [42] Degano, M, Garcia, KC, Apostolopoulos, V, Rudolph, MG, Teyton, L and Wilson, IA Immunity 12 (2000)251.
Drug Discovery and Design: Medical Aspects J. Matsoukasand T. Mavromoustakos (Eds.) 1OS Press, 2002
267
Effects of Angiotensin II, III and IV on Memory Retention of Rats: Interaction with Adenosine AI Receptor Related Ligands J. Tchekalarova and V. Georgiev Laboratory "Experimental Psychopharmacology", Institute of Physiology, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria Abstract. Angiotensins II, III and IV (Ang II, Ang III and Ang IV) exert a crucial role in learning and memory processes either alone or interacting with other neurotransmitters and neuromodulators. The present study examines the functional interaction between angiotensin II, III, IV and adenosine AI receptorrelated drugs on passive avoidance (step-through) conditioning in rats. Ang II and III facilitated while the heptapeptide decreased the retention response. Adenosine A, receptors could modulate the realization of angiotensins memory effects. Adenosine AI and Ang II receptors interact in adaptation to long-term theophylline administration in step-through memory task. Taken together, the obtained data suggest that angiotensin peptides are closely implicated in the regulation of memory retention processes and mutual interaction with adenosine A, receptors could play an important role in its realization.
Introduction Increasing evidence suggests that biologically active forms of angiotensins, II, in and IV (Ang II, Ang III and Ang IV) including their specific binding sites are present in the central nervous system (CNS). The accumulated data indicate that the angiotensin peptides play major role in the central neural excitability (memory processes included) either alone or in close relationship with certain neurotransmitters and neuromodulators. The effects of angiotensin ligands (Ang II, Ang III, Ang IV) are mediated through their interaction with specific angiotensin receptors. Due to the development of selective peptidic or nonpeptidic Ang II ligands several subtypes of angiotensin receptors have been identified. The AT] subtype binds with high affinity the imidazolic nonpeptidic compound DuP 753 or losartan but not the spinacme derivative PD 123319 or the pseudopeptidic Ang II analog CGP 42
268
J. Tchekalarova and V. Georgiev I Effects of Angiotensin II, III and IV
112 A. AT| receptor is a seven-transmembrane and G-protein-coupled receptor. It is present in high densities in areas closely related with the "classic functions" of the brain RAS (Renin Angiotensin System) including regulation of body fluid homeostasis, heart rate and blood pressure and control of vasopressin release. The AT2 subtype binds with high affinity to PD 123319 and CGP 42 112 A but not to DuP 753. AT2 is also a seven-transmembrane domain and G-protein coupled receptor. It is present in brain areas rather different of those for ATi site. The AT2 subtype is supposed to play a role in vascular growth, the autoregulation of cerebral blood flow, cGMP regulation and modification of signalling of other receptors. The AT4 subtype is distinct from the AT| and AT2 sites. It has been determined that [I25I] Ang IV binds to the AT4 site reversibly, saturably, and with high affinity. This subtype is not G-protein linked. The AT4 site is expressed in high density predominately in brain areas involved in the enhancement of memory acquisition and retrieval, regulation of blood flow, angiogenesis, etc. Divalinal- Ang IV is a specific antagonist of AT4 receptor [ 1 ]. The observed effects of ANG n on cognitive processes have been inconsistent. Thus, ANG n, infused into the dorsal striatum after passive avoidance conditioning, disrupted the retention 24 h later [2]. On the other hand, we have demonstrated that Ang n exerted retention-improving effect in shuttle-box active avoidance and facilitated retention in step-trough passive avoidance task [3, 4]. Angiotensin ATi receptor subtype has been implicated in the mediation of the octapeptide's Ang II effects on cognition. Braszko et al. first showed that i.c.v. infused Ang IV, in the equivalent dose of 1 nmol with Ang II, improved recall of passive avoidance conditioning and acquisition of active avoidance conditioning [5]. Wright et al. extended these finding revealing that ANG IV effect on passive avoidance response is dose-dependent and the selective AT4 receptor antagonist Divalinal-Ang IV (10 nmol), severely disrupted recall of this memory effect [6]. Furthermore, metabolically resistant analogs of Ang IV facilitated acquisition and retention in a circular water maze task in scopolamine treated rats, perforant path damaged rats and ischemia-induced hippocampal damaged gerbils [7]. Adenosine AI receptors play a significant role in the cognition. Thus, the selective agonists for AI receptor as N^-cyclopentyladenosine (CPA) and N^-U-methyl-2phenyl)adenosine (R-PIA) have been shown to impair the retention [8, 9] while the AI receptor antagonists as theophylline produce memory enhancement in passive avoidance tests [9]. There are several reports that have considered adenosinergic and angiotensinergic interactions in the periphery and the central nervous system (CNS). Thus, ANG n leads to a release of adenosine from the rat lung [10] and to an increase in the plasma adenosine concentration in renovascular hypertensive rats [11]. In the CNS, endogenous angiotensin modulates responses on blood pressure and heart rate to adenosine when microinjected into the area postrema of rats [12]. In our previous studies, we suggested that angiotensins n, in and IV as well as adenosine AI receptors could interact in the regulation of the seizure susceptibility [13, 14, 15]. The process of long-term potentiation (LTP) in hippocampus represents a basic physiological mechanism of memory storage in the brain. Accumulating data reveal that angiotensins play a major role in the excitability of hippocampus, including LTP. Wayner et al. has shown that Ang II microinjected in the hippocampus inhibited LTP in perforant path-stimulated dentate granule cells [16]. This Ang H-induced inhibition could be blocked with saralasin or DuP 753 but not by PD 123319. Ang III was found to be 40 to 50-fold less potent than Ang II at inhibition of LTP [16]. Further, AT4 receptor stimulation via native Ang IV or its more selective analog Nle'-Ang IV, enhanced synaptic transmission and LTP. while infusion of the AT4 receptor antagonist, Nle'-Llual^-Ang IV, disrupted stabilization of LTP [17]. These results are in agreement with the above mentioned behavioral work
J. Tchekalarova and V. Georgiev /Effects
of Angiotensin II, III and IV
269
indicating that AT| receptors mediated inhibition while AT4 receptors activation enhance the cognition in different memory paradigms. The present review aims to summarize the post-trial effects of bilogically active forms of angiotensin II, III and IV as well as of their interaction with adenosine AI receptor-related drugs on inhibitory avoidance (passive avoidance) task in rats. It is beyond the mission of this review to present detailed treatments of this topic, but rather to summarize the major findings of the pertinent literature and particularly those from our laboratory. Ang II-, HI-, IV- and adenosine Aj- receptor interactions: There is increasing evidence of interactions between Ang n and adenosine system in the CNS. Previously, in our laboratory we have found agonistic angiotensin-adenosine relationships in seizure susceptibility [13, 14, 15, 18]. Further, we extended this study on cognition processes, revealing antagonistic angiotensin-adenosine AI receptor-mediated interactions. For the purpose, we used steptrough method with punishment reinforcement where the rats were trained on a one-trial passive avoidance task. Retention was assessed 24 hours and 7 days later, using a procedure identical to that for training, except that footshock was not delivered. The latency to cross from the outer to the inner compartment with the four legs was recorded. Retentionenhancing effect exerted by Ang II was abolished by adenosine AI receptor agonist N6cyclohexyladenosine (CHA) upon re-testing of rats 24 but not 7 days after passive avoidance training (Table 1) [19]. Table 1. Latencies for the rats to enter the dark compartment in passive avoidance task measured 24 h and 7 days after training. The drugs were injected i.c.v. or i.p. immediately after training trial. n=10 animal/group; *p<0.05, ** p<0.01 vs. controls; °p<0.05, °°p<0.01 vs. angiotensin II, III or IV, respectively; +p<0.05, ++p<0.01 vs. CHA or theophylline, respectively.
Ang II 0. 1 u,g CHA 0.1 u.g Ang 0+CHA theoph.75mg/kg AngllO.l+theoph Ang III 0.1 ng
24th h Latency (%) 44% 70% 15% 40% 44% 30% 16%
Ang III+CHA
60%
Anglll+theoph Ang IV 0.1 [ig Ang IV+CPA AnglVl+theoph
2% 70% 34.3 % 72%
Drugs Controls
%2 3.8* 2.39* 7.2°° 3.83* 3.4* 4.34°° 3.83++ 3.86+ 5.79* 3.35° 6.99+0
7th day Latency (%)
30% 23% 22% 29% 20% 4%
*2 6.39**
26%
3.2*
2% 67% 42% 20%
4.5++ 4.78* 2.39°
Theophylline is a non-selective A]/A2 adenosine receptor antagonist that is known to improve cognitive performance. Prolonged exposure of adenosine AI receptors to their ligands lead to adaptive changes. Thus, chronic treatment with either adenosine agonist or antagonist results in cognitive responses that differ from those observed following acute administration of these compounds. Recently, we have demonstrated that the increasing effect of adenosine AI receptor antagonist 8-p-sulfophenyl-theophylline (8-p-SPT) on retention was potentiated by Ang II after chronic (14 days) systemic treatment of rats with
J. Tchekalarova and V. Georgiev /Effects
270
ofAngiotensin II, III and IV
theophylline (Table 2) [Tchekalarova, Georgiev, unpublished results]. On the contrary, the non-specific angiotensin receptor antagonist, sarilesin, showed opposite, memoryimproving effect, on theophylline-treated animals compared with the acute experiment where it demonstrated amnesic effect in passive avoidance task (Table 2). Table 2. Latencies for the rats to enter the dark compartment in passive avoidance task measured 24 h after training. Chronic theophylline was administered 14 days in dose of 75 mg/kg, i.p. The drugs were injected i.c.v. immediately after training trial. n=10 animal/group; *p<0.05 vs. respective controls.
Drugs controls 1 controls 2 (chronic theophylline) 8-p-SPT 50 ng (acute) 8-p-SPT 50 jig (chronic theophylline) sarilesin 1 pig (acute) sarilesin 1 |Xg (chronic theophylline)
24*11 Latency (%) 37%
X2
29%
-
46%
-
82%
3.8*
5%
4.89*
60%
-
The heptapeptide Ang HI, which impaired long-term memory upon re-testing of rats 24 h and 7 days later in step-through task, produced an opposite, enhancing effect on the retention when was co-administered with CHA (Table 1) [19]. Furthermore, Ang ID attenuated the memory-improving effect induced by theophylline after re-testing the rats 24 h and 7 days later. We confirmed and extended the findings reported by Braszko et al. and Wright et al. [5, 61] that when Ang IV is injected i.c.v., improved the recall of a passive avoidance response. Thus, this facilitatory effect of the hexapeptide was reduced by adenosine AI receptor agonist N6-cyclopentyladenosine (CPA) while combination of theophylline and Ang IV, both injected in ineffective doses, facilitated the retention 24 hours after training but not 7 days later (Table 1) [20]. Conclusions The present results demonstrate that Ang n and Ang ID produce opposite effects in passive avoidance conditioning as the octapeptide facilitated while the heptapeptide decreased the retention response. Furthermore, adenosine AI receptors could modulate the realization of angiotensin's memory effects. In addition with the proposed close relationships between Ang H/Ang HI and adenosine-related drugs, we have shown that the enhancing effect of Ang IV upon passive avoidance conditioning might be due to the concurrent interaction of AT4 and adenosine AI receptors. So far, most of the effects of angiotensins on cognition seem to be mediated by AT| or AT4 receptor subtypes. The role of AT2 receptor subtype is less clear although there are few studies suggesting that this receptor might mediate the cognitive enhancing action of Ang n [7]. Ang II has been recognized as the major peptide activating ATi receptors in the periphery while Ang HI could be the predominant effector peptide in the brain [7]. It is tempting to suggest that Ang II enhancing effects on cognition could be mediated by AT4 receptors. In this respect, AT4 receptors have to be expected to being excited indirectly by Ang II through its degradation to Ang IV. This point of view is supported by the observed failure of AT| and AT2 receptor antagonists to block subsequent
J. Tchekalarova and V. Georgiev / Effects of Angiotensin II, HI and IV
271
Ang II facilitation of acquisition of a conditioned response. The latter was explained by Wright et al. [7] with degradation of Ang II to Ang III, then to Ang IV, the ligand that by acting on AT4 receptors, improves the performance. On the other hand, the observed Ang HI decreasing effect on memory retention might be realized through interaction with ATi receptors. The intriguing finding of the present study is based upon the observation that the selective adenosine AI receptor agonist CHA modulates the memory effects of Ang II, III and IV. Furthermore, Ang III was able to convert memory-enhancing effect produced by theophylline not only 24 h but also 7 days after the trial test which fits well with the longterm memory effects observed by this heptapeptide in the present study. The persistence of increased retention in Ang IV-treated mice 7 days after training might be explained with stimulation of the protein synthesis thus influencing late consolidation and long-term memory storage [21, 22]. On the other hand, long-lasting action of sarilesin seems to be a sequence of its resistance to peptidases allowing it to remain in the vicinity of the receptors as proactive material for a longer period. The exact mechanism of Ang's n, HI, IV and adenosine AI receptor-related drugs relationships in cognition processes is not quite clear and needs further studies for its elucidation. Acknowledgements This work was supported by European Commission through the COPERNICUS programme, contract No. CIPA CT 94-0239, and by Grant VFP-L-6 from the National Fund for Scientific Research at the Bulgarian Ministry of Education and Science, Sofia. References [I] M. De Gaspare, K.J. Catt, T. Inagami, J.W. Wright J.W., T. Unger, Pharmacol. Rev. 52 (2000) 415. [2] M.R. Zarrindast, B. Shafahi, Eur. J. Pharmacol. 256 (1994) 233. [3] V. Georgiev, D. Yonkov, Meth. Find. Exptl. Clin. Pharmacol. 7 (1985) 415. [4] V. Georgiev, D. Yonkov, T. Kambourova, Neuropeptides 12 (1988) 155. [5] J.J. Braszko, B. Kupryszewski, B. Witczuk, K. Wisniewski, Neuroscience 27 (1988) 777. [6] J.W. Wright, L.T. Krebs, J.W. Stobb, J.W. Harding, Front. Neuroendocrinol. 16 (1995) 23. [7] J.W. Wright, J.W. Harding, Brain Res. Rev. 25 (1997) 96. [8] H.L. Normile, R.A. Barraco, Brain Res. Bull. 27 (1991) 101. [9] D.K.J.E Von Lubitz, K.A. Jacobson, Behavioral effects of adenosine receptor stimulation. In " Adenosine and Adenine Nucleotides: from Molecular Biology to Integrative Physiology", Edited by L.Belardinelli and A. Pelleg, Kluver Academic Publs., Boston, Dordrecht, London, 1995, pp. 489–498 [10] D.F. Bottiglieri, K. Curtis, K.E. Jackson, J. Cardiovasc. Pharmacol. 16 (1990) 101. II1] A. Ohnishi, P. Li, A.R. Brauch, O.I. Biaggioni, K.E. Jackson, Hypertension 12 (1988) 152. [12] H.C. Lin, F.J. Wan, C. Tung, C.J. Tseng, J. Auton. Nerv. Syst. 51 (1995) 19. [13] V. Georgiev, J. TchekalarovaBrain Res. 779 (1998) 259. [14] J. Tchekalarova, V. Georgiev, J. Physiol. (Paris) 93 (1999) 191. [15] J. Tchekalarova, T. Kambourova, V. Georgiev, Brain Res. Bull. 52 (2000) 13. [16] M.J. Wayner, D.L. Armstrong, J.L. Polan-Curtain, J.B. Denny, Pharmacol. Biochem. Behav. 45 (1993) 455. [17] E. Kramar, D. Armstrong, S. Ikeda, M. Wayner, J. Harding, J. Wright, Brain Res. 897 (2001) 114. [18] J. Tchekalarova, T. Kambourova, V. Georgiev, Brain Res. Bull, (in press) [19] J. Tchekalarova, T. Kambourova, V. GeorgievBehav. Brain Res. (in press) [20] J. Tchekalarova, T. Kambourova, V. Georgiev, Behav. Brain. Res. 123 (2001) 113. [21] T. Herdegen. J.D. Leah, Brain Res. Bull. 28 (1998) 370. [22] K.A. Robert, L.T Krebs, E.A. Kramar, M.J. Shaffer, J.W. Harding, J.W. Wright, Brain Res. 682 (1995) 13.
272
Drug Discovery and Design: Medical Aspects J. Matsoukasand T. Mavromoustakos (Eds.) IOS Press, 2002
Molecular Targets and Compounds for AntiHIV Therapy E. De Clercq Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium Abstract. Virtually all the compounds that are currently used, or under advanced clinical trial, for the treatment of HIV infections, belong to one of the following classes: (1) nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs): i.e., zidovudine (AZT), didanosine (ddl), zalcitabine (ddC), stavudine (d4T), lamivudine (3TC), abacavir (ABC), emtricitabine [(-)FTC], tenofovir (PMPA) disoproxil fumarate; (ii) non-nucleoside reverse transcriptase inhibitors (NNRTIs): i.e., nevirapine, delavirdme, efavirenz, emivirine (MKC-442); and (iii) protease inhibitors (Pls): i.e., saquinavir, ritonavir, indinavir, nelfinavir, amprenavir, and lopinavir. In addition to the reverse transcriptase and protease step, various other events in the HIV replicative cycle are potential targets for chemotherapeutic intervention: (i) viral adsorption, through binding to the viral envelope glycoprotein gp120 (polysulfates, polysulfonates, polyoxometalates, zintevir, negatively charged albumins, cosalane analogues); (ii) viral entry, through blockade of the viral coreceptors CXCR4 and CCR5 [bicyclams (i.e. AMD3100), polyphemusins (T22), TAK-779, MlP-lct LD780 isoform]; (iii) virus-cell fusion, through binding to the viral glycoprotein gp41 (T-20 (DP-178), T-1249 (DP-107), siamycins, betulinic acid derivatives]; (iv) viral assembly and disassembly, through NCp7 zinc finger-targeted agents [2,2'-dithiobisbenzamides (DIBAs), azadicarbonamide (ADA) and NCp7 peptide mimics]; (v) proviral DNA integration, through integrase inhibitors such as L-chicoric acid and diketo acids (i.e. L-731,988); (yj) viral mRNA transcription, through inhibitors of the transcription (transactivation) process (fluoroquinolone K-12, Streptomyces product EM2487, temacrazine, CGP64222). Also, in recent years new NRTIs, NNRTIs and Pis have been developed that possess respectively improved metabolic characteristics (i.e. phosphoramidate and cyc/osaligenyl pronucleotides of d4T), or increased activity against NNRTI-resistant HIV strains [second generation NNRTIs, such as capravirine and the novel quinoxaline, quinazolinone, phenylethylthiazoly-lthiourea (PETT) and emivirine (MKC-442) analogues], or, as in the case of Pis, a different, non-peptidic scaffold [i.e. cyclic urea (DMP 450), 4-hydroxy-2-pyrone (tipranavir)]. Given the multitude of molecular targets with which anti-HIV agents can interact, one should be cautious in extrapolating from cell-free enzymatic assays to the mode of action of these agents in intact cells. A number of compounds (i.e. zintevir and L-chicoric acid, on the one hand; and CGP64222 on the other hand) have recently been found to interact with virus-cell binding and viral entry in contrast to their proposed modes of action targeted at the integrase and transactivation process, respectively.
E. De Clercq /Molecular Targets and Compounds for Anti-HIV Therapy
273
Introduction Combination therapy, comprising at least three anti-HIV drugs, has become the standard treatment of AIDS or HIV-infected patients. Virtually all drugs that have been licensed for clinical use (or made available through expanded access programmes) for the treatment of HIV infections fall into one of the following three categories: (i), nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), that, following two phosphorylation steps (tenofovir) or three phosphorylation steps (zidovudine, didanosine, zalcitabine, stavudine, lamivudine, abacavir), act, as chain terminators, at the substrate binding site of the reverse transcriptase; (ii), non-nucleoside reverse transcriptase inhibitors (NNRTIs), that interact with the reverse transcriptase at an allosteric, non-substrate binding site (nevirapine, delavirdine, efavirenz); and (iii), protease inhibitors (PIs), that specifically inhibit, as peptidomimetics, the virus-associated protease (saquinavir, ritonavir, indinavir, nelfinavir, amprenavir, lopinavir). Guidelines to the major clinical trials with these compounds have been recently published [1]. In numerous studies, combinations of NRTIs, NNRTIs and PIs have been found to decrease HIV viral load, increase CD4 count, decrease mortality and delay disease progression, particularly in AIDS patients with advanced immune suppression [2]. When initiated during early asymptomatic HIV infection, highly active antiretroviral (combination) therapy (HAART) initiates rapid reversal of disease-induced T-cell activation, while preserving pretherapy levels of immune function, suggesting that therapeutic benefit may be gained from early aggressive anti-HIV chemotherapy [3]. Combination therapy that produces sustained suppression of plasma HIV RNA may also be able to reduce the virus burden in the lymphoid tissues [4], although clearance of plasma viremia is not invariably associated with immune restoration [5] and not at all paralleled by a reduction in viral DNA burden in the peripheral blood mononuclear cells [6]. The introduction of HAART or combined anti-HIV drug regimens has had profound repercussions on various AIDS-associated diseases. While partial immune restitution induced by HAART in patients with advanced HIV infection can exacerbate clinically apparent cryptococcal meningitis [7] or CMV vitritis, HAART may reduce the prevalence of cervical squamous intra-epithelial lesions in HIV-seropositive women caused by HPV [8]. Several studies have indicated that HAART significantly improves the prognosis of AIDS patients with progressive multifocal leukoencephalopathy [9–11]. Likewise, HAART has been shown to favorably alter the prognosis of CMV retinitis in HIV-infected individuals, as attested by long-lasting remission of CMV retinitis without CMV maintenance therapy [12], lack of reactivation of CMV retinitis after stopping CMV maintenance therapy [13], decrease of CMV replication (viremia) [14,15] and increased survival [16,17]. In the era of combination antiretroviral therapy the management of CMV diseases in patients with AIDS has undergone dramatic modifications [18,19]. Although the long-term goal of eradicating the virus from latently and chronically infected cells remains forbidding [20] the advent of so many new compounds, other than those that have been formally approved, for the treatment of HIV infections, will undoubtedly improve the prognosis of patients with AIDS and AIDS-associated diseases. Here I will primarily address those new anti-HIV compounds that (i) have emerged as promising anti-HIV drug candidates during the last few years, that (ii) are in preclinical or early-clinical development, and that (iji) are targeted at well-defined steps in the HIV replicative cycle. In particular, reverse trascriptase (RT) targeted at the substrate and allosteric site will be covered.
274
E. De Clercq /Molecular Targets and Compounds for Anti-HIV Therapy
Reverse transcriptase inhibitors targeted at the substrate binding site The substrate (dNTP) binding site of the HIV-1 reverse transcriptase (RT) is the target for a large variety of NRTI analogues, which have for several years [21] been recognized as efficacious agents for the treatment of HIV infections: i.e., zidovudine (AZT), didanosine (ddl), zalcitabine (ddC), stavudine (d4T), lamivudine (3TC), abacavir (ABC). Other agents in clinical trial include tenofovir (PMPA) and emtricitabine [(-)FTC]. As a rule, all of these compounds must be phosphorylated to their 5'-triphosphate form, before they can act as competitive inhibitors/substrate analogues/chain terminators at the reverse transcriptase level (Figure 1). In contrast to the nucleoside analogues, the nucleotide analogues PMEA and PMPA (Figure 2) are already equipped with a phosphonate group, and thus only need two phosphorylation steps to be converted to the active metabolite [22]. From PMEA and PMPA the oral prodrug forms [bis(POM)-PMEA or adefovir dipivoxyl, and bis(POC)-PMPA or tenofovir disoproxil fumarate, respectively] have been prepared. The former is now in phase III clinical trials for the treatment of hepatitis B virus (HBV) infections, whereas the latter has gone through several phase III clinical trials for the treatment of HIV infections [23]. Tenofovir has proven to be antivirally active in heavily antiretroviral-experienced patients; it has a favorable activity/resistance profile in that it by itself leads to little, if any, resistance, while retaining full activity against HIV-1 RT mutations that have developed resistance to 3TC (Ml84V mutants) or multinucleosids (Ql51M mutants) [24]. NH,
bis(POM)-PMEA Adefovir dipivoxU
o
II
(CH&CH—O—C—O—CH2
O.
(CHjfeCH—O—C—O—CHj
O
O
bis(POC)-PMPA Tenofovir disoproxil Figure 1. Mechanism of anti-HFV action of 2',3'-dideoxynucleoside analogues, as exemplified forzidovudine (azidothymidine, AZT, AzddThd).
E. De Clercq / Molecular Targets and Compounds for Anti-HIV Therapy
275
CH,—C—S—CHj—CH2—O' II O Bis-S-acetyl-thioethylester
H,CO—C—CH— HN O
CH,
Aryloxyphosphoramidate
d4T
Cyclo saligenyl
Figure 2. Acyclic nucleoside phosphonates: PMEA (adefovir) and PMPA (tenofovir) in their prodrug forms, adefovir dipivoxil and tenofovir disoproxil. Prodrugs of ddNMPs: Bis(S-acetyl-thioethyl)ester of ddAMP, (Aryloxy)phosphoramidate of d4T, and cycloSal-d4TMP and cycloSal-ddAMP.
In addition to 3TC and (-)FTC, the structurally related 2'-deoxy-3'-oxa-4'thiocytidine (BCH-10652, dOTC) [25], the dioxolane purine nucleoside analogues [26] and the methylenecyclopropane nucleoside analogues (and their phosphoro-L-alaninate diesters) [27,28] have recently been described as new anti-HIV agents. Emtricitabine [(-) FTC] is in phase III trials for HIV and phase I/II trials for HBV; it is considered for use in the multidrug combination therapy of HIV-1 and HBV infections [29]. DAPD, (-)-|3-D-2,6diaminopurine dioxolane, which is converted by adenosine deaminase to (-)-p-D-dioxolane guanine (DXG) (which is then further metabolized intracellularly to its active metabolite DXG-TP, a strong alternative substrate inhibitor of the HIV RT) [30], has proven active against AZT- and 3TC-resistant HIV-1 strains and has proceeded to phase I/II clinical studies [31]. BCH-10652 (dOTC) has demonstrated activity against HIV-1 in the SCID-hu Thy/Liv model; despite its structural similarity to 3TC it proved also active against 3TCresistant HIV-1 (Ml84V), albeit at a relatively high dosage level (400 mg/kg/day) [32]. All the nucleoside analogues mentioned above may ultimately, upon their intracellular metabolism, act as chain terminators, akin to AZT (Fig. 9). Chain-terminating nucleotides inhibit HIV replication because their incorporation at the 3' end of the nascent DNA chain prevents further elongation. However, it has been recently shown that under physiological conditions RT can remove these chain terminators and thus unblock the primer termini [33,34]. Mutant RTs associated with AZT resistance would be capable of removing the chain terminating residue with much greater efficiency than wild-type RT and this should, in turn, facilitate rescue of DNA synthesis. On the other hand, the Ml84V mutation, which confers high-level resistance to 3TC, would severely impair the removal of chain-terminating nucleotides and hence suppress the rescue of AZT-terminated DNA synthesis [35]. Similarly, foscarnet resistance mutations would also suppress the removal of
276
E. De Clercq / Molecular Targets and Compounds for Anti-HIV Therapy
AZT monophosphate from the blocked primer/templates, thus providing a likely explanation for the resensitization of AZT-resistant HIV-1 strains to AZT [36]. The bottleneck in the metabolic pathway leading from AZT and the other 2',3'dideoxynucleoside (ddN) analogues to their active 5'-triphosphate form is the first phosphorylation step. Therefore attempts have been made at constructing 2',3'dideoxynucleotide (ddNMP) prodrugs, that, once they have been taken up by the cells, deliver the nucleotide (ddNMP) form. This approach has proven particularly successful for a number of NRTIs such as 2',3'-dideoxyadenosine (ddA) and d4T (Fig. 2). Thus, the bis(S-acetyl-2-thioethyl)phosphotriester of ddA [bis(SATE)ddAMP] was synthesized and found to be 1000-fold more potent against HIV than the parent compound [37]. By directly delivering bis(SATE)ddAMP into the cells, one circumvents the adenosine deaminase step that would otherwise rapidly degrade ddA to ddI. Similarly, aryloxyphosphoramidate derivatives (Fig. 2) of d4T [i.e., So324, a d4TMP prodrug containing at the phosphate moiety a phenyl group and the methylester of alanine linked to the phosphate group through a phosphoramidate linkage] have been constructed [38–40]. Once taken up by the cells, d4TMP is released intracellularly (via the alaninyl d4TMP intermediate) [41]. The latter could be considered as an intracellular depot form of d4TMP [41]. This phosphoramidate prodrug approach does not seem to work so well with zidovudine, where the main metabolite formed from the alaninyl AZTMP intermediate is AZT rather than AZTMP [42] thus explaining why d4T phosphoramidate prodrugs, but not AZT phosphoramidate prodrugs, retain anti-HIV activity in HlV-infected thymidine kinase-deficient cell cultures. In resting monocytes/macrophages (M/M) the aryloxyphosphoramidate derivatives of d4T, d4A and ddA provided an anti-HIV activity that was 25- to 625-fold greater than that of the parent nucleosides (d4T, d4A and ddA) [43]. The thymidine kinase (in the case of d4T) and the adenosine deaminase (in the case of ddA) can also be bypassed by using the cyclic saligenyl approach [44,45]. CycloSaligenyl pronucleotides of d4T and ddA (Fig. 10) deliver exclusively the nucleotides d4TMP and ddAMP, not only under chemical-simulated hydrolysis conditions but also under intracellular conditions [46,47]. The cycloSal approach has also been applied, with success, to F-ara-ddA (lodenosine, 2'-fluoro-ara-2',3'-dideoxyadenosine) [48]. Conclusions In recent years, an ever increasing number of compounds have been uncovered as anti-HIV agents targeted at virtually any step of the virus replicative cycle: adsorption, entry, fusion, uncoating, reverse transcription, integration, transcription (transactivation), and maturation. In addition to the "newer" NRTIs, NNRTIs and PIs, various other compounds, i.e. those that are targeted at viral entry (i.e. CXCR4 and CCR5 antagonists) and virus-cell adsorption/fusion (i.e. compounds interacting with either gp120 or gp41), offer great potential for the treatment of HIV infections. Most of the anti-HIV agents seem to accomplish their anti-HIV activity through an interaction with the presumed molecular target. However, quite a number of compounds are capable of interacting with more than one target so their true mechanism of action may not be what was expected. Three examples in point are the G-octet containing 17-mer oligonucleotide zintevir, the dicaffeoyltartaric acid L-chicoric acid, and the nonapeptoid CGP 64222. Zintevir and Lchicoric acid were originally identified as integrase inhibitors, and the nonapeptoid as a transactivation (Tat) antagonist, and their anti-HIV activity in acutely infected cells was ascribed to interference with the integration and transactivation process, respectively. As it now appears, zintevir and L-chicoric acid primarily interact as virus adsorption inhibitors, and the nonapeptoid as a CXCR4 antagonist, and thus these compounds owe their anti-HIV
E. De Clercq / Molecular Targets and Compounds for Anti-HIV Therapy
277
activity mainly to interference with an early event (adsorption, entry) of the HIV replicative cycle. Caution should therefore be exercised in postulating from the alleged interaction of a certain anti-HIV agent with a certain viral protein (or enzyme) that the anti-HIV activity is necessarily due to this interaction [49,50]. The compound(s) may well be able to interact with other targets that are more crucial in the anti-HIV activity that is eventually achieved. Acknowledgements Prof. Erik De Clercq holds the Professor P. De Somer Chair of Microbiology at the Katholieke Universiteit Leuven School of Medicine. I thank Christiane Callebaut for her invaluable editorial assistance. References [1] J. A. Tavel, K. D. Miller, and H. Masur, Clin. Infect. Dis. 28 (1999) 643. [2] K. Henry, A. Erice, C. Tierney, H. H. Jr Balfour, M. A. Fischl, A. Kmack, S.-H. Liou, A. Kenton, M. S. Hirsch, J. Phair, A. Martinez, and J. O. Kahn, J. Acquir. Immun. Defic. Syndr. Hum, Retrovirol 19 (1998) 339. [3] L. R. Bisset, R. W. Cone, W. Huber, M. Battegay, P. L. Vernazza, R. Weber, P. J. Grob, M. Opravil, and the Swiss HIV Cohort Study, AIDS 12 (1998) 2115. [4] M. Harris, P. Patenaude, P. Cooperberg, D. Filipenko, A. Thorne, J. Raboud, S. Rae, P. Dailey, D. Chernoff, J. Todd, B. Conway, J.S.G. Montaner, and the INCAS Study Group, J. Infect. Dis. 176 (1997) 1388. [5] N. G. Pakker, E.D.M.B. Kroon, M.T.L. Roos, A. S. Otto, D. Hall, F.W.N.M. Wit, D. Hamann, M. van der Ende, F.A.P. Claessen, H. R. Kauffmann, P. K. Koopmans, F. P. Kroon, C.H.H. ten Napel, H. G. Sprenger, H. M. Weigel, J.S.G. Montaner, J.M.A. Lange, P. Reiss, P.T.A. Schellekens, and F. Miedema, AIDS 13 (1999) 203. [6] J. J. Zaunders, H. P. Cunningham, D. A. Kelleher, R. G. Kaufmann, B. A. Jaramillo, R. Wright, D. Smith, P. Grey, J. Vizzard, A. Carr, and A. D. Cooper, J. Infect. Dis. 180 (1999) 320. L. M. Woods II, R. MacGinley, P. D. Eisen, and M. A. Allworth, AIDS 12 (1998) 1491. Heard, V. Schmitz, D. Costagliola, G. Orth, and M. D. Kazatchkine, AIDS 12 (1998) 1459. H. Albrecht, C. Hoffmann, O. Degen, A. Stoehr, A. Plettenberg, T. Mertenskotter, C. Eggers, and H.J. Stellbrink, AIDS 12 (1998) 1149. [ 10] P. Miralles, J. Berenguer, D. Garcia de Viedma, B. Padilla, J. Cosin, J. C. L6pez-Bernaldo de Quiros, L. Munoz, S. Moreno, and E. Bouza, AIDS 12 (1998) 2467. [ 11] M. S. Dworkin, P.-C. T. Wan, D. L. Hanson, J. L. Jones, and the Adult and Adolescent Spectrum of HIV Disease Project, J. Infect. Dis. 180 (1999) 621. [12]C. Tural, J. Romeu, G. Sirera, D. Andreu, M. Conejero, S. Ruiz, A. Jou, A. Bonjoch, L. Ruiz, A. Arno, and B. Clotet, J. Infect. Dis. 177 (1998) 1080. [ 13] J. C. Macdonald, F. J. Torriani, L. S. Morse, M. P. Karavellas, J. B. Reed, and W. R. Freeman, J. Infect. Dis. 177 (1998) 1182. [14] C. E. O'Sullivan, W. L. Drew, D. J. McMullen, R. Miner, J. Y. Lee, R. A. Kaslow, J. G. Lazar, and M. S. Saag, J. Infect. Dis. 180 (1999) 847. [15] J. Deayton, A. Mocroft, P. Wilson, V. C. Emery, M. A. Johnson, and P. D. Griffiths, AIDS 13 (1999) 1203. [16] J. L. Casado, M. J. Perez-Elias, P. Marti-Belda, A. Antela, M.,Suarez, E. Ciancas, B. Frutos, M. D. Perez, and A. Guerrero, J. Acquir. Immun. Defic. Syndr. Hum. Retrovirol 19 (1998) 130. [17] J. C. Walsh, C. D. Jones, E. A. Barnes, B. G. Gazzard, and S. M. Mitchell, AIDS 12 (1998) 613. [18] R. J. Whitley, M. A. Jacobson, N. D. Friedberg, N. G. Holland, D. A. Jabs, D. T. Dieterich, D. W. Hardy, A. M. Polis, T. A. Deutsch, J. Feinberg, A. S. Spector, S. Walmsley, W. L. Drew, G. W. Powderly, P. D. Griffiths, C. A. Benson, and H. A. Kessler, Arch. Intern. Med. 158 (1998) 957. [19] M. Jouan, and C. Katlama, Int. J. Antimicrob. Agents 13 (1999) 1. [20] Z.-Q. Zhang, T. Schuler, M. Zupancic, S. Wietgrefe, K. A. Staskus, A. K. Reimann, A. T. Reinhart, M. Rogan, W. Cavert, J. C. Miller, S. R. Veazey, D. Notermans, S. Little, S. A. Danner, D. D. Richman, D. Havlir, J. Wong, L. H. Jordan, W. T. Schacker, P. Racz, K. Tener-Racz, L. N. Letvin, S. Wolinsky, and T. A. Haase, Science 286 (1999) 1353. [21] E. De Clercq, J. Med. Chem. 38 (1995) 2491. [22] L. Naesens, R. Snoeck, G. Andrei, J. Balzarini, J. Neyts, and E. De Clercq, Antiviral Chem. Chemother. 8(1997) 1.
278
E. De Clercq / Molecular Targets and Compounds for Anti-HIV Therapy
[23] A. Fridland, Tenofovir, Curr. Opin. Anti-Infect. Invest. Drugs 2 (2000) 295. [24] O. M. Miller, N. A. Margot, K. Hertogs, B. Larder, and V. Miller, 4th International Workshop on Drug Resistance & Treatment Strategies, 12–16 June 2000, Sitges, Spain. Antiviral Ther. 5, Suppl. 3, p. 4, abstract 4. (2000) [25] J.-M. De Muys, H. Gourdeau, N. Nguyen-Ba, D. L. Taylor, P.S. Ahmed, T. Mansour, C. Locas, N. Richard, M. A. Wainberg, and R. F. Rando, Antimicrob. Agents Chemother. 43 (1999) 1835. [26] Z. Gu, M. A. Wainberg, N. Nguyen-Ba, L. L'Heureux, J.-M. De Muys, T. L. Bowlin, and R. F. Rando, Antimicrob. Agents Chemother. 43 (1999) 2376. [27] H. Uchida, E.N. Kodama, K. Yoshimura, Y. Maeda, P. Kosalaraksa, V. Maroun, Y.-L. Qiu, J. Zemlicka, and H. Mitsuya, Antimicrob. Agents Chemother. 43 (1999) 1487. [28] K. Yoshimura, R. Feldman, E. Kodama, M. F. Kavlick, Y.-L. Qiu, J. Zemlicka, and H. Mitsuya, Antimicrob. Agents Chemother. 43 (1999) 2479. [29] M. Daneshtalab, Emtricitabine, Curr. Opin. Anti-Infect. Invest. Drugs 2 (2000) 272. [30] P.A. Furman, J. Jeffrey, L. L. Kiefer, J. Y. Feng, K. S. Anderson, K. Borroto-Esoda, E. Hill, W. C. Copeland, C. K. Chu, J.-P. Sommadossi, I. Liberman, R. F. Schinazi, and G. R. Painter, Antimicrob. Agents Chemother. 45 (2001) 158. [31] P.A. Furman, R. Chin, K. Borroto-Esoda, B. Dent, A. Bartholomeusz, and S. Locarnini, (2000) 4th International Workshop on Drug Resistance & Treatment Strategies, 12–16 June 2000, Sitges, Spain. Antiviral Ther. 5, Suppl. 3, p. 5, abstract 5. [32] C. A. Stoddart, M. E. Moreno, V. D. Linquist-Stepps, C. Bare, M. R. Bogan, A. Gobbi, R. W. Jr. Buckheit, J. Bedard, R. F. Rando, and J. M. McCune, Antimicrob. Agents Chemother. 44 (2000) 783. [33] P. R. Meyer, S. E. Matsuura, A. G. So, and W. A. Scott, Proc. Natl. Acad. Sci. USA 95 (1998) 13471. [34] P. R. Meyer, A. G. So, and W. A. Scott, Int. Antiviral News 7 (1999) 124. [35] M. Gotte, D. Arion, M. A. Parniak, and M. A. Wainberg, J. Virol. 74 (2000) 3579. [36] P. R. Meyer, R. Chopra, E. Pendarvis, S. Matsuura, H. Bazmi, A. G. So, J. W. Mellors, and W. A. Scott, (2000) 4th International Workshop on Drug Resistance & Treatment Strategies, 12–16 June 2000, Sitges, Spain. Antiviral Ther. 5, Suppl. 3, p. 13, abstract 14. [37] C. Perigaud, A.-M. Aubertin, S. Benzaria, H. Pelicano, J.-L. Girardet, G. Maury, G. Gosselin, A. Kirn, and J.-L. Imbach, Biochem. Pharmacol. 48 (1994) 11. [38] C. McGuigan, H.-W. Tsang, D. Cahard, K. Turner, S. Velazquez, A. Salgado, L. Bidois, L. Naesens, E. De Clercq, and J. Balzarini, Antiviral Res. 35 (1997) 195. [39] A.Q. Siddiqui, C. Ballatore, C. McGuigan, E. De Clercq, and J. Balzarini, J. Med. Chem. 42 (1999) 393. [40] A.Q. Siddiqui, C. McGuigan, C. Ballatore, F. Zuccotto, I. H. Gilbert, E. De Clercq, and J. Balzarini, J. Med. Chem. 42 (1999) 4122. [41] J. Balzarini, A. Karlsson, S. Aquaro, C.-F. Perno, D. Cahard, L. Naesens, E. De Clercq, and C. McGuigan, Proc. Natl. Acad. Sci. USA 93 (1996) 7295. [42] D. Saboulard, L. Naesens, D. Cahard, A. Salgado, R. Pathirana, S. Velazquez, C. McGuigan, E. De Clercq, and J. Balzarini, Mol. Pharmacol. 56 (1999) 693. [43] S. Aquaro, O. Wedgwood, C. Yarnold, D. Cahard, R. Pathinara, C. McGuigan, R. Calio, E. De Clercq, J. Balzarini, and C. F. Perno, Antimicrob. Agents Chemother. 44 (2000) 173. [44] C. Meier, M. Lorey, E. De Clercq, and J. Balzarini, Bioorg. Med. Chem. Lett. 7 (1997) 99. [45] C. Meier, T. Knispel, E. De Clercq, and J. Balzarini, Bioorg. Med. Chem. Lett. 7 (1997) 1577. [46] C. Meier, M. Lorey, E. De Clercq, and J. Balzarini, J. Med. Chem. 41 (1998) 1417. [47] C. Meier, T. Knispel, E. De Clercq, and J. Balzarini, J. Med. Chem. 42 (1999) 1604. [48] C. Meier, T. Knispel, V. E. Marquez, M. A. Siddiqui, E. De Clercq, and J. Balzarini, J. Med. Chem. 42(1999) 1615. [49] E. De Clercq, Trends Pharmacol. Sci. 21 (2000) 167. [50] E. De Clercq, Int. Antiviral News 8 (2000) 53.
279
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Proteomics in Drug Discovery: Potential and Limitations M. Fountoulakis F. Hoffmann-La Roche Ltd., Pharmaceutical Research, Genomics Technologies, Basel, Switzerland Abstract. Proteomics is the large-scale, high throughput analysis of proteins. Its goals are the detection of novel drug targets, diagnostic markers and the investigation of biological events. The bloom that proteomics enjoys today has resulted from corresponding developments in other sciences and technologies, such as computer and software, mass spectrometry, other analytical methods and in particular genomics, which delivered an enormous amount of information from the sequencing of the genome of many organisms. Proteomics is still under development and it possesses the potential of becoming a powerful tool in medicinal research. In this article, selected examples of application of proteomics in drug discovery, the potential of contribution in future discoveries and the limitations of the current technologies are discussed.
What is proteomics Proteomics is a relatively new term and refers to the technology-based science which studies the proteome, i.e. the proteins of an organism or an organ, their post-translational modifications, their interactions and in particular the changes in their levels and their modifications. The changes can be the result of specific diseases or the influence of various external factors, such as toxic agents. Proteomics has as goal the detection of novel drug targets, diagnostic markers and the investigation of biological events [1]. It is comprised of two steps, (i) the separation of protein mixtures, usually by two-dimensional (2-D) electrophoresis and (ii) the identification of the separated proteins by analytical techniques, mainly by mass spectrometry [2]. The process is facilitated by the use of highly sophisticated software for protein identification and advanced image analysis. The 2-D electrophoresis itself comprises two steps (dimensions): (i) separation of the proteins on the basis of differences in their net charge by isoelectric focusing (IEF) and (ii) separation of the focused proteins on the basis of differences in their molecular masses by sodium dodecyl sulfate (SDS) polyacrylamide gels [3–4]. For the performance of IEF, a pH gradient is required. As such, immobilized pH gradient (IPG) strips are usually employed on which the pH gradient has been already formed by acrylamide derivatives, called immobilines. They are weak acids and bases with a buffering capacity, copolymerized with acrylamide and the polyacrylamide gel is then dried on a plastic sheet [5]. The increased application of proteomics today is due to a large
280
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
extent to the introduction of the IPG strips, which are commercially available. The major advantage of using IPG strips is the ability to maintain a high reproducibility while a relatively large protein amount is applied [6]. The increase in reproducibility has allowed the high throughput analysis of proteomes and the application of larger sample quantities has enabled the subsequent identification of a large number of proteins. The use of narrow pH range strips (for example of 1 pH unit) provides a higher resolution, allows the detection of a larger number of protein isoforms and represents an additional advantage of employing IPG strips. Figure 1 shows the 2-D electrophoretic analysis of the soluble proteins of the bacterium Escherichia coli, separated first on a broad pH range IPG strip and then on a SDS polyacrylamide gel of large format.
Figure 1. Two-dimensional electrophoretic analysis of Escherichia coli proteins. One mg of cytosolic proteins were separated on a pH 3–10 nonlinear immobilized pH gradient strip, followed by an 11% sodium dodecyl sulfate polyacrylamide gel (dimensions of the full gel about 18 x 20 cm). The gel was stained with colloidal Coomassie blue.
Following separation by 2-D electrophoresis, proteins are usually identified by mass spectrometry. The proteins are in-gel digested with a protease, such as trypsin. For the proteins that have their genomic sequence in a database, the most efficient identification method presently available is the matrix-assisted laser desorption ionisation time of flight mass spectrometry (MALDI-TOF-MS) [7]. By this method more than 1000 proteins can be analysed daily by one person [8–10]. About 1000-3000 protein spots can be visualised on one 2-D gel, where 1 mg of the total protein amount has been applied and stained with Coomassie blue. Approximately half of the visible spots are available at sufficient quantities to be analysed for identity assignment. From one single gel, up to 1500 spots can be analysed by MALDI-MS and up to 1000 proteins are usually identified which however are the products of about 200-400 different genes. Following this approach, 2-D databases
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
281
for various proteomes have been constructed, usually including a few hundred different gene products [11–18]. The largest 2-D proteome databases, such as of the bacterium Haemophilus influenzae, include approximately 500 mapped proteins [19]. Many of the unidentified proteins are probably not expressed at sufficient amounts to be visualised and can not be detected on account of limitations of the technology (see below). Fig. 2 shows the workflow in proteomics in its classical definition as above, i.e. protein separation by 2-D electrophoresis and identification by mass spectrometry, as well as alternative approaches for a proteome analysis. Moreover, other proteomics technologies also exist, Protein Sample Preparation (cell disruption, protein extraction, fractionation)
Two-dimensional Electrophoresis (isoelectric focusing, SDS-gels) Protein Identification
j
j
Detection of Protein Changes
(construction of databases)
i
j
(comparison of protein levels, modifications)
Coomassie blue-stained gels
"1
In-gel digestion
!^
Electrotransfer to PVDF membrane "" T
N-terminal
Hydrolysis
sequencing
I Amino acid analysis
Mass spectrometry (MALDI-MS, electrospray)
Search in Databases (data storage)
Figure 2. Workflow in proteomics. Proteomics in its classical definition involves protein separation by two-dimensional electrophoresis and protein identification by mass spectrometry as well as detection of changes in the level and modifications of proteins. MALDI-MS, matrix-assisted laser desorption ionization mass spectrometry; PVDF, polyvinylidene difluoride;SDS, sodium dodecyl sulfate.
currently in a developmental stage. Their goal is also the high throughput proteome analysis, based on other separation and identification approaches, for example the use of protein chips. Proteins are often represented by more than one spot in 2-D gels. Examples of heterogeneity of brain, cerebrospinal fluid and liver proteins, which are represented by multiple spots, are shown in Fig. 3. We found that on average, 2-5 spots of brain and 10-20 spots of liver proteins correspond to one gene product [17-18 and unpublished results]. Presently, we do not know either the origin or the biological significance of most cases of observed heterogeneity. It may be the consequence of post-transational modifications, such as phosphorylation, glycosylation or deamidation, which result in the alteration of the pI of the molecule and its focusing position. Heterogeneity may also result from artifacts of the
282
M. Fountouhkis / Proteomics in Drug Discovery: Potential and Limitations
technology, such as carbamylation of the proteins upon prolonged contact of the sample with urea.
Figure 3. Partial 2-D gel images showing the heterogeneity of three proteins derived from the indicated proteomes. CSF,cerebrospinal fluid.
Drug discovery and potential of proteomics Drug dscovery is a lengthy and costly process. It takes approximately 10-15 years until a drug comes on the market, and the average costs for the development and introduction of a drug product can soar to 500 million dollars. Drugs are usually lowmolecular mass, organic compounds, synthesized in chemical laboratories. There are also many proteins, such as interferons, used as drugs, which are mainly prepared by biotechnological approaches. Their production costs are higher than those of the synthetic drugs, but the development and introduction costs are comparable. Many of the drugs currently in use have been discovered in an empirical way and some show strong side effects. The aim of the pharmaceutical science is to develop specific drugs against certain disorders with possibly few side effects. To reach such a goal, the contribution of new sciences is necessary.
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
283
In the development of a drug many basic sciences are involved, i.e. Biology, Biochemistry, Medicine and New Technologies. Proteomics and genomics belong to the New Technologies and represent alternative approaches to search for drug targets. The genomic analysis quickly provides us with data about the mRNA levels (here the corresponding terms are transcriptome and transcriptomics). However, knowledge of the transcription levels does not necessarily inform us about the protein levels or the number of modified protein forms, information, which can only result from the proteome analysis. Life is a dynamic process and not all-possible proteins are expressed at any time point. Moreover, the pattern of protein expression changes depending on various factors, as the stage of development of an organism and an organism's physiological state. It seems that the more complex the genome, the less of the total possible proteins will be expressed at any particular moment. Thus, proteomics supplements the genomics data with information about what gene products are being made, in what cell type, at what amounts and under what conditions and how protein levels and modifications change as a result of specific diseases or external challenges. Various expressions exist for essentially the same science, such as functional proteomics, large-scale proteomics, pharmaceutical proteomics, neuroproteomics etc. All of them have the same goal, the identification of proteins and the functions they exert in the various biological pathways [20-26]. In the drug discovery process, first a drug target needs to be identified, usually a protein that is involved in a biological pathway. Since proteomics is the study of proteins, their modifications, their changes and interactions, it represents the ideal method to search for protein drug targets. Its major advantage is that it enables the simultaneous observation of thousands of unknown proteins. No other method is as efficient at the present time. Thus, the search for unknown drug targets can be enormously facilitated, in particular the search for unknown species associated with other proteins, known to be involved in certain disorders. The identification of disease-related differences is usually a difficult task. Proteomics can relatively easily identify changes in bacterial proteomes and also in cell lines. The analysis of mammalian tissues, such as brain, is more difficult, as many factors are involved, such as differences in age [27], sex [28-29], possibly other diseases, treatment with medicines, as well as technical, disease-unrelated factors, such as post-mortem time [30], improper treatment of the samples etc., which all can affect a clear discrimination between healthy and diseased states of interest. Until now, the research in proteomics mainly resulted in development of the technologies, but the real, systematic application of the advanced proteomics methods in drug discovery has yet to come. Proteomics has been applied as a diagnostic tool in the investigation of infectious diseases, cancer, heart and diseases of the central nervous system, which are summarized in a series of review articles [31-38]. Here we shortly describe our findings of using proteomics technologies in the investigation of neurological disorders. Application of proteomics in neurodegeneration studies: We applied proteomics in the investigation of protein changes in disorders of the central nervous system, such as Alzheimer's disease (AD), Down syndrome (DS) and Pick's disease. Alzheimer's disease is a well-studied dementia affecting mainly elderly people, for which no reliable premortem diagnosis marker exists today. Down syndrome is the most frequent genetic cause of dementia. Although the trisomic state is responsible for the phenotype, the pathomechanisms are not well understood. Almost all subjects with Down syndrome over 40 years show neuropathological and neurochemical abnormalities on post-mortem brain examinations, indistinguishable from those seen in Alzheimer's disease [39-42]. Thus, the results from the DS may be useful in the AD studies. Pick's disease is also a neurodegenerative disorder not well defined presently.
284
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
Figure 4. Two-dimensional database of human brain proteins from the parietal cortex of a control individual. In the map, spots are indicated representing proteins with deranged levels in Alzheimer's disease and Down syndrome. The proteins were identified and the differences were quantified applying proteomics technologies.
We analyzed tissue from seven regions of the brain from the disease groups and from controls with no history of neurological disease. Fig. 4 shows a 2-D map of the human brain proteins, where species are indicated for which deranged levels in AD and DS were found. The proteins with deranged levels, some of which may be potential drug targets, have mainly neurotransmission, guidance, transport and signal transduction functions, are involved in metabolism and detoxification pathways and in conformational changes. We found changed levels for dihydropyrimidinase-related protein (DRP) 2 [43].
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
285
Dihydropyrimidinase-related proteins show homology to chicken collapsin 62 kDa and the rat turned on after division protein. DRPs are expressed in relatively large amounts in the brain, DRP-1, -3 and -4 mainly in the neonatal brain and DPR-2 in both neonatal and adult brain and show a high heterogeneity (Fig. 3) [27, 44]. The functions of DRPs are not clear. It is possible that these molecules are involved in neuronal migration during brain development. When the latter becomes less pronounced in the adult brain, DRP-2, which is almost equally expressed in both adult and neonatal brain, may serve as a guidance cue in repair and remodeling in adult brain. Reduced levels of DRP-2 in the Alzheimer and Down syndrome brain may have consequences in the functions of the protein [43]. We also observed reduced levels of synaptosomal-protein beta-soluble Nethylmaleimide-sensitive factor attachment protein (beta-SNAP) and synaptotagmin 1 in the AD and DS brain. SNAPs participate in vesicular transport between endoplasmic reticulum and Golgi apparatus. Synaptotagmin 1 is a calcium-binding protein, localized at synaptic vesicles and chromaffin granules, which forms complexes with SNAPs. It has probably a regulatory role in membrane interactions during trafficking of synaptic vesicles at the active zone of the synapse [45]. The levels of dynamin 1 were also decreased by about 50% in the disease groups. Dynamin 1 is a phosphoprotein with GTPase activity, abundant in nerve terminals, which is involved in endocytosis and neural transmission. Decreased dymanin levels may count for deficient wiring in the diseased brain (unpublished results). In the AD and DS groups, we found a manifold increase in the levels of the glial fibrillary acidic protein (GFAP), a known marker of neuronal decay and brain damage, which distinguishes astrocytes from other glial cells during development of the central nervous system [46]. We also observed increased levels for the 14-3-3y and 14-3-3e proteins in the disease groups by about 1.5-fold [47]. The 14-3-3 proteins exert complex functions in signal transduction pathways. Deranged levels may reflect impaired signaling and apoptosis in the brain. The levels of synaptosomal associated protein 25 kDa (snap-25) decreased in the two disease groups to approximately 40% [46]. Snap-25 is widely distributed in the brain, is an integral constituent of the synaptic core complex, participates in synaptic vesicle exocytosis and is involved in the formation of presynaptic sites. Decreased snap-25 levels may lead to deranged functions in exocytosis and neurotransmission. Changes were also observed in the levels of histamine-releasing factor [48] glyceraldehyde 3-phosphate dehydrogenase [49], NADH: ubiquinone oxidoreductase [50], superoxide dismutase, carbonyl reductase, alcohol dehydrogenase, voltage-dependent anion channel proteins VDAC1 and VDAC2 [51], heat shock proteins [52-53] and others. Further applications of proteomics: Proteomics has just started contributing to drug development. Various "Human Proteome" projects are currently running and the knowledge that will be accumulated is probably going to revolutionize the protein science in the future. On account of the large heterogeneity and variations, the analysis of a proteome is much more laborious than that of a genome. In the near future, proteomics can supplement the effort of genomics by detecting differences in protein levels and posttranslational modifications and species involved in biological events. Post-translational modifications resulting from various diseases may carry important biological information and proteomics will undertake the task for their identification and understanding of their meaning. Moreover, proteomics is already used and could be to a larger extent employed in the investigation of the toxicity of drug candidates [54-56]. In order to study their efficacy, these substances are subjected to three phases of human clinical trials, which generate enormous costs to the pharmaceutical companies. Early detection of eventual toxicity can save costs and effort and allocate resources in the efficient search of new drug candidates.
286
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
We have applied proteomics to study liver toxicity in mice treated with acetaminophen, the known antipyretic and analgesic agent, which is toxic at high doses. The mechanism of the toxicity is not well understood [57]. We found that the levels of about 30 proteins changed following acetaminophen administration. The changes were in the order of 10-50% of the control value and appeared mainly at a high dose of acetaminophen [16]. Administration of carbon tetrachloride in rats also resulted in significant changes in the liver protein levels (unpublished results). We further studied the effect of administration of kainic acid, a potent neurotoxin and excitatory amino acid, on the levels of brain proteins. The proteomic analysis revealed altered regulation of heat shock proteins, neuronal death, cytoskeletal disruption and mitochondrial derangement by systemic kainic acid administration [58]. Limitations The current proteomics technologies show certain limitations, which need to be overcome in order for the technology to develop its full power and capabilities. The limitations are of two kinds: (i) those related to the composition of the proteome to be analyzed, mainly concerning the protein expression levels and (ii) technical limitations of the technology. Proteomics is therefore simultaneously confronted with two problems, namely to increase the quantity of the low-abundance proteins so that they can be detected and to apply the proper analytical technologies to detect all species of a mixture. For the detection of a protein, three prerequisites must be fulfilled: (i) The protein should be available in a sufficient amount in the protein mixture prior to the 2-D electrophoretic analysis (otherwise the protein needs to be enriched, for example by chromatographic methods), (ii) The protein should be brought into solution with mild detergents and chaotropes, reagents that are compatible with the isoelectric focusing and kept in solution during the first dimensional separation (for proteins difficult to solubilize, strong solubilizing agents should be employed, such as sodium dodecyl sulfate). (iii) The protein should belong to the category of proteins that can be visualized by 2-D electrophoresis, i.e. it should have average pI (usually between pH 4 and 12) and molecular mass (between 10 and 150 kDa) values and should not include strong hydrophobic stretches (otherwise variations of the 2-D electrophoresis or alternative separation procedures should be applied) [59]. Here we shortly describe possibilities to overcome the limitations mentioned. Enrichment of low-abundance proteins: Not all-possible gene products of an organism are expressed at the same level at a certain time point. Some species, such as structural proteins, heat shock proteins or elongation factors, are produced at many thousands of copies per cell, whereas others, such as transcription factors, exert their functions at very low concentrations, maybe at a few copies per cell. In the small sample volume (about 10500 ul) usually used for the proteomic analysis, the majority of the expressed proteins are not present in sufficient quantities to be visualized and identified. Therefore, proteins present in low copy numbers can not be readily detected during the analysis of total proteins. The study of the low copy number gene products may be more interesting in investigating biological events than of their high abundance counterparts. To the present, samples representing the total protein mixture have been usually analyzed and mainly only the abundant, hydrophilic components have been visualized. These proteins could be solubilized with reagents compatible with isoelectric focusing. Such an analysis provides us with a limited image of the proteome and is insufficient for the study of biological events. For the enrichment of low-abundance proteins from crude extracts, biochemical protein enrichment methods are usually employed [60]. These methods separate the original protein mixture into simpler fractions, so that each contains a lower number of total proteins in
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
287
comparison with the starting material. This increases the likelihood of detection of lowabundance proteins manifold. Fig. 5 shows the general scheme followed for the enrichment of low-copy-number bacterial proteins, but similar pathways are valid for eukaryotic systems [59]. The first step is usually the separation of the mixture into organelles, i.e. cytosolic, membranal, mitochondrial etc. protein fractions. In bacteria, cell envelope and membrane proteins represent approximately 15% of the total cellular proteins, so that the probability of detection of a low-abundance membrane protein increases by about 10-fold if a pre-fractionation is employed. In eukaryotic systems, the separation of additional organelles, such as Golgi apparatus, increases the likelihood of detection of the species specific to that organelle. After the separation of the organelles, a further protein enrichment from larger volumes can be achieved by fractionation of the protein mixture, using chromatographic steps or preparative electrophoretic procedures [59-60].
Bacterial Cell Paste (cell disruption, centrifugation)
Pellet (cell envelopes, membranes)
Cytosol (soluble cytoplasmic proteins)
Solubilization (mild detergents and chaotropes)
Chromatography (affinity, hydrophobicity, ion exchange etc.)
Two-dimensional Electrophoresis (broad-, narrow-, basic-range 1PG strips, Tricine gels, 2-detergent system)
Figure 5. General scheme showing the steps that can be followed for the enrichment of low- abundance bacterial proteins. In eukaryotic systems, more "pellet" fractions can be prepared, for example mitochondria, membranes, Golgi apparatus etc.
Enrichment of low-abundance proteins from large volumes can be achieved by selective fractionation, prior or in addition to the chromatographic approaches, using precipitation agents, such as ammonium sulfate, polyethylene glycol or organic solvents. Chromatographic methods separate complex protein mixtures into simpler fractions on the basis of different binding principles and every approach adds a unique resolving power. The proteins are usually separated on the basis of affinity, charge, hydrophobicity, size etc.
288
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
[60]. The choice of the chromatographic method best suited to fulfill the experimental requirements is essential for the success of the experiment. Sequential chromatographic steps are often required. We used four chromatographic steps to enrich low-abundance cytosolic bacterial proteins from larger volumes: affinity chromatography on Heparin [6162], ion exchange chromatography on Polybuffer Exchanger (chromatofocusing) [63], hydrophobic interaction chromatography on a Phenyl column [64] and hydroxyapatite chromatography [65]. Heparin, a highly sulfated glycosaminoglycan, has an affinity for nucleic acidbinding proteins and also functions as a high capacity cation exchanger. Heparin chromatography is relatively easy to perform. We applied this step to enrich low-abundance proteins of the bacterium Haemophilus influenzae. Approximately 150 different proteins were identified in the pools collected from the column and 50 of them were low-abundance species, which had not been found in the 2-D gel of the starting material. Approximately 40% of the heparin-bound proteins were nucleic-acid binding proteins, including ribosomal proteins and t-RNA synthetases. Basic proteins, in particular ribosomal proteins, could be efficiently enriched using this step [66]. Heparin chromatography was also used to enrich cytosolic brain proteins [44]. Chromatofocusing is an ion exchange chromatographic step. The proteins are bound to the gel matrix, Polybuffer Exchanger, and are eluted with a specific buffer, Polybuffer, in the order of their decreasing isoelectric points. Proper choice of the pH of equilibration and elution buffers and of the dimensions of the column can result in an efficient protein concentration and high resolution. This step is usually chosen because the ion exchanger has a high protein binding capacity and can discriminate and enrich proteins with minor differences in their isoelectric points (pI values). We employed the chromatofocusing step to enrich low-abundance proteins of H. influenzae. Most fractions, in particular those eluted with salt, contained mainly acidic proteins. About 125 unique proteins were identified in the pools. Approximately 30 of them were low-abundance gene products not identified before [63]. Fig. 6 shows the enrichment of two enzymes of the microorganism using this step. In hydrophobic interaction chromatography, proteins are separated on the basis of differences in their hydrophobicity. The proteins are adsorbed onto an uncharged matrix carrying hydrophobic groups, in the presence of salts. Elution is achieved by lowering the salt concentration. Due to the various numbers of hydrophobic sites carried by proteins, the hydrophobic interaction chromatography can efficiently fractionate complex protein mixtures. We used hydrophobic interaction chromatography to enrich low-abundance proteins of H. influenzae. About 200 different proteins bound to the column were identified, 30 of which were low-abundance gene products. This step mainly enriched proteins with catalytic activities as well as hypothetical proteins of a wide spectrum of pI values, including basic proteins [64]. Hydroxyapatite chromatography represents a standard protein purification procedure today and the gel binds a wide range of proteins by different mechanisms from the other separation techniques. The hydroxyapatite matrix carries positively charged (calcium) and negatively charged (phosphate) sites. Proteins bind either by nonspecific, electrostatic interactions between their positive charges and the negative charge on the matrix, or by complexation of their carboxyl groups with the calcium sites. Due to the complexity of the protein-hydroxyapatite interactions and in order to achieve a high resolution, various steps for the development of the column are usually applied. We used this step to enrich low-abundance proteins of the bacterium Escherichia coli. Approximately 300 different proteins were identified in the pools collected from the column and about 100 of them were low-abundance proteins. Basic as well as many low
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
289
molecular mass proteins and the proteolytic products of many proteins as well as their fulllength counterparts were enriched by this method [65].
Figure 6. Partial 2-D images, showing the enrichment of two Haemophilus influenzae proteins by chromatofocusing. A, total soluble proteins; B, proteins of a fraction collected from the column.
In addition to the classical chromatographic steps, preparative electrophoresis methods can be applied to enrich proteins from large volumes. They comprise separation of protein mixtures by preparative polyacrylamide gel electrophoresis on the basis of protein size usually in the presence of ionic detergents and preparative isoelectrofocusing on the basis of protein charge in the presence of ampholines. Multicompartment electrolyzers operating with isoelectric membranes may be very useful in the fractionation process [67]. Changes in the levels of low-abundance proteins, resulting from diseases or the effect of external factors, can only be detected after enrichment of the corresponding gene products from large volumes. For example, we employed ion exchange chromatography prior to proteomic analysis to enrich low-copy number cytosolic rat brain proteins from controls and animals treated with kainic acid. We observed changes in the levels of four lowabundance brain proteins. Three out of the four proteins with changed levels had not been detected in the cytosolic fraction before and the detection of the changes was possible only after protein enrichment [68]. The deranged protein levels suggest for the involvement of an apoptotic pathway, the recruitment of the heat shock protein machinery, the generation of an antioxidant response and probably the induction of repair mechanisms in the brain. Chromatographic approaches for protein enrichment have certain limitations. We observed a significant overlap between the H, influenzae proteins enriched by heparin chromatography, chromatofocusing and hydrophobic interaction chromatography. An estimated 40% of the species enriched by hydrophobic interaction chromatography were also enriched by heparin chromatography, and about the same percentage was also enriched by chromatofocusing. About 20% of the proteins were enriched by all three methods. After having applied two or more fractionation methods, the use of additional chromatographic separations may not result in the detection of significant numbers of new proteins not detected or enriched by the previous methods. This points to the limits of the chromatographic enriching techniques and suggests that a large percentage of the proteins not detected are expressed at very low levels. The various chromatographic steps, which we employed, resulted in the enrichment of both low- and high-abundance proteins. This is to be expected, since the high- and low-
290
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
abundance proteins bind to the column matrix on the basis of the same principles. Certain enriched proteins may represent up to 50% of the protein content in a particular fraction, thereby suppressing the low-abundance proteins and hindering their detection in 2-D gels (Fig. 6). High-abundance proteins, for example heat shock proteins and certain housekeeping enzymes in bacteria or albumin in plasma, can be removed prior to 2-D electrophoresis by affinity chromatography using specific antibodies. Protein analysis by 2-D electrophoresis:Following enrichment by any of the methods described above or similar approaches, the visualization of the proteins is usually performed by 2-D electrophoresis [69]. Technical details on the performance of the 2-D electrophoresis can be found in Walsh and Herbert [70] or in Westermeier [71]. Often, it is not sufficient to enrich a protein for a successful detection. Protein detection and identification has certain limitations. The detection in 2-D gels and identification of lowabundance proteins, already successfully enriched from dilute solutions, might be technologically limited because of their charge and size. Fig. 7 shows approximately 400 brain proteins identified from 2-D gels in our laboratory sorted according to their molecular mass and pI values. The majority of the proteins have average molecular mass and pI values. No protein was detected with a pI below 4 and relatively few were detected with a pI value higher than 10. No proteins were found with a molecular mass below 10 kDa or above 160 kDa. In such cases, alternative separation techniques may need to be employed, for example Tricine gels for the detection of the low molecular proteins [72]. In Tricine gels, polypeptides with a mass of about 4 kDa can be detected. Another reason for an unsuccesful detection might be related to the hydrophobicity and low solubility of certain proteins. A protein can only be visualized and analyzed if it can be brought and kept in solution during the whole enrichment process and 2-D electrophoretic analysis. In general, the solubility problem arises at two time points along the performance of the 2-D electrophoresis: (i) during the initial extraction step to solubilize membrane proteins (the cytosolic proteins are already in solution) using agents that are compatible with isoelectric focusing, such as urea and CHAPS, and (ii) during the performance of the first dimensional separation, when hydrophobic proteins could precipitate at their application positions. Poorly soluble proteins can be brought in solution with the use of strong detergents and chaotropes, such as SDS [73]. Hydrophobic proteins with multiple transmembrane regions do not usually enter the immobilized pH gradient strips during the first dimensional separation and thus they can not be detected. It seems that not the hydrophobicity of the entire protein is decisive whether it will enter the IPG strip but the amino acid sequence of the hydrophobic stretches. For example, cytochrome P450 2D6, a hydrophilic protein with one transmembrane region, was not detected in 2-D gels, but at the sample application position (unpublished results). No single solubilizing agent is sufficient for the solubilization, resolution and visualization of all proteins of a proteome. Membrane proteins can also be separated in a discontinuous, two-detergent system [19]. In this system, the first dimensional separation is performed in polyacrylamide gels in the presence of benzyldimethyl-n-hexadecylammonium chloride (16-BAC) and the second dimensional separation in typical SDS-gels.
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
120
100
1 -
80 60
29!
_
a
T
•
40 20
- -
Molecular Mass (kDa) Figure 7. Distribution of about 400 fetal brain proteins identified from 2-D gels by mass spectrometry in our group in relation to their theoretical pI (A) and molecular mass (B) values. The bars indicate the number of proteins found in the pI and molecular mass intervals indicated. No protein with a pI value below 4 and no protein smaller than 10 kDa were found.
Conclusions Proteomics is still in a developmental stage. In spite of the current limitations of a proteomic study, this is the only method for a high throughput qualitative and quantitative analysis of a protein mixture. Moreover, with the sequencing of the genomes of more than 30 organisms to date, many new proteins with no known functions have been deduced. These proteins are referred to as "hypothetical" or as "predicted coding regions", and some of them might be of biological interest. Many of these proteins are produced in low-copy numbers and no literature information is available today for their function or isolation. In our hands, a large number of hypothetical or unknown proteins have been identified following enrichment using chromatographic steps. The analysis of the proteomes of higher organisms is anticipated to be very complex and will probably require a combination of many analytical approaches, comprising multiple enrichment steps and efficient separation and detection methods. The completion of the sequencing of more genomes together with improvements in the analytical techniques will add new search dimensions and will lead to a more widespread application of the technology in drug discovery and the investigation of biological events in general.
292
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
References [1] M. Fountoulakis, Amino Acids (2002) (in press). [2] M. Fountoulakis and H.-W. Lahm J. Chromatogr. 826 (1998) 109. [3] P. H. O'Farrell, J. Biol. Chem. 250 (1975) 4007. [4] M. Fountoulakis, Encyclopedia of Separation Science ll/Electrophoresis, Academic Press, London, 2000, pp. 1356-1363. [5] B. Bjellqvist, K. Ek, P. G. Righetti, E. Gianazza, A. Gorg, R. Westermeier and W. Postel J. Biochem. Biophys. Methods 6 (1982) 317. [6] A. Gorg, C. Obermaier, G. Boguth, A. Harder, B. Scheibe, R. Wildgruber and W. Weiss, Electrophoresis 21 (2000) 1037. [7] W. J. Henzel, T. M. Billed, J. T. Stults, S. C. Wong, C. Grimley and C. Watanabe, Proc. Natl. Acad. Sci. USA 90 (1993) 5011. [8] M. Fountoulakis and H. LangenAnal. Biochem. 250 (1997) 153. [9] P. Berndt, U. Hobohm and H. Langen, Electrophoresis 20 (1999) 3521. [10] H.-W. Lahm and H. Langen, Electrophoresis 21 (2000) 2105. [11] M. Fountoulakis, J.-F. Juranville and P. Berndt, Electrophoresis 18 (1997) 2968. [12] H. Langen, C. Gray, D. Roder, J.F. Juranville, B. Takacs and M. Fountoulakis, Electrophoresis 18 (1997) 1184. [13] G. Lubec, O. Labudova, N. Cairns, P. Berndt, H. Langen and M. Fountoulakis, J. Neural Transm. Suppl. 57 (1999) 21. [14] H. Langen, P. Berndt, D. Roder, N. Cairns, G. Lubec and M. Fountoulakis, Electrophoresis 20 (1999) 907. [15] M. Fountoulakis, E. Schuller, R. Hardmeier, P. Berndt and G. Lubec, Electrophoresis 20 (1999) 3572. [16] M. Fountoulakis, P. Berndt, U. A. Boelsterli, F. Crameri, M. Winter, S. Albertini and L. SuterElectrophoresis 21 (2000) 2148. [17] M. Fountoulakis, J.-F. Juranville, P. Berndt, H. Langen and L. Suter, Electrophoresis 22 (2001) 1747. [18] M. Fountoulakis, P. Berndt, H. Langen and L. Suter, Electrophoresis (2002) (in press). [19] H. Langen, B. Takacs, S. Evers, P. Berndt, H.-W. Lahm, B. Wipf, C. Gray and M. Fountoulakis, Electrophoresis 21 (2000) 411. [20] S. Borman, Chem. Engin. News, issue July 9 (2001) 43. [21] A. Abbott, Nature 402 (1999) 715. [22] S. Borman, Chem. Engin. News, issue July 31 (2001) 31. [23] A. Dove, Nat. Biotechn. 17 (1999) 233. [24] D. Eisenberg, E. M. Marcotte, I. Xenarios and T. Yeates, Nature 405 (2000) 823. [25] A. Pandey and M. Mann, Nature 405 (2000) 837. [26] J. H. Wang and R. M. Hewick, Drug Discovery Today 4 (1999) 129. [27] M. Fountoulakis, R. Hardmeier, E. Schuller and G. Lubec, Electrophoresis 21 (2000) 673. [28] I. Miller, P. Haynes, M. Gemeiner, R. Aebersold, C. Manzoli, M. R. Lovati, M. Vignati, I. Eberini and E. Gianazza, Electrophoresis 19 (1998) 1493. [29] S. Steiner, D. Wahl, M. C. Varela, L. Aicher and P. Prieto, Electrophoresis 16 (1995) 1969. [30] M. Fountoulakis, R. Hardmeier, H. Hoger and G. Lubec, Exp. Neurology 167 (2000) 86. [31] P. R. Jungblut, U. Zimny-Arndt, E. Zeindl-Eberhart, J. Stulik, K. Koupilova, K.-P. Pleissner, A. Otto, E.C. Muller, W. Sokolowska-K6hler, G. Grabher and G. Stoffler, Electrophoresis 20 (1999) 2100. [32] A. A. Alaiya, B. Franzen, G. Auer and S. Linder, Electrophoresis 21 (2000) 1210. [33] R. E. Banks, M. J. Dunn, D. F. Hochstrasser, J.-C. Sanchez, W. Blackstock, D. J. Pappin and P. J. Selby, Lancet 356 (2000) 1749. [34] P. Cash, Electrophoresis 21 (2000) 1187. [35] D. F. Hochstrasser, Proteomics. In M. R. Wilkins, K. L. Williams, R. D. Appel, D. F. Hochstrasser (Eds), Proteome research: New Frontiers in Functional Genomics. Springer, Berlin, 1997, pp. 187-219. [36] V. E. Bichsel, L. A. Liotta and E. F. Petricoin III, The Cancer Journal 7 (2001) 69. [37] S. J. Cordwell, A. S. Nouwens and B. J. Walsh, Proteomics 1 (2001) 461. [38] C. Rohlff, Electrophoresis 21 (2000) 1227. [39] S. E. Antonarakis, Genomics 51 (1998) 1. [40] N. J. Cairns, J. Neural Transm. Suppl. 57 (1999) 61. [41] S. M. de la Monte. J. Neural Transm. Suppl. 57 (1999) 1. [42] B. L. Shapiro. J. Neural Transm. Suppl. 57 (1999) 41. [43] G. Lubec, M. Nonaka. K. Krapfenbauer, M. Gratzer. N. Cairns and M. Fountoulakis. J. Neural Transm. Suppl. 57 (1999) 161. [44] K. Karlsson. N Cairns. G Lubec and M. Fountoulakis. Electrophoresis 20(1999) 2970.
M. Fountoulakis / Proteomics in Drug Discovery: Potential and Limitations
293
[45] B. C. Yoo, N. Cairns, M. Fountoulakis and G. Lubec, Dement. Geriatr. Cogn. Disord. 12 (2001) 219. [46] S. Greber, G. Lubec, N. Cairns and M. Fountoulakis, Electrophoresis 20 (1999) 928. [47] M. Fountoulakis, N. Cairns and G. Lubec, J. Neural Transm. Suppl. 57 (1999) 323. [48] S. H. Kim, N. Cairns, M. Fountoulakis and G. Lubec, Neurosci Lett. 300 (2001) 41. [49] G. Lubec, O. Labudova, N. Cairns and M. FountoulakisNeuroscience Lett. 260 (1999) 141. [50] S. H. Kim, R. Vlkolinsky, N. Cairns, M. Fountoulakis and G. Lubec, Life Science 68 (2001) 2741. [51] B. C. Yoo, M. Fountoulakis, N. Cairns and G. Lubec, Electrophoresis 22 (2001) 172. [52] B. C. Yoo, S. H. Kim, N. Cairns, M. Fountoulakis and G. Lubec, Biochem. Biophys. Res. Communications 280 (2001) 249. [53) B. C. Yoo, R. Vlkolinsky, E. Engidawork, N. Cairns, M. Fountoulakis and G. Lubec, Electrophoresis 22 (2001) 1233. [54] Y. Qiu, L. Z. Benet and A. L. BurlingameJ. Biol. Chem. 273 (1998) 17940. [55] S. Steiner and N. L. Anderson, Toxicol. Lett. 112-113 (2000) 467. [56] S. Steiner and F. A. Witzmann, Electrophoresis 21 (2000) 2099. [57] S. D. Cohen and E. A. Khairalaah, Acetaminophen. In: G. Sipes, C. A. McQueen and A. J. Gandolfi (Eds.), Comprehensive toxicology, Vol. 9: Hepatic and gastrointestinal toxicology. Pergamon, Cambridge, UK, 1997, pp. 329-343. [58] K. Krapfenbauer, M. Berger, G. Lubec and M. Fountoulakis, Electrophoresis 22 (2001) 2086. [59] M. Fountoulakis and B. Takacs, Methods Enzymology (2002) (in press). [60] B. J. Takacs, Protein purification: Theoretical and methodological considerations. In: R. A. Meyers (Ed.), Encyclopedia of Analytical Chemistry. John Wiley and Sons Ltd., Chichester, 2001, pp. 5955-5970. [61] M. Fountoulakis, H. Langen, S. Evers, C. Gray C and B. Takacs, Electrophoresis 18 (1997) 1193. [62] M. Fountoulakis and B. Takacs, Protein Expression Purification 14 (1998) 113. [63] M. Fountoulakis, H. Langen, C. Gray and B. Takacs, J. Chromatogr. 806 (1998) 279. [64] M. Fountoulakis, M.-F. Takacs and B. Takacs, J. Chromatogr. 833 (1999) 157. [65] M. Fountoulakis, M.-F. Takacs, P. Berndt, H. Langen and B. Takacs, Electrophoresis 20 (1999) 2181. [66] M. Fountoulakis, B. Takacs and H. Langen, Electrophoresis 19 (1998) 761. [67] P. G. Righetti, A. Castagna and B. Herbert, Anal. Chem. 73 (2001) 321. [68] K. Krapfenbauer, M. Berger, G. Lubec and M. Fountoulakis, Eur. J. Biochem. 268 (2001) 3532. [69] H. Langen, D. Roder, J.-F. Juranville and M. Fountoulakis, Electrophoresis 18 (1997) 2085. [70] B. J. Walsh and B. Herbert, (1998) http://rbams3115/Pages/2DPAGE/ABRFNews_2dpage.html [71] R. Westermeier, Electrophoresis in Practice. VCH Verlagsgesellschaft, Weinheim, 1993. [72] M. Fountoulakis, J.-F. Juranville, D. Roder, S. Evers, P. Berndt and H. LangenElectrophoresis 19 (1998) 1819. [73] M. Fountoulakis and B. Takacs, Electrophoresis 22 (2001) 1593.
294
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Evaluation of Tumor Capabilities for Recurrence in Patients with Larynx and Pharynx Malignancies on the Basis of DNA Criterion S. Andreychenko O.Kolomiychenko Institute of Otolaryngology of Academy of Medical Sciences Kiev, 03057, Ukraine
Abstract. A special criterion adequate for prediction of tumor behavior as well as for an evaluation of post-operation recovery duration in patients with larynx and pharynx malignancies has been developed. The criterion in question was called a coefficient of tumor aggressiveness - A. It presents itself a proportion showing a relation of DNA concentration in tumor to that in nearby normal on convention tissue. A depends on tumor localization, varying in the range 1,15 - 2,02 for larynx and 1,55 - 2,62 for pharynx, respectively. Values of A 1,8 -1,9 are the most beneficial for post-operation recovery in patients with larynx malignancies, while for pharynx strong rehabilitation was observed in the diapason of 2,0 - 2,2. Beyond these borders a chance for tumor recurrence significantly increases. A correlates with the appearance of polyploid cell clones persistent to oncogenic transformation.
Introduction Nowadays cancer presents itself a most serious threat for a mankind because of its wide spreading in different races and populations all over the world. According to statistical data in Ukraine appear 2 new oncology patients among each 1000 of inhabitants, while in USA more than 40 000 new cases only of head & neck cancer are expected to be revealed in the coming year [1]. However, in Ukraine and several other countries of Eastern Europe the
5. Andreychenko / Evaluation of Tumor Capabilities for Recurrence
295
situation is being dramatically deteriorated by the consequences of Chernobyl catastrophe happened on April 25, 1986 on the Chernobyl atomic power station. This incident damaged a lot of people because of their continuous exposure to low-dose ionizing irradiation in the period of disaster and afterwards. As a result of the accident, cancer begun to grow in an unpredictable manner, causing either rapid metastases formation or sudden impetuous progressing to the ultimate stages of the disease. Apart from this, an obvious downfall in lower age limit for malignant transformation has been observed. In the connection to all aforesaid a search of an adequate criterion for tumor behavior evaluation appears to be of urgent need. This could be indicative as to the tumor capabilities in further recurrence after surgical intervention. Materials and Methods The examinations were carried out in 2 groups of patients with cancer of larynx and pharynx, respectively. Each group contained 30 persons. The malignant nature of tumors was confirmed on the basis of histopathological analyses executed on biopsies stained with hematoxylin and eosin after they had been primarily fixed in Holland's fluid, embedded in paraffin wax and sectioned at 5 um [2]. All the patients underwent surgery for extirpating malignancies. Under the operations samples of tumors as well as nearby normal on convention tissue were taken off to be processed both for total DNA concentration quantitative determination and nuclear DNA contents cytophotometry. The oncology patients in the age of 40 to 65 years were in stage 11/111 of the disease, subsequently followed the same standard scheme of treatment. Determination of total DNA concentration in tissue biopsies:The analyses were performed according to specially developed protocol. Thus, the minced biopsies were previously fixed in cold methanol (-18 C ) and stored in it for at least 24 h. Afterwards the samples were washed 3 times in 1 N perchloric acid (HC1O4) to extract free nucleotides and centrifuged at 1500 g , the supematants being discarded. The saved sediments were treated with 0,5 N HC1O4 twice at 70 °C for 20 min each time and then centrifuged at 1500 g, the supematants being united together and used in Dishe reaction with diphenylamine in Burton's modification [3]. According to the procedure to a sample aliquot 2 volumes of reagent consisting of 1,5 % diphenylamine, 1,5 % sulfuric acid (H2SO4) and 0,008% acetaldehyde in glacial acetic acid (CH3COOH) were added. Color intensity in reaction mixture was developing for 18 h at 30° C and then measured at 598 nm on spectrophotometer "SF-46" (Russia). In the samples DNA content was determined by the aid of calibration curve built beforehand for known quantities of similar hydrolyzed calf thymus DNA (Sigma). DNA cytophotometry:Tissue samples immediately after extirpation were put into physiological solution of 0,9% sodium chloride (NaCl) and then used for smear preparations. Following fixation in Carney's solution - ethanol: chloroform: acetic acid (6:3:1) - for at least 3 h the smears were passed through descending ethanol series (50 %, 30 %, 15 %) up to water, rinsed with cold 0,1 N hydrochloric acid (HC1) and then hydrolyzed in 1 N HC1 for 9 min at 60 ° C. Staining with basic fuchsin dye was performed during 2 h in the dark at room temperature [4]. For removing free dye and color fastening the preparations were placed in three changes of SO2 - water for 10 min in each, washed with water and passed through ascendant ethanol series (15 %, 30 %, 50 %, 70 %, 90 %, 96 %, absolute), ethanol: xylene mixture and xylene. After embedding in DPX mountant
296
S. Andreychenko / Evaluation of Tumor Capabilities for Recurrence
(Fluka) the preparations were analyzed in two rays automatic cytophotometer "MFTX 2M" (Russia) linked with computer IBM-486 at the wavelength of 546 nm. The specially developed program made us possible to record exclusively total nuclear DNA specific absorption. For control measurements really diploid mucous cells and haploid spermatozoa from volunteers were analyzed to establish properly the correlation coefficient between DNA absorption and ploidy level. The aforesaid technique permitted to specify cell distribution in tissue populations according to the value of their ploidy. Estimation of tumor aggressiveness coefficient - A: It is calculated on the basis of proportion showing the relation of DNA concentration in tumor (cDNAT) to that in nearby normal on convention epithelial tissue (cDNAN ): A = cDNAT/ cDNAN Statistical analysis: For statistical evaluation of data Student's t-test was used at the level of significance not less than 0,95 when the coincidence between control and experimental results is observed only at a value of p < 0,05 [5]. Results Analysis of DNA concentration in tumors in patients with larynx malignancies revealed the diapason of its variability ranging from 1,15 to 1,89 ug/mg (p>0,99) while for pharynx this parameter has shown a tendency for some increase reaching upper limit of 2,72 ug/mg. In the nearby normal on convention tissues of larynx and pharynx the values for DNA contents were 0,95 ± 0,16 ug/mg and 1,04 ± 0,17 ug/mg, respectively (Table 1). In connection to these data a tumor aggressiveness coefficient A for larynx malignancies ranged in the interval 1,15-1,91 and was some few different from that for pharynx, a latter one showing a broader range of variability and a tendency for manifestation at a higher level of significance - 2,04 ± 0,59. Table 1. Analysis of DNA concentration in malignant tissue in comparison to unaffected one in patients with tumors of larynx and pharynx*
Localization
Larynx Pharynx
DNA concentration variability range in tissue, ug/mg Malignant (cDNAT) 1.15–1,89 1,31–2,72
Normal on convention (cDNAN) 0,79–1,11 0,87–1,21
Tumor aggressiveness coefficient A variability range 1,15-2,02 1,55–2,62
* Full variability range of experimental data for each group of patients is presented Each patient was examined as to the duration of post-operation recovery. According to the data presented in Fig.1 evident correlation exists between A and patient's resistance to tumor recurrence. Thus, several patients after larynx surgery manifested explicit stability in recovery for more than 4 years long with A ranging from 1,8 to 1,9. Meanwhile for pharynx the same result was achieved at values of A equal to 2,15±0,20.
S. Andreychenko / Evaluation of Tumor Capabilities for Recurrence
297
For larynx rapid abatment was observed of recovery duration at A values below 1,5 with further little rise at 1,15. The plotted two curves (see fig.1) both for larynx and pharynx show similarities having obvious peak with two descending branches beyond it, although larynx manifests additional ascendant branch at low values. In both cases A value growth causes the appearance of recovery beneficial interval then replaced by pronounced downfall in recovery duration.
6,0 5,5 5,0 4,5 4,0 3,5 3,0 2,5 2,0 1,5 1,0 0,5 1,0
1,2
1,4
1,6
1,8
2,0
2,2
2,4
2,6
A (units) Figure 1. Dependence of recovery duration on the DNA ratio coefficient A in oncology patients after tumor removal.
The cytophotometric analysis of nuclear DNA contents revealed that cell distributions in larynx and pharynx normal on convention tissues were in the range of 2n to 4n showing usual realization of cell cycle in overall cell population. Meanwhile under strong recovery conditions the subpopulations of autopolyploid cells, i.e. tetraploids and octaploids were discovered. Apart from this, the accumulation of aneuploid cell clones with degraded genome (<2n) also was recorded. This reflects the depression of immune system as well as the reduction of macrophage activities. These data agree well with the present in literature information concerning head and neck cancer [6]. The patients having a tendency for short-term recovery were characterized by the appearance of intermediate ploidy cell clones, thus manifesting an inclination for selective gene amplification. In the case of pharynx malignancies partial DNA increase took place both in diploid and tetraploid cells while for pharynx malignancies selective DNA replication only at the diploid level was recorded.
298
S. Andreychenko / Evaluation of Tumor Capabilities for Recurrence
Discussion - Conclusion The obtained data show the possibility to evaluate tumor aggressiveness in relation to postoperation recurrence on the basis of DNA criterion that presents itself a ratio equal to proportion of DNA concentration in tumor to that in nearby normal on convention epithelial tissue. A is easy to determine in patients undergone surgical intervention by extirpating samples of corresponding tissues under the operation for their next processing in DNA quantitative analysis. A correlates well with the duration of post-operation recovery. However it depends on the cancer primary localization. At the range of A values most beneficial for long recovery A reflects the appearance in target tissue special cell clones undergone genome polyploidization while partial DNA amplification used to be incident to short-term recovery state. In the former case whole DNA repeated duplication might lead to the increase of genome stability and safety, on the contrary, in the latter one the sequence of events caused cell genome disbalance and casual proliferation of deleterious oncogenes. In this connection the ascendant short branch on larynx curve (see fig. 1) could be explained by diminution of oncogene dose below the crucial limit at low A. Launching of extra DNA replication in target tissue might be due to lowering of extracellular pH, activation of H+/Na+ antiport and concomitant growth in Ca2+ influx into cells, the latter one being a potent promoter of cell proliferation [7,8,9]. Moreover, it is known that DNA synthesis counteracts pH lowering and provides for normalization of intracellular pH as well as immobilization of superfluous Ca2+ in newly synthesized chromatin motifs [10,11]. Thus, when driving autopolyploidization program cells can acquire stability to oncogenic transformation and persist in unfavorable environment. This tendency can be registered by coefficient A measurement. The application of A criterion in clinic let us avoid a number of complications in post-operation period and outline properly the strategy for rehabilitation of oncology patients with aforementioned cancer localizations.
References [1] Buntze J., Basecke S., Weinaug R., Glatzel M., Kuttner K., Frohlich D., (2000). Liposomal doxorubicin in the palliative treatment of head and neck cancer. In: Jahnke K. and Fischer M. (ed.), "4th European Congress of Oto-Rhino-Laryngology Head and Neck Surgery", Berlin (Germany), May 13–18, 2000. Monduzzi Editore, Bologna, Italy, vol.2, p. 1377–1381. [2] Silverberg R.G., DeLellis R.A., Frable W.J. "Principles and Practice of Surgical Pathology and Cytopathology". 3rd edn. New York, Churchill-Livingstone, (1997). [3] Burton K.A., Biochem. J. 62 (1956) 315. [4] Darlington S.D., La Cour A.F. "Handling of Chromosomes". 6* edn. Atomizdat, Moscow, Russia, (1980). [5] Rokitsky P.F. "Biological Statistics". Higher School, Minsk, Belorussia, (1973). [6] Hadden J.W., Int. J. Immunopharmacol. 19 (1997) 629. [7] Rogers M.S., Strehler E.E., (1996). In: Celio R.C. (ed.), " Guidebook to the Calcium - Binding Proteins",A Sambrook & Tooze Publication AT Oxford University Press, New York, U.S.A., p. 34 - 40. [8] Ruas M., Peters G., Biocliim. Biophys. Acta 1378 (1998) 115. [9] Lee A.S., Tannock I.F, Cancer Research. 58 (1998) 1901. [10] L'Allemain P., Paris S., Poussegur J. J. Biol. Chem. 260 (1985) 4877. [11] Lewin B. "Genes", Oxford Univ. Press, New York, U.S.A. (1997)
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Significance of G-protein-coupled receptor polymorphisms : The case of a2-adrenergic receptor A.S. Manolis1, A. Lymperopoulos, E.A. Bouga, M. Scheinin2 and CS. Flordellis Department of Pharmacology and1 Department of Cardiology, School of Medicine, University of Patras, 261 10 Rion, Patras Greece and 2Department of Pharmacology and Clinical Pharmacology, University of Turku, Finland Abstract. Until recently it was assumed that G-protein coupled receptors ( GPCRs ) were genetically invariant in the human population. This assumption has been challenged by the growing evidence demonstrating the existence of frequently occurring genetic variants of GPCRs. Using a2-adrenergic receptor subtypes as example, this work summarizes briefly current knowledge on the issue and describes the clinical and pharmacological significance of these polymorphisms in cardiovascular disease.
Polymorphism is defined as the occurrence in the same locality of two or more discontinuous genetically determined phenotypes in such proportions that the rarest of them cannot be maintained merely by recurrent mutations [1]. Simply stated, genetic polymorphisms are common genetic variants (allele frequency > 2%). In a medical context the advantage of dealing with genetic polymorphisms is that associations between genotype and phenotype can be expressed in terms of frequency. Their importance consists in the fact that such variations in receptors , drug metabolizing enzymes, transporters and other potential targets of drug interventions , in many cases entail functional consequences, which may provide the basis for observed inter-individual variability in clinical phenotypes and pharmacological responses [2]. The G-protein-coupled receptors ( GPCRs ) represent a very suitable system for investigating the potential contribution of receptor polymorphism to propensity for disease and response to drug therapy. These receptors are widely expressed and mediate the action of a variety of hormones, growth factors, neurotransmitters , peptide and local mediators involved in numerous cellular processes and consequently physiological functions, such as metabolism, neurotransmission and cardiovascular regulation among others. Their clinical importance is underlined by the fact that almost one third of all subscription medications are GPCR agonists or antagonists [3].
300
A.S. Manolis et al. / Significance of G-protein-coupled Receptor Polymorphisms
GPCRs are nearly devoid of intrinsic activity. Upon agonist stimulation they only catalyze the release from the Ga subunit of the tightly bound GDP , driving the heterotrimeric Gprotein into the process of G-protein GTPase cycle . The generated Ga and G (Jy subunits regulate downstream effectors, such as adenylate cyclase [4], PI3 Kinase [5], and MAPK ([6] among others. In addition , the Cfty complex activates GRKs, Ser/Thr kinases, which phosphorylate the agonist-bound receptor, thus initiating the process of receptor desensitization. The way genetic polymorphisms may modify the receptor's biochemical phenotype, becomes evident when considering the functional domains of GPCRs. Seven transmembrane receptors in general and alpha2-adrenergic receptors (ct2-ARs ) in particular have an extracellular aminoterminus, usually glycosylated , an intracellular carboxyl terminus and a peptide backbone that folds back and forth through the plasma membrane seven times forming the ligand-binding pocket (in the transmembrane domains ) and loops, notably the third cytoplasmatic loop, which has domains both for coupling to the cognate G-protein and for phosphorylation by G protein-coupled Receptor Kinases ( GRKs ) and subsequent desensitization [7]. In the case of <x2-ARs there is subtype-specific agonistpromoted phosphorylation and desensitization : a2A and tt2B undergo agonist-promoted phosphorylation and desensitization,whereas the o^c receptor does not, despite the fact that the aac has within the 3rd intracellular loop the sequence DESS, which is similar to the a2A sequence EESSS, shown to be an excellent substrate for GRK-induced phosphorylation [8]. Mutations of G-protein coupled receptors: They can cause a number of relatively rare diseases. In the majority of mutations that modify receptor function, the variant allele imparts constitutive activation (agonist -independent increased function) or decreased agonist-promoted function. Examples of the former are the familial male precocious puberty ( FMPP ), that results from a mutation in the luteinizing hormone receptor [9] and the hyperfunctioning thyroid adenomas due to somatic or germline mutation of the Gs-coupled TSH receptor [10]. Polymorphisms of GPCRs on the other can be minor risk factors for complex multifactorial diseases ( e.g cardiovascular disease, diabetes mellitus etc. ), modifiers of disease characteristics ( onset, progression or outcome ) or alter responses to drug therapies. Many of them have been described, but in the present context we mention representatively only some of them. The Gly16 form of the pVadrenergic receptor becomes more easily down regulated and desensitized after exposure to the agonist, compared with the wild-type Arg16 form. This genetic variant may predispose to some forms of asthma and hypertension in AfricanCaribbeans [11]. On the contrary the Glu27form of the receptor confers resistance to down regulation compared with the wild-type Gln27 form [12]. Consequently subjects with this variant receptor are predicted to have less reactive airways. Finally, the Deu164 pY adrenergic receptor polymorphism, displays a small reduction in agonist binding affinity and defective agonist-promoted coupling to Gs and has been found to affect adversely the outcome in congestive heart failure [13]. A mutation has been identified at the beginning of the first intracellular loop of (J 3adrenergic receptor resulting in a Trp64Arg substitution. The 0 3-AR is expressed mainly in brown fat adipocytes and impairement of its function could reduce adipocyte sensitivity to lipolysis. The allele frequency of the above variant was found similar in normal subjects and in patients with morbid obesity ( BMI > 40 kg*/m2 ) ( 0.1 vs. 0.08 respectively ), implying that the allele is not the cause of obesity. The presence of the allele may
A.S. Manolis et al. / Significance of G -protein-coupled Receptor Polymorphisms
301
predispose patients to abdominal obesity, decreased resting metabolic rate, insulin resistance and earlier onset of NIDDM ([14]. In recent studies several polymorphisms have been identified in the coding or the 5' and the 3' flanking regions of the ATl receptor for AngII [15]. The polymorphism A1166C ( an Adenine to Cytosine base substitution at position 1166, located in the 3' untranslated region of the ATl receptor gene), was recently found to be associated with myocardial infarction and essential hypertension. The C allele of this polymorphism is an independent determinant of aortic stiffness [16] and also shows a synergism with the D allele of the angiotensin converting enzyme insertion/deletion polymorphism on risk of myocardial infarction. A role of the renin-angiotensin system in the aetiology of coronary atherosclerosis has been indicated by the finding that the DD genotype of an insertion/deletion polymorphism (I/D) in the gene for the Angiotensin Converting Enzyme ( ACE ) is strongly associated significantly with the levels of circulating enzyme and is more frequent in patients with myocardial infarction [17]. The genetic analysis of aa-ARs is limited. Initial studies on small populations have indicated that all cta-AR genes are polymorphic, and because the mutations occur in the 3rd cytoplasmatic loop the variant alleles have altered biochemical properties in relation to the wild-type genes. The Del322-325 form of the human aac-adrenergic receptor results in impaired coupling'to multiple effectors [18]. In other cases however, the polymorphism confers enhanced agonist-promoted G-protein coupling. Thus, the Asn to Lys at position 251 in the third cytoplasmatic loop of the human aaA-AR imparts enhanced G-protein coupling [18]. Finally, a common genetic variant of ct2B- adrenergic receptor was identified , which consists in a deletion of three glutamic acid residues in the third cytoplasmatic loop [19]. This deletion results in reduced ( ~60 % ) agonist-promoted phosphorylation of the variant receptor relative to the wild-type receptor and in loss of short-term agonistpromoted desensitization and is associated with reduced metabolic rate in obese subjects of a Finnish population [19]. The clinical and pharmacological consequences of the 012-AR genetic variation can be predicted, if one considers the physiological function of the respective receptor subtype and the kind of biochemical modification resulting from the mutation. It has been anticipated for example, that the above mentioned (X2A-AR polymorphism would result in enhanced inhibition of depolarization-induced norepinephrine release and therefore predispose to hypotension and bradycardia and display a milder course of disease, in case of development of hypertension. For the same reason the polymorphism would result in potential protection of the myocardium from catecholamines in congestive heart failure. Similarly, the antihypertensive efficacy of (X2-AR agonists i.e. clonidine would be enhanced, whereas the central nervous system side-effects of such pharmacological agents would be pronounced limiting their therapeutic utility [18]. Role of alpha2-ARs in cardiovascular regulation: Three distinct and well characterized tt2AR subtypes ( v,2\, 0.23, «2C) have been described. They belong to the family of G-protein coupled receptors and they are linked to the inhibitory Gi/o signalling system modifying the activity of several effectors including adelylate cyclase, ion channels , MAPK and cPLA2 [20] among others. They differ in their structure, chromosomal localization tissue distribution and pharmacological properties and participate in the regulation of cardiovascular system [7]. Despite progress in dissecting the function of alpha2-ARs as a group, knowledge on the physiological roles of the individual a2-AR subtypes was very limited until recently due to the lack of subtype-selective antagonists. However, In the last five years, knock-out (KO) mouse lines generated by the employment of homologous recombination methodologies [21, 22] have allowed determination of the physiologic roles of the (X2-AR
302
A.S. Manolis et al. / Significance of G-protein-coupled Receptor Polymorphisms
subtypes in mice. Studies on such mice have indicated that central a2A-ARs mediate the long-known hypotensive effects of 02-AR agonists, such as clonidine [7], whereas 02adrenergic vasoconstriction is mainly mediated by postsynaptic or extrajunctional aie-ARs on vascular smooth muscle cells. Both <X2A- and c^c-AR mediate presynaptic inhibition of neurotransmitter release [23]. tt2-ARs exert important functions in renal physiology. It has been shown that ct2B-ARs participate in the development of salt-induced experimental hypertension and mice lacking the full complement of a2B receptor fail to develop effectively salt-induced hypertension [24]. Studying genetic polymorphisms of d2-ARs: Cardiovascular disease is a multifactorial disease that results apparently from the synergism of several genes interacting in a complex way with environmental factors. Recent clinical genetic studies have shown significant associations between a 2B-AR gene variant and basal metabolic rate and the risk for acute coronary events [19, 25]. We have been interested in the a2B-adrenergic receptor polymorphism and have started to study its association with cardiovascular disease and characterize the biochemical properties of the receptor in order to find : the clinical association of this alpha2 receptor polymorphism with coronary heart disease phenotypes in populations of Greek patients and the mechanisms by which genetic polymorphisms contribute to the development of cardiovascular diseases Determination of genotype: The study population used for genotype determination and association analysis consists of patients hospitalized for ischemic coronary events (unstable angina or AMI ) evaluated and classified in the Cardiology Department of the University Hospital, Patras-Greece. The a 2B-AR insertion/deletion genotypes are determined by electrophoretic separation of PCR-amplified DNA fragments. The size difference of the long and short alleles of this a 2B-AR polymorphism makes it possible to reliably and reproducibly distinguish them by their different electrophoretic migration on high density agarose gels [25]. To confirm the reliability of agarose gel electrophoresis in determining the genotype some of the DNA samples in our study were also analyzed by PAGE. A representative example of such analysis is shown in figure 1. So far we have analyzed 70 patients and in some cases their normal relatives. The frequencies for the I and D alleles in this population appears to be 0.8 and 0.2 respectively.
Figure 1. Determination of genotype by electrophoretic separation of PCR-amplified DNA fragments.
DNA for genotyping was axtracted from a small quantity of peripheral blood according to standard methods and amplified with the following pair of primers : 5'-CAA GCT GAG GCC GGA GAC ACT G-3' and 5'- AGG GTG TTT GTG GGG CAT CTC CT-3' yielding a DNA product of 112 bp for the long allele ( I ) and a DNA product of 103 bp for the short, deleted allele ( D ). The PCR reaction contained ~100 ng genomic DNA, 0.8 mM dNTPs, 0.3 nM of each primer and 0.5 units of Taq polymerase in a total volume of 20 ul. The conditions included an initial denaturation step at 94 oC for 2 minutes and 35 cycles of amplification as follows : 96 oC ( 40 sec ), 69 oC ( 30 sec ) and 72 oC ( 30 sec ) followed
A.S. Manolis et al. / Significance of G-protein-coupled Receptor Polymorphisms
303
by a final extension step at 72 oC for 6 minutes. After amplification the PCR products were separated electrophoretically on a 4% agarose gel with ethidium bromide staining. The long and short alleles were distinguished on agarose gels ( left panel ) based on their different electrophoretic migration rate as a result of their 9 bp size difference. The analytical power of this agarose gel system was verified by parallel analysis of PCR products in polyacrylamide gels ( right panel). Desensitization properties of polymorphic a.2B-adrenergic receptor Previously, it has been shown that an acidic stretch of amino acids in the 3rd cytoplasmatic loop of a 2B-adrenergic receptor regulates phosphorylation of the receptor by GRKs and experimentally induced deletion mutation in this region, is accompanied by reduced agonist-promoted phoshorylation and subsequent desensitization of the receptor [26]. In order to analyze the consequences of this deletion variant on short-term agonist-promoted receptor phosphorylation, we use Chinese Hamster Ovary ( CHO ) cells stably expressing the wild-type and the deleted form of a2B-AR. We have standardized the conditions for monitoring the state of receptor phosphorylation using a specific anti-human aas-AR monoclonal antibody (generously provided from the Turku laboratory ) to immunoprecipitate the receptor, after whole-cell phosphorylation. A representative result is shown in figure 2.
Figure 2. Detection of a2B-AR expressed in CHO cells. ( 1 ) Confluent cultures of CHO cells expessing the wild-type a2B-AR were solublized in RIPA buffer and the receptor was immunoprecipitated with a specific monoclonal anti-human a2u-AR , followed by Western blotting and visualization of the receptor using the same antibody and ECL detection. ( 2 ) Confluent cultures of CHO cells expessing the wild-type O2B-AR were incubated with [32P] orthophosphate ( 400uCi/ml) for 2h at 37oC, washed with ice-cold PBS 5 times, solublized in RIPA buffer and the receptor was immunoprecipitated with a specific monoclonal anti-human a2e-AR, followed by SDS-PAGE and autoradiography.
These genetic and biochemical studies are currently in progress. The rapid progress in functional genomics is expected to have a profound impact in the medical-pharmaceutical system. Detailed analysis of the genetic mechanisms of disease will clarify our understanding of multigenic diseases, will facilitate the assessment of individual risk for disease and will aid in the design of novel approaches to disease prevention and drug therapies. Pharmacogenomics promises the translation of functional genomics into rational drug therapy. Genetic tests for more rapid diagnosis will help classifying heterogenous populations of patient into genetically distinct groups. Genotyping before prescription of certain drugs will enable to predict how a patient will respond to a given drug. This will
304
allow on the one hand selection of appropriate therapies, from which patients are going to be benefited the most and on the other avoidance of the prescription of potentially toxic drugs, that are likely to yield dangerous side-effects. On the other side, the utilization of flexible and time-saving assays ( e.g. gene expression monitoring ) for predicting pharmacological and toxicological outcomes, will decrease the cost of clinical drug testing and may allow companies to develop drugs for small markets such as limited, genetically defined subpopulations and patients in poor countries. Furthermore, the new knowledge will facilitate the drug companies to sharpen the properties of their drugs, so that drug intervention is expected to become more individualized. These starting shifts in the existing paradigm of health care and drug development show that the medical-pharmaceutical system will be further challenged in the coming years. References [1] Ford EB. Ecological Genetics, 3rd edition, London Chapman & Hall. 1971. [2] Insel PA., Seminars in medicine of the Beth Israel Hospital, Boston. New Engl. J.Med. 334 (19%) 580. [3] Kallal L, Benovic JL, Trends Pharm.Sci. 21 (2000) 175. [4] Schaak S, Cayla C, Lymperopoulos A, Flordellis C, Cussac D, Denis C, Paris H., Mol. Pharmacology 58 (2000) 821. [5] Hawes BE, Luttrell LM, van Biesen T, Lefkowitz RJ., J.BioiChem. 271 (19%) 12133. [6] Flordellis CS, Berguerand M, Gouache P, Barbu V, Gavras H, Handy DE, Bereziat G, Masliah J., J.Biol.Chem. (1995) 3491. [7] MacDonald E, Kobilka BK, Scheinin M, Trends Pharmacol.Sci. 18 (1997) 211. [8] Jewell-Motz EA, Small KM, Theiss CT, Liggett SB., J.Biol. Chem. 275 (2000), 28989. [9] Shenker A, Laue L, Kosugi S, Merendino JJ Jr, Minegishi T, Cutler GB Jr., Nature 365 (1993 ) 652. [10] Parma J, Duprez L, Van Sande J, Cochaux P, Gervy C, Mockel J, Dumont J, Vassart G. , Nature 365, 649. [11] Reishaus et al. (1993 ) Am.J.Respir.Cell.Mol.Biol. 8 (1993) 334. [12] Green SA, Turki J, Bejarano P, Hall IP, Liggett SB., Am.J.Respir.Cell.Mol.Biol. 13 (1995) 25. [13] Liggett SB, Wagoner LE, Craft LL, Hornung RW, Hoit BD, Mclntosh TC, Walsh RA., J.Clin.Investigation 102 (1998) 1534. [14] Walston J, Silver K, Bogardus C, Knowler WC, Celi FS, Austin S, Manning B, Strosberg AD, Stem MP, Raben N, et al., New Engl. J Med. 333 (1995) 343. [15] Bonnardeaux A, Davies E, Jeunemaitre X, Fery I, Charm A, Clauser E, Tiret L, Cambien F, Corvol P, Soubrier F., Hypertension 24 (1994) 63. [16] Benetos A, Gautier S, Ricard S, Topouchian J, Asmar R, Poirier O, Larosa E, Guize L, Safar M, Soubrier F, Cambien F., Circulation (1996) 698. [17] Cambien F, Poirier O, Lecerf L, Evans A, Cambou JP, Arveiler D, Luc G, Bard JM, Bara L, Ricard S, et al. Nature 359 (1992) 641. [18] Small KM, Forbes SL, Rahman FF, Bridges KM, Liggett SB., J.Biol. Chem. (2000) 23059. [19] Heinonen P, Koulu M, Pesonen U, Karvonen MK, Rissanen A, Laakso M, Valve R, Uusitupa M, Scheinin M., J. Clin.Endocrinology & Metabolism, 84 (1999) 2429. [20] Audubert F, Klapisz E, Berguerand M, Gouache P, Jouniaux AM, Bereziat G, Masliah J., Bioch.Bioph.Acta 1437 (1999) 265. [21] Link RE, Desai K, Hein L, Stevens ME, Chruscinski A, Bernstein D, Barsh GS, Kobilka BK., Science 273 (1996) 803. [22] MacMillan LB, Hein L, Smith MS, Piascik MT, Limbird LE, Science 273 (1996) 801. [23] Hein L, Altman JD, Kobilka BK., Nature 402 (1999) 181. [24] Makaritsis KP, Handy DE, Johns C, Kobilka B, Gavras I, Gavras H., Hypertension 33 (1999) 14. [25] Snapir A, Heinonen P, Tuomainen TP, Alhopuro P, Karvonen MK, Lakka TA, Nyyssonen K, Salonen R, Kauhanen J, Valkonen VP, Pesonen U, Koulu M, Scheinin M, Salonen JT., J. Am.Coll. Cardiol. 37 (2001) 1516. [26] Jewell-Motz EA, Liggett SB., Biochemistry 34 (1995) 11946.
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
305
Computer Graphics Applications on Molecular Biology and Drug Design K. Perdikuri1, A. Tsakalidis1 1
Department of Computer Engineering and Informatics, University of Patras, 26500 Patras, Greece
Abstract. Molecular structure-based drug design is an art and a science. As graphics hardware has matured and software continues to mature, the applications of computer algorithms on molecular biology provides a rigorous basis for many drug design efforts. In this paper we try to present the basic principles in molecular graphics as long as it concerns molecular representations and modelling, and outline some important challenges and open problems in the field of computer aided drug design.
Introduction During the last 20 years, the process of drug discovery and drug design has been enormously affected from the increased available information, concerning 3D structures of biomolecules, determined by X-ray crystallography and NMR structure determination techniques. Exploiting this structural information, scientists can design novel pharmaceutical molecules (often referred to as ligands), which bind tightly and selectively to a target macromolecule, such as a protein (often referred to as receptor). The binding of a ligand to a specified receptor is based on the "lock-and-key" principle, which was firstly recognized by Emil Fischer more than 100 years ago. Along with this principle, the active site of a receptor binds with a ligand both spatially and chemically, just as only a specific key fits a given lock. The design of pharmaceuticals molecules based on this principle is usually referred to as structure-based drug design [1]. Modern methods for structure-based drug design can be divided into two categories. The first category, which is usually referred as Database Searching, searches ligands for a given receptor, through large-scale screens in databases of known 3D molecules. The goal of the search is to retrieve molecules, with shape and chemical complementarity to a given active site of a receptor. The second category of structurebased drug design, which is usually referred as de novo design, "builds" gradually the proper ligands [2]. In this case, ligands, are built up within the constraints (both geometrical
306
K. Perdikuri and A. Tsakalidis / Computer Graphics Applications
and chemical) of the binding receptor, by assembling atoms or small fragments in a stepwise manner. A number of such approaches have already been reported. Several ligands designed in this manner are now in clinical trials. The success of structure-based drug design has encouraged the development of various computational methods that can combine structural information and modem molecular graphics techniques to suggest novel structures, which may prove to be useful lead compounds. Ideally, these methods should be fast, objective and produce a set of diverse yet chemically reasonable structures. In this paper we will present and analyze the current methods used and the problems encountered in the field of Molecular Graphics and especially in the field of structure-based drug design.
Figure 1. A 3D molecule represented with ball and sticks Molecular Representations
Molecular Graphics deals with the representation and manipulation of biological molecules in a computer. It is a newly established scientific field encompassing the theoretical and application areas of computer science that deal with geometry and visualization. Among these areas are computer graphics, computer animation, computer vision, computational geometry and computer-aided geometric design. In this section we will try to introduce some definitions for the concept of molecular surface and molecular volume. The three-dimensional geometric structure of a molecule is often represented as a set of atoms (Figure 1). In this representation each atom is modeled as a "hard" sphere with a certain atomic radius. The three-dimensional placement of the spheres is expressed according to the internal distances between the centers of each pair of atoms (interatomic distances) and their relative angles (Table 1). The equilibrium configuration of this representation is defined as the "low energy conformation". In many representations, the spheres are allowed to interpenetrate one another. In structure-based drug design, one wishes to manipulate a molecule in the presence of another molecule to see whether they fit together. During such manipulation the molecules should have only limited interpenetration. Table 1. Table with internal coordinates of C2H6
1
C
2
C
1.54
1
3
H
1,0
1
109,5
2
4
H
1,0
2
109,5
1
180,0
3
5
H
1,0
1
109,5
2
60,0
4
6
H
1,0
2
109,5
1
-60,0
5
7
H
1,0
1
109,5
2
180,0
6
8
H
1,0
2
109,5
1
60,0
7
K. Perdikuri and A. Tsakalidis / Computer Graphics Applications
307
Molecular Surfaces The determination of the molecular surface of a collection of atom spheres of a molecule is of great importance in various applications in molecular biology for the interpretation of molecular properties, interactions and processes. Molecular surfaces are classified as contact surfaces and re-entrant surfaces. Various definitions have been proposed in the relative bibliography. The van der Waals surface (Figure 2), Lee and Richards' solvent accessible surface [4], and Richards' smooth molecular surface. In Lee and Richard's approach, a sphere is rolled on a reference surface, to obtain a new surface described by the center of the rolling sphere, which is assumed to be fairly small. In a simplified approach the molecular surface of a molecule can be described as the accessible parts of the modeled spheres (assuming that the molecules are modeled by the hard sphere model), or as the boundary of the union of the balls in the hard sphere model. Molecular Volumes A critical issue in most 3D molecular graphics packages is the choice of the model for representing, the three dimensional structure of a molecule. Obviously the molecular geometry is affected from the selected model. Generally the use of any model means that a significant level of detail is lost as molecules are visualized to have surfaces and volumes similar to our perception of surfaces and volumes of macroscopic objects. Although it is clear that the more accurate the model used, the better are the chances in predicting molecular interactions certain abstractions need to be made in order to calculate molecular interactions efficiently. The sphere model is the most popular model for representing approximately the volume of a molecule. A sphere is drawn around the centre of every atom of the molecule. The radius of each sphere reflects the space requirements of the corresponding atom and consequently the total volume of the molecule (the used radii have been determined by a combination of experimental observations and quantum mechanical calculations). Sticks models are also used to represent the bonds between two consecutive atoms and the angles they form in 2 dimensions. The bond lengths and bond angles are degrees of freedom (DOF) for each molecule. This way a conformation of a molecule is obtained by assigning values to all the DOF of the molecule.
Figure 2. Representation of Van der Waals surfaces
Color Coding and Texture Mapping Techniques The molecular surface concept is not only useful for a representation of the shape of molecules. These surfaces can be used as screens for the visualization of arbitrary properties using color coding techniques. Color coding is a popular means of displaying scalar information on a surface [3]. In interactive molecular graphics, high contrast color code variation can be realized by using texture mapping techniques which are available on
308
K. Perdikuri and A. Tsakalidis / Computer Graphics Applications
graphical workstations and PC's to represent a color ramp as a ID texture. Texture mapping is a technique that applies an image to an object's surface. Conformational Analysis Having represented the surface and volume of a molecule the next step in molecular modeling is to find the global energy minimum conformation of a molecule, regardless of the size of the molecule under consideration. This procedure is known as conformational analysis. A variety of conformational analysis methods exist and fall into two classes: deterministic and stochastic. Deterministic methods search all the rotatable bonds in a molecule. The time required for this type of search increases exponentially to the number of rotatable bonds. The success of a deterministic method relies on the selected granularity, which is difficult to define. For example it may be possible to perform a complete search if one limits the number of rotamers per bond to a small number. On the other hand the same search may become computationally infeasible if the number of scanned rotamers increases. Stochastic methods are based on algorithms that limit the conformational search space to the lowest energy conformations of a molecule. Some of the stochastic methods use molecular dynamics, Monte Carlo, genetic algorithms, random perturbations to the coordinates, or a combination in order to find and optimize local minima [9]. Unfortunately stochastic methods are not guaranteed to converge to the same set of low energy conformations as produced with a deterministic method. A slight deviation in a torsion angle may miss the lowest energy state, although we are very close to structurally to the global minimum. Open Challenges and Problems in Molecular Graphics Studying a hard sphere model from a computational geometry point of view, we have to take into consideration several properties such as: (i) the radii ranges in a fairly restricted area, and (ii) sphere centers cannot get too close to one another. One of the tasks of molecular modeling packages is to display molecules. This should preferably be done so fast that the user can interact with the model by turning it around to look at it from different directions or by moving different molecules with respect to each other, to see for example whether they fit together. In this paragraph we will refer to problems and challenges that arise in the field of molecular graphics and are crucial for application in computer-aided structure-based drug design. Reconstructing a 3D Model The reconstruction of a three-dimensional set of points using information about its inter-point distances is a task of great importance in determining molecular structure. In particular, spectroscopic methods such as two-dimensional NMR provide a mean for determining a labeled subset of the distances between atoms in large structures such as proteins and RNA, and such methods have therefore proved extremely valuable in conformational studies of such molecules. Determining the structure of n labeled points in R3 is easy if the exact value of every inter-point distance is given. However, this is far from what one obtains using NMR and its variants. In particular, not all distances can be measured, and the distance values that one does obtain are susceptible to a wide variety of sources of error.
K. Perdikuri and A. Tsakalidis / Computer Graphics Applications
309
NMR experiments are frequently based on measuring proton resonances, since hydrogen atoms are easily detected by these techniques; this is in general sufficient for conformational studies, since hydrogen atoms are abundant in organic molecules. To determine spatial information, one typically requires two NMR experiments: one based on spin-spin coupling to determine protons that are close in the covalent geometry of the molecule (COSY, Correlated Spectroscopy), and another to determine distance information for pairs of protons that are close in space but not necessarily close in the covalent structure (NOESY, Nuclear Overhauser Enhancement Spectroscopy). Combining information from these two types of experiments, one obtains labeled interatomic distance data- more precisely which distance corresponds to which pair of atoms. The nuclear Overhauser Effect (NOE), on which the latter technique is based, is manifested in cross-peaks in an NMR spectrum that arise from dipole- dipole coupling of one proton with nearby protons. NOE intensity is given by a formula with a term for dij-6, where dij is the Euclidean distance between two protons. If one has approximate knowledge of the other variables involved, then it is feasible to recover a value for dij. Distance calculations based on NOESY are subject to numerous sources of systematic error. Some basic examples are the following: i) Spin-diffusion is a phenomenon whereby NOE effects between protons i and j are in some sense "transferred" through a third proton k, resulting in spurious cross-peaks. These effects often result in a compression of the apparent distances derived from transferred NOE experiments. ii) For reasons of experimental efficiency, the relaxation delays between successive NMR scans are typically too short to allow for full recovery of the magnetization effects between nearby protons This is particularly a problem in RNA structure determination, where proton relaxation times are quite long and vary considerably [8]. iii) As the NMR spectrum becomes more and more dense, it is possible for two peaks to essentially coincide, and thus for one or more of these peaks to escape detection when the data is interpreted. This is a problem that becomes increasingly significant precisely when more of the inter-point distances are measurable. iv) As noted above, in order to produce labeled distance data from a NOESY experiment- in other words to determine that a given distance d is in fact associated with protons i and j— one must solve a complex assignment problem using information from an accompanying COSY experiment. Several error-correcting algorithms have been proposed in order to deal with arbitrary errors in distance measurements and reconstruct an accurate 3D model. Most of them belong in the area of distance geometry and use general model-building tool. Hidden Surface Removal Hidden surface raises a difficult problem among sets of intersecting spheres. In practice most 3D graphics workstations available nowadays, use a Z-buffer algorithm either in software or in their graphics hardware. Unfortunately, such an implementation can only handle polyhedral objects, which means that triangular meshes can approximate spheres. To get a reasonable approximation for a sphere one needs more than 100 triangles and these implementations lead to a large number of needed triangles in 3 dimensional space in order to model a molecule. Another common approach to hidden surface removal used in computer graphics is the painter's algorithm. Here one tries to define a depth order on the objects, sorting them from back to front. Next one draws the objects in this order on top of each other where each new object hides the parts of other objects that lie below it. Such an approach does not require special graphics hardware. The problem with this approach is that it requires a valid depth order on the objects. Such an order does not always exist, as in the case of intersecting objects. In such applications the combinatorial representation of visible pieces,
310
K. Perdikuri and A. Tsakalidis / Computer Graphics Applications
can be seen in the visibility map. The visibility map can be defined as the subdivision of the viewing plane into maximal connected regions in each of which a single object is seen, or no object is seen. Modelling Water What is important in a software package is the ability to simulate the presence of water molecules, in order to understand the effects of water on the shapes of biological molecules. Taking into consideration when building a cell that it contains billions of water molecules and the space not occupied by the atoms of biological molecules is filled with water, it is clear why water plays an important role in molecular interactions. A single water molecule (H2O) has a tetrahedral geometry, which gives water a loosely packed structure compared with that of most other liquids, such as oils. To construct a computer aided model of water, we need to take into account two different types of forces: intramolecular and intermolecular. Moreover there are two types of waters that must be considered when building a computer model of a biological molecule in aqueous solution: the "ordered waters" that surround and strongly interact with the molecule and the "bulk waters" that may be buried within the molecule [6]. Subsequent simulations of DNA in water have revealed that water molecules are able to interact with nearly every part of DNA double helix, including the base pairs that constitute the genetic code. In other words water help us to reveal the structure of several biomolecules. In contrast, water is not able to penetrate deeply into the structures of proteins. Experimental results have shown that water profoundly influences the interactions of proteins and DNA. The polarity and hydrogen-bonding capability of water make it a highly interacting molecule. Water can greatly weaken the electrostatic forces and H-bonds between polar molecules by competing for their attraction, play both roles of an H-bond donor and, by lending electron pairs of the oxygen atom, an acceptor, and form a variety of bridges between molecular donors and acceptors [7]. Molecular Docking One important application in the field of computer aided molecular modeling is related to the molecular docking process. An effective and interactive handling of the molecular complementarity is needed in order to investigate possible docking partners. Hidden surface problem is highly related to the protein docking process. Although the mechanisms of docking reactions are not well understood, two complementarity principles seem to be important for the recognition and binding of docking partners. The first principle is the shape complementarity principle: the shapes of the molecules that build a docking complex are complementary, that is, there is a large fit between the surfaces of the docking partners. The second complementarity principle is the chemistry principle: there is a strong chemical complementarity (with respect to hydrogen bonds, electrostatic interactions, hydrophobicity and so on), between the sites of docking partners. Although the second principle is the more important one, it is possibly to identify many docking sites solely with the help of the shape complementarity principle. In order to find these sites for two molecules A, B with n and m atoms, the following 3D. matching problem has to be solved: determine all transformations of B such that there is a large fit between the surface of A and the surface of B, and no penetration of B into the interior of A. For all the candidates with a good geometric fit the potential energy function of the docking conformation and the molecules A and B has to be computed. In the above description of the geometric 3D matching problem two strong assumptions were made. 1)
K. Perdikuri and A. Tsakalidis / Computer Graphics Applications
311
the molecules are considered rigid and 2) there is no penetration. Of course not all molecules are rigid and certain parts of the molecules are very flexible influencing their chemical activity. More over local changes of the shape of the docking sites happen during the docking reactions. So with the structure of even one target protein, and the knowledge of function of its receptor or active site, it is now possible to use computer tools to build and dock a ligand or inhibitor ("new leads") prior to investing time and resources for synthesis and testing. Thus molecular modeling is essential for understanding and exploring the structure-function relationship and evaluating novel compounds before being synthesized. Based on Fischer's "lock and key" principle, the mechanical view of molecular interactions can be easily understood and applied to biomolecules. However, even "rigid" molecules have local flexibility, as previously mentioned, and water molecules as previously described are usually a structural appendage of both the "lock" and the "key," which means the in vivo structure may differ significantly from that on the display screen. Conclusion Researchers in the areas of computer graphics, molecular biology and computational chemistry have long studied representations of molecular models as well as other computational problems related to the geometry of molecules. Current trends incorporate tools from computational geometry in order to provide algorithms that are provably efficient and at the same moment fast in practice. Taking as an input the information determined by X-ray crystallography and NMR structure determination techniques, scientists can move from sequence level to structure level, which is an important step for understanding the molecular details of biological processes. Various fields start to develop like genome modeling and annotation; comparative protein modeling and fold assignment; in silica drug design and modeling of cellular processes. Acknowledgments We would like to express our thanks to Prof. J. Matsoukas and T. Mavromoustakos for their support and useful guidelines. This work was partially supported by the General Secretariat of Research and Technology of Greece during the research project EPET II/ EKBAN 115. References [ 1 ] D. Kuntz, Science 257 (1992) 1078. [2] R. Wang, Y. Gao, L. Lai, LigBuilder, J. Mol. Modeling 6 (2000) 498. [3] J. Brickmann, T. Exner, M. Keil, R. Marhofer, J.Mol.Modeling, 6 (2000) 328. [4] F. Richards, Ann.Rev.Biophys.Bioeng 6 (1977) 151. [5] F. Richards, Methods in Enzymology 115 (1985) 440. [6] M. Gernstein, M. Levitt, Scientific American (November 1998). [7] Y. Deng, J. Glimm, Y. Wang, A. Korobka, M. Eisenberg, A. Grollman, J.Mol.Modeling 5 (1999) 125. [8] B. Berger, J. Kleinberg, T. Leighton, Journal of the ACM 46 (1999) 212. [9] Spellmeyer, D. Wong, A. Bower, M. Blaney, J., J.Mol.Gmphics Mod., Vol. 15, 1997. [10] H. Lenhof, An algorithm for the protein docking problem. From Nucleic Acid and Proteins to Cell Metabolism, D.Schomburg and U.Lessel (ed),1995 125-139.
312
Drug Discovery and Design: Medical Aspects J. Matsoukas and T. Mavromoustakos (Eds.) IOS Press, 2002
Investigation of Novel DNA Gyrase Inhibitors Using the High Resolution NMR Spectroscopy S.G. Grdadolnik, M. Oblak, T. Solmajer and R. Jerala Laboratory for Molecular Modeling and NMR Spectroscopy, National Institute of Chemistry, POB 660, Hajdrihova 19, 1001, Ljubljana, Slovenia
Abstract. DNA gyrase is a well-established antibacterial target, because it is restricted to prokaryotic organisms. Novel DNA gyrase inhibitors are still searched in order to overcome the limitations of the known inhibitors connected with resistance and cytotoxicity. In our efforts to find new lead compounds, the high resolution NMR Spectroscopy has been utilized, which has recently become a high-throughput screening method. The binding of new compound has been determined by observation of chemical shift changes of resonances of the 24 kDa N-terminal fragment of gyrase B, which contains the entire ATP binding site. The resonances of the protein have been sequentially assigned by application of the threedimensional heteronuclear correlation methods in order to identify the interactions of inhibitor with the protein binding site. The location and orientation of the new molecule in the protein binding site has been studied by molecular modeling methods and analysis of chemical shift differences.
Introduction High resolution NMR Spectroscopy is a powerful technique for studying the conformational and dynamic properties of drug molecules. The information derived from the conformational analysis studies aids in rationalizing the drug design of new synthetic molecules with better physicochemical properties. However, the relationship between bioactivity and conformation is indirect. In addition, conformational analysis in general follows the synthesis of novel drug molecules. Recently, NMR Spectroscopy has been applied very efficiently at an earlier stage of drug research, in screening for novel ligands (1-4). In particular, its ability in identification of weakly binding ligands has been utilized, which offers the identification of binding sites at unique resolution. Systems of proteins
S.G. Grdadolnik et al. / Investigation of Novel DNA Gyrase Inhibitors
313
with weakly binding ligands exist in a dynamic equilibrium between free and bound species, which usually can not be crystallized and the NMR is the method of choice for studying conformational and dynamic properties of such complexes. The search for the ligands with low binding affinity is important, because it reduces the number of compounds, which have to be synthesized. These molecules can be further improved on the basis of structural information and the knowledge of their interactions with the protein target. A number of low affinity ligands can be combined as building blocks for novel high binding affinity ligands. Thus the number of compounds, which have to be synthesized, is reduced and there is no need for investigation of a huge number of compounds by random screening, which can fail in searching for the new lead structures. Binding of new compounds to the protein target can be monitored by observation of chemical shift changes of the protein signals in the "fingerprint" 2D HSQC spectrum (5). When the molecule binds to the protein it changes the local chemical environment and causes the change of the local magnetic field, which manifests in the change of chemical shifts of the residues at the binding site. Chemical shifts can be rapidly measured even for larger proteins. The measuring time for NMR experiments mentioned above is approximately 30 minutes with conventional NMR equipment and approximately 10 minutes with the new cryogenic probe technology (2). In this article, we are presenting our efforts in investigation of novel DNA gyrase inhibitors. DNA gyrase is an essential prokaryotic type II topoisomerase, which is involved in vital processes of DNA replication, transcription and recombination (6,7). Its role as an attractive antibacterial target arises from the fact that there are no mammalian gyrases (8). The active enzyme consists of an A2B2 complex. The A subunit (97 kDa) is involved in direct interactions in DNA breakage and reunion, while the B subunit (90 kDa) catalyses the hydrolysis of ATP. Several classes of potent DNA gyrase inhibitors have been found e.g. quinolones, cyclothialidines and coumarins. Especially quinolones, which bind to the A subunit and inhibit the DNA-breakage-reunion cycle (9), are widely used in medicine as broad-spectrum antibiotics against bacteria such as B.anthracis, which causes anthrax (10). Cyclothialidines and coumarins bind to the ATP recognition site located in the subunit B and inhibit the ATP-ase activity, as well as dimerization of the protein (11,12). However, the known inhibitors have limitations due to bacterial resistance and cytotoxicity for human cells (13,14), which are stimulating the search for novel compounds. The methodology applied in our searching for novel high affinity DNA gyrase inhibitors is high resolution NMR spectroscopy and consists of the following steps. 1st step: Resonance assignment of the protein target. The assignment of resonances in the NMR spectra to specific atoms in the protein sequence enables to identify the residues of protein, which are influenced upon binding of ligands. The molecular weight of the DNA gyrase subunits is far beyond the capabilities of high-resolution NMR spectroscopy, which in nowadays are limited to around 40 kDa. Fortunately, the 24 kDa Nterminal part of the B subunit contains the entire ATP binding site (15,16) and can be separately overexpressed in bacteria, isotopically labeled and prepared in sufficient concentrations for NMR measurements to perform sequential resonance assignment of the protein. 2nd step: Exploring the binding of ligands. In the second step binding of low molecular weight ligands is determined by observation of chemical shifts changes in the 2D 15 N HSQC spectra of the protein. Detailed analysis of chemical shift differences allows the determination of location of the protein binding site. 3rd step: Optimization of the ligand binding. Information from chemical shift changes is used in combination with the results from the molecular modeling of ligands complexed with the protein to predict their location and orientation in the protein binding site, which is essential for the optimization of ligands and rational design of more potent
314
S.G. Grdadolnik et al. / Investigation of Novel DNA Gyrase Inhibitors
inhibitors. The fragments, which are identified as ligands and bind to the individual protein subsites are linked together in order to find more potent DNA gyrase inhibitors. The NMR methodology described above is using the principles of the so-called SAR-byNMR approach, which was developed in the group of S. Fesik at Abbot Laboratories (1–4). Materials and methods Sample preparation: The 24 kDa fragment of gyrase B from E.coli was overexpressed in the same organism and isolated by affinity chromatography on novobiocin-Sepharose based on the modification of a method of Staudenbauer et al. (17). Protein was eluted from the novobiocin-Sepharose column in a buffer containing 8 M urea in 20 mM Tris pH 7.5 and refolded by extensive dialysis against 50 mM Tris pH 7.5, 10% glycerol, 1 mM EDTA, 2 mM DTT and 0.02 % NaN3. The last buffer exchange contained 20 mM K-phosphate buffer at pH 7.2, ImM EDTA and 2 mM DTT. For NMR experiments protein sample was concentrated to approximately 0.4 mM concentration and 5% of D2O was added. For NMR experiments involving ligands, which are poorly soluble in water, DMSO was added to the sample up to the final concentration of 15%, at which concentration the protein still displayed the full dispersion of chemical shifts, indicating the preservation of defined tertiary structure. Stock of gyrase inhibitors were prepared at 10-100 mM concentration in DMSO and used for NMR titration of 24 kDa fragment. NMR spectroscopy: All NMR experiments were recorded at 303 K on a Varian INOVA 600 MHz NMR spectrometer using the pulse sequences provided in the ProteinPack Varian library. All experiments employed pulsed field gradients and gradient-selected sensitivity enhancement. For water suppression selective water flip-back pulses and gradient coherence selection were utilized (18). The 1H, 13Ca and I3C0/^ spectral widths were 10, 2.2, 4.8 and 12 kHz, respectively. The three-dimensional (3D) HNCA (18,19), HN(CO)CA (20) and HNCACB (18,19,21) spectra were recorded with the 512x64x32 complex data points, 24 scans and a relaxation delay of ls. The two-dimensional (2D) I5N HSQC spectra (5) were recorded with 512x64 complex points, 16 scans and a relaxation delay of ls. Data processing has included linear prediction in the incremented dimensions, apodization with sine-bell square function shifted by Ti/2 and zero filling. Molecular Modeling Molecular models of the 24kDa N-terminal fragment of gyrase subunit B in the complex with ligands novobiocin and FQ-115 were built by using the experimental X-ray structures. Two structures deposited in the Protein Data Bank were used to model the complex with novobiocin: N-terminal 43kDa fragment of E. coli GyrB subunit (g43) with ADPNP in the binding site (PDB entry 1EII) (22) and N-terminal 24kDa fragment of E. coli GyrB subunit (g24) with novobiocin in the binding site (PDB entry 1AJ6) (23). For building of the model of ligand FQ-115 in the complex with 24kDa N-terminal fragment of gyrase subunit B we used the experimental X-ray structure of the DNA gyrase (N-terminal 43kDa fragment of E. coli GyrB subunit) with ADPNP in the binding site (15) and, in addition, structures of ATP binding site in a structurally similar enzyme protein tyrosine kinase-substrate/inhibitor analogue complexes: protein tyrosine kinase in complex with a chromone derivative (PDB entry 2HCK) and tyrosine kinase with a nucleotide analog AMPPNP in the binding site(PDB entry 1AD5) (24). By superimposing the backbones of experimental structures of two protein tyrosine kinase complexes and ATP binding site of the gyrase the starting position of FQ-115 in the
S.G. Grdadolnik et al. / Investigation of Novel DNA Gyrase Inhibitors
315
DNA gyrase (subunit B) was determined. Next, molecular mechanics computations were performed in order to refine the initial structural models by using the Discover module as available in InsightII, Version 97 software package (MSI). The inhibitor binding sites were soaked with water molecules using radius of hydration R = 15A. All calculations were performed at pH 7.4 to determine the charge state of ionizable amino-acid residues, and consistent valence force field (CVFF) was used throughout. Potential energy of all systems described above was minimized using a combination of steepest descents (1000 steps) and conjugate gradients (5000 steps). The minimization was terminated when the energy gradients did not exceed 0.001 kcal/A.
Results Assignment of the protein: The assignment of the NMR signals to specific atoms in the protein sequence is done in a sequential way i.e. the signals of the neighboring amino acids are searched on the basis of inter- and intra-molecular correlations of the backbone nuclei. A variety of three-dimensional (3D) heteronuclear correlation experiments have been developed for sequential resonance assignment of the protein backbone (25,26). These methods are designed to overcome the problems of large signal overlap and short transverse relaxation times, which are connected with the large molecular weight of the proteins and prevent the application of conventional 2D NMR. Additional dimensions are implemented in the NMR experiments (3D or 4D NMR) to reduce signal overlap and larger heteronuclear coupling constants are used for magnetization transfer to overcome fast relaxation. For the proteins with molecular weight above ca. 20 kDa the use of partial deuteration to reduce relaxation is beneficial (27, 28). In addition, sample concentrations must be kept sufficiently low to prevent aggregation of the protein. Therefore, isotopic labeling of proteins with 15N and/or 13C nuclei is required to obtain the 3D spectra in a reasonable measuring time. The N and C uniformly labeled 24-kDa N-terminal fragment of gyrase B was prepared to perform the 3D heteronuclear correlation methods. The most stable condition of the protein is achieved during the complexation. Thus, the addition of novobiocin (Fig. 1), which belongs to the family of coumarin antibiotics (11) and is one of the most potent DNA gyrase inhibitors with binding affinity in nanomolar range caused stabilization of the complex with DNA gyrase protein. The novobiocin - gyrase B complex remained stable over several weeks, thus the measuring of a series of 3D experiments under stable conditions was possible. As shown bellow the experiments performed on uniformly 15N, C-doubly-labeled protein with combination of comparative studies of the free and bound protein were sufficient to assign the protein and the expensive procedure of the protein partial deuteration could be avoided.
H2N
w
OH
H \\ O
CH3
Figure 1 :The chemical structure of novobiocin, which is one of the most potent DNA gyrase inhibitors.
316
S.G. Grdadolnik el al. / Investigation of Novel DNA Gyrase Inhibitors
In order to select the most efficient way for sequential resonance assignment of the protein the NH signal comparison of the commonly used 3D methods was performed. The 24 kDa molecular weight is already critical for success of the high resolution NMR measurements and after sensitivity comparison it was realized that only a few of the 3D experiments can be successfully applied. For the sequential assignment HNCA and HN(CO)CA experiments were chosen. In the HNCA spectrum intra- and inter-residue correlations of amide 1HN and 15 N nuclei with Ca nuclei are observed (Fig.2, left), while in the HN(CO)CA spectrum only the inter-residue correlations between the same nuclei are present (Fig. 2, right). With the comparison of both spectra the intra- and inter-residue correlations can be distinguished and search for the resonances of the neighboring residues can be performed. Promising sensitivity was observed also for the HNCACB spectrum, where besides the correlations present in HNCA spectrum also the intra- and inter-residue correlations of the amide 1HN and 15N nuclei with I3C^ nuclei are observed. However, the sensitivity of the experiment was insufficient to be used for sequential assignment, because mostly only the intra-residue correlations resulted in the spectrum. The HNCACB spectrum contains helpful information of C chemical shifts, which have characteristic values for certain amino acids like alanine, serine or threonine and significantly facilitate the identification of the residues and thus the resonance assignment of the protein. In order to overcome the assignment problems due to the limited number of 3D experiments comparative NMR studies of the free and bound protein were performed and the information from the X-ray structure of the novobiocin-Gyrase B complex regarding the observed hydrogen bonds and hydrophobic interactions between the novobiocin and protein was used (16). The Ala47, Asn46, Asp73 and Arg136 form hydrogen bonds directly with novobiocin, while the Val43, Glu50, Gly77, Val71 and Thrl65 form hydrogen bonds across the ordered water molecules. The Arg76 and Pro79 form hydrophobic interaction with the coumarin ring of the novobiocin. HNCA
HN(CO)CA Q72
cu,,
El 8.1 056
" if
G77
F.I 83
Tt
F.I83? 1 LI.W t
*i
HiC
!<••„. 1 C '(i 1 •
» V7I Y184 >CW
12.0
II.0
10.0
H Ippm]
9.0
8.0 \ = i : s ~ < . ppin
12.0
II.0
10.0 H|ppm|
9.0
8.0 s = i:s^f>ppm
Figure 2: Planes from the 3D HNCA and HN(CO)CA spectra. With combination of both spectra inter (i-l) and intra (i) - residue correlations of amide nuclei ( 1 HN, I5 N) with 13Ca nuclei in HNCA spectrum can be distinguished. The sequential assignment is performed by searching for the amide resonances, which has the Ca ( i - l ) and C a (i) correlations at the same chemical shift. In the presented planes the amide resonance of the previous residue to Y184 was found in the same plane: the C" (i-l) correlation peak of amide Y184 has the same chemical shift as the Ca correlation peak of amide El83, thus identifying the signals of the two neighboring residues.
S.G. Grdadolnik et al. /Investigation of Novel DNA Gyrase Inhibitors
317
Figure 3: The model of novobiocin in complex with the 24 kDa N-terminal fragment of gyrase B. The residues participating in hydrogen bonds and hydrophobic interactions with novobiocin are labeled. The part of the protein, which could not be traced in the X-ray structure, is colored in black.
In the Brookhaven Protein Data Bank only the crystal structure of the N-terminal 24kDa fragment R136H mutant of DNA gyrase complexed with novobiocin is deposited. This structure lacks some structural information, which is vital for our study." Therefore, a structural model of g24-novobiocin complex (Fig. 3) has been constructed, which includes Argl36 and addition of missing amino acid residues not traced in the X-ray structure. Modeled positions of residues are in good agreement with previously published analyses (16,22). For example, Argl36 forms a direct hydrogen-bonding interaction with novobiocin, which is vital in directing the noviose sugar to the appropriate site. In the wild type structure the ester and carbonyl oxygens of the coumarin ring are 3.2 and 2.6A from the guanidinium nitrogens of Argl36, while the distances in our model are 3.18 and 2.92A, respectively (Fig. 4).
Figure 4: Detail from the model of novobiocin in complex with the 24 kDa N-terminal fragment of gyrase B: interaction of Argl36 with the coumarin ring of novobiocin.
318
S.G.Grdadolnik et al. /Investigation of Novel DNA Gyrase Inhibitors
Figure 5: The 2D 1SN-HSQCspectra of the free (left) and bound (right) protein. The signals of residues, which are involved in hydrogen bonds and hydrophobic interactions with novobiocin are labeled.
The chemical shifts of residues of the protein involved in intermolecular interactions with novobiocin are expected to be the most significantly affected. The "N HSQC spectra of the free and bound protein (Fig. 5) show that the chemical shifts of most of the residues, directly involved in..intermolecular interactions are significantly changed upon addition of novobiocin and served to check and complete the resonance assignment of the protein. Finally, almost the entire protein (90%) was assigned and the complete assignment of the residues of the protein active site was achieved, which is of the most importance for search for the new ligands. Identification of the protein binding site: The binding site of ligands in the protein can be identified on the basis of the most pronounced chemical shift differences upon addition of ligands. In the case of novobiocin the chemical shift differences clearly indicate binding of the antibiotic to the ATP binding site of the protein. Most of the residues with pronounced chemical shift differences are concentrated around the residues, which are involved in intermolecular interactions with the novobiocin (Fig. 6). Besides the interactions mentioned in the previous chapter, we can observe pronounced effect on the chemical shifts of the helix, which contains Val120 (Fig. 6). The side chain of Val120 is in a hydrophobic contact with the methyl groups of the noviose part of the novobiocin. The comparative NMR studies of the free protein and protein in complex with novobiocin were not important just for the assignment purposes but also enabled us to identify the residues of the protein, which are inflpenced upon binding of the novobiocin. This information can help us to determine the location of the new ligands in the ATP binding pocket.
S.G. Grdadolnik et al / Investigation of Novel DNA Gyrase Inhibitors
319
Figure 6: Presentation of residues with the most pronounced chemical shift differences upon addition of novobiocin. The residues, with the chemical shifts differences larger than 0.1 ppm for 1H or 0.4 ppm for 15N nuclei are colored in gray and black. The residues, which were mentioned in the X ray studies to participate in the interactions with novobiocin are colored in black.
Searching for the new ligands: The binding of various low molecular weight compounds, which were expected to bind to individual subsites of the ATP binding pocket according to our structural models, has been tested. Binding of a very promising compound namely FQ115 was characterized. This compound is structurally different than novobiocin. The effect on the chemical shifts of the protein upon binding of FQ-115 was observed in the 2D 15NHSQC spectra (Fig. 7).
H'[ppin]
H [ppm]
Figure 7: Parts of the 2D 15N-HSQC spectra of 24 kDa N-terminal fragment of gyrase B in complex with FQ115 (left) and novobiocin (right), which are overlapped with the corresponding region of 2D 15N-HSQC of the free protein (dotted contours) recorded under the same conditions. Generally lower chemical shift differences are observed for FQ-115 in comparison with novobiocin as shown for R76 and R136 residues.
320
S.G. Grdadolnik et al. / Investigation of Novel DNA Gyrase Inhibitors
The binding affinity of FQ-115 to 24 kDa fragment of gyrase B is in 10 micromolar range as determined by fluorescence measurements, which might account for lower chemical shift differences in comparison to novobiocin. Measurements of gyrase inhibition by FQ-115 confirmed that it indeed inhibits the activity of gyrase B (29). Location of the new ligand in the protein binding site: All chemical shift differences due to addition of the FQ-115 to the protein are located around the same residues as in the case of novobiocin (Fig. 8) indicating that the FQ-115 binds to the ATP binding site of DNA gyrase. The location of molecules in the ATP binding site can be predicted on the basis of induced chemical shift differences (Fig. 8, left) and comparison with the effect of novobiocin on the chemical shifts (Fig. 8 right). The NMR experimental data are in good agreement with the model of FQ-115 in complex with gyrase B, which was obtained using molecular modeling methods (Fig.8). By mapping the chemical shift differences of affected residues by different colors according to their size (Fig 8) the reduced effect of FQ-115 in comparison with novobiocin on the residues R136, and R76 (which interact with the coumarin ring of novobiocin) can be observed. This indicates that the FQ-115 is probably not located in the part of the binding pocket, which is occupied by the coumarin ring of novobiocin. On the other hand, the FQ-115 is significantly perturbing the chemical shifts of the residues, which are participating in the hydrogen bonding network with noviose moiety of novobiocin and are located close to the adenine part of ADPNP, as well as the V120, which forms a hydrophobic contact with methyl groups of noviose sugar. Thus the experimental NMR data support our model, in which the FQ-115 is located in the region of binding site occupied by ADPNP and is showing overlap with the noviose sugar moiety of novobiocin.
Figure 8: Models of 24 kDa N-terminal fragment of gyrase B in complex with FQ-115 (left) and novobiocin (right). In the model of complex with FQ-115 (in black) the locations of novobiocin (in white) and ADPNP (in gray) in the protein binding site are also shown. The influence of ligand FQ-115 on the chemical shifts of the protein residues is mapped on the left figure and for comparison the influence of novobiocin is presented on the right figure. The residues, which show the chemical shifts differences larger than 0.1 ppm for 1H or 0.4 ppm for 15N nuclei are colored in black. The residues with the less pronounced chemical shift differences i.e. smaller than 0.1 ppm for 1H nuclei or 0.4ppm for I5N nuclei are colored in gray.
S.G. Grdadolnik et al. / Investigation of Novel DNA Gyrase Inhibitors
321
Model of ligand docking to the gyrase B fragment will be improved and more information about intermolecular interactions will be obtained also from measurements of structurally dependent NMR parameters, especially NOE effects. However, the initial information about location and orientation of ligands as shown in the example of compound FQ-115 in the protein binding site can be rapidly obtained from the chemical shift analysis once the protein has been fully assigned. Conclusion The NMR approach is applied (1-4) to search for the new DNA gyrase inhibitors. In order to identify the binding site of new ligands and to determine their location and orientation in the binding site the sequential resonance assignment of the protein has been performed. With combination of 3D heteronuclear correlation methods and comparative studies of the free and bound protein the backbone of 90% of the residues has been assigned. Binding of new molecules to protein target is monitored by observation of chemical shift changes in the 2D 15N HSQC spectra of the protein. The binding of a new DNA gyrase ligand (FQ-115) is identified which was found to be biologically active. The detailed analysis of chemical shift differences upon addition of FQ-115 and molecular modeling of the 24kDa Gyrase B fragment in complex with FQ-115 provided initial information about the location and orientation of ligand in the ATP binding pocket. On the basis of this information we are currently optimizing this promising DNA gyrase inhibitor and searching for compounds, which would occupy the rest of the binding pocket. In the next stage the fragments, which bind to the individual protein subsites could be linked together in order to find more potent DNA gyrase inhibitors, which could serve us as leads for rational drug design. Acknowledgment This work was supported by the Ministry of Education, Science and Sport of the Republic of Slovenia. References: [ 1] P. J. Hajduk, R. P. Meadows and S. W. Fesik, Q. Rev. Biophys. 32 (1999) 211. [2] P. J. Hajduk, T. Gerfin, J. M. Boehlen, M. Haberli, D. Marek and S. W. Fesik, J. Med. Chem. 1 (1999) 2315. [3] P. J. Hajduk, R. P. Meadows and S. W. Fesik, Science 278 (1997) 497. [4] S. B. Shuker, P. J. Hajduk, R. P. Meadows and S. W. Fesik, Science 274 (1996) 1531. [5] L. E. Kay, P. Keifer and T. Saarinen, J. Am. Chem. Soc. 114 (1992) 10663. [6] J. C. Wang, Annu. Rev. Biochem. 54 (1985) 665. [7] R. J. Reece and A. Maxwell, Crit. Rev. Biochem. Mol. Biol. 26 (1991) 335. [8] A. Maxwell, Biochem. Soc. Trans. 27 (1999) 48. [9] A. Maxwell, J. Antimicrob. Chemother. 30 (1992) 409. [10] S. I. D'iakov. V. V. Katsalukha, I. K. Lebedeva, A. V. Lukashina, and V. Raiskaia, Antibiot. Khimioter. 39 (1994) 15. [11] A. Maxwell, Mol. Microbiol. 9 (1993) 681. [12] N. Nakada, H. Gmuender, T. Hirata and M. Arisawa, Antimicrob. Agents. Chemother. 38 (1994) 1966. [13] J. S. Wolfson, Etir. J. Clin. Microbiol. Infect. Dis. 8 (1989) 1080. [14] M. Stieger, P. Angehrn, B. Wohlgensinger and H Gmuender, Antimicrob. Agents. Chemother. 40 (1996) 1060. [15] D. B. Wigley, G. J. Davies, E. J. Doson, A. Maxwell and G. Dodson, Nature. 351 (1991) 624.
322
S.G. Grdadolnik et al. / Investigation of Novel DNA Gyrase Inhibitors
[16] R. J. Lewis, O. M. P. Singh, C. V. Smith, T. Skarzynski, A. Maxwell, A. J. Wonacott and, EMBO J. 15 (1996) 1412. [17] W. L. Staudenbauer and E. Orr, Nucleic.Acids.Res. 9 (1981) 3589. [18] D. R. Muhandiram and L. E. Kay, J. Magn. Reson. Ser. B, 103 (1994) 203. [19] L. E. Kay, G. Y. Xu and T. Yamazaki, J. Magn. Reson. Ser. A, 109 (1994) 129. [20] T. Yamazaki, W. Lee, C. H. Arrowsmith, D. R. Muhandiram and L. E. Kay, J. Am. Chem. Soc., 116 (1994) 11655. [21] M. Wittekind and L. Mueller, J. Magn. Reson. Ser. B, 101 (1993) 201. [22] L. Brino, A. Urzhumtsev, M. Mousli, C. Bronner, A. Mitschler, P. Oudet and D. Moras, J. Biol. Chem.. 275 (2000) 9468. [23] G. A. Holdgate, A. Tunnicliffe, W. H. J. Ward, S. A. Weston, G. Rosenbrock, P. T. Barth, I. W. F. Taylor, R. A. Pauptit and D. Timms, Biochemistry, 36 (1997) 9663. [24] F. Sicheri, I. Moarefi, and J. Kuriyan, Nature, 385 (1997) 602. [25] L. E. Kay, M. Ikura, R. Tschudin and A. Bax, J Magn. Reson., 89 (1990) 496. [26] L. E. Kay, M. Ikura and A. Bax, J. Magn. Reson., 91 (1991) 84. [27] R. A. Venters, C. C. Huang, B. T. Farmer R. Trolard, L. D. Spicer and C. A. Fierke, J. Biomol. NMR. 5 (1995) 339. [28] K. H. Gardner and L. E. Kay, Annu. Rev. Biophys. Biomol. Struct., 27 (1998) 357. [29] M. Golob, A. Plaper, S. Zalar, R. Jerala, In vitro study of effects of flavonoids on DNA gyrase activity. In: (T. Lah, T. Turk, M. Dolinar eds.) 3rd Meeting of the Slovenian Biochemical Society with International Participation, Portoroz, 25–29. September, 1999. Book of abstracts. [Ljubljana: Slovensko biokemijsko drutvo, 1999], p. 30.
323
Author Index Andreu-Vieyra, C.V. Andreychenko, S. Apostolopoulos, V. Athanasellis, G. Blagbrough, I.S. Bonvin, A. Bouga, E.A. Camoutsis, Ch. Chatzantoni, K. Constantinou-Kokotou, V. Cordopatis, P. Courty, J. Daliani, I. Damianakos, C. De Clercq, E. Demetzos, C. Deraos, S. Detsi, A. Flordellis, C.S. Foscolos, G.B. Fountoulakis, M. Fragiadaki, M. Fytas, G. Galanis, A. Gates, P. Gavrielatos, E. Georgiev, V. Gerothanassis, I.P. Giatas, N. Grdadolnik, S.G. Habibi, H.R. Hayden, L.J. lakovidou, Z. Igglessi-Markopoulou, O. Iliodromitis, E. Jerala, R. Kapou, A. Karakantza, M. Karamanos, N.K. Karayianni, V. Karestou, E. Katsoris, P. Keivish, T. Kidric, J.
41 294 258 25 64 180 299 97 241 76 188,205,217 251 116,125 53 103,272 125,131 116,180 25 299 103 279 217 103 188 53 25 267 180 3 167,174,312 41 236 20,97 25 3 312 150,167 241 229 97 251 251 3 161
Kokotos, G. 76 Kolocouris, A. 103 Kolocouris, N. 103 Koumentakos, S. 217 Kovala-Demertzi, D. 150 Kremastinos, D. 3 Kyriakidis, D.A. 33 Kyrikou, I. 142,150 Liakopoulou-Kyriakides, M. 13,20 Lymperopoulos, A. 299 Magafa, V. 205,217 Magrioti, V. 76 Makridis, T. 76 Manolis, A.S. 299 Markopoulos, J. 25 Maswadeh, H. 125 Matsoukas, J. 3,116,180,241 Mavromoustakos, T. 3,116,125,142, 150,167,174 Militsopoulou, M. 53 Mioglou, E. 20,97 Moore, D. 236 Moore, G.J. 236 Mourelatos, D. 97 Mouzaki, A. 241 Murray, G. 236 Mutule, I. 3 Neal, A.P. 64 Neyts, J. 103 Nikolaropoulos, S. 167 Nounesis, G. 125 Oblak, M. 312 Padalko, E. 103 Pantazaka, E. 53 Papadimitriou, E. 251 Papaioannou, D. 53 116,241 Papathanassopoulos, P. Papazacharias, S. 188 Perdikuri, K. 305 Plebanski, M. 258 Polevaya, L. 3 Polykratis, A. 251 Poulos, C. 142 Pristovsek, P. 161
324
Pritsa, A.A. Probert,L. Raptis, D. Roumelioti, P. Roy, S. Sarigiannis, Y. Scheinin, M. Slaninova,J. Solmajer,T. Soulakis, V. Spyroulias, G.A. Stavropoulos, G. Tchekalarova,J. Theocharis, A.D. Thymianou, S. Troganis, A.N.
33 116 217 3 236 13 299 217 312 97 188,205,217 13,20 267 229 241 116,180
Tsakalidis, A. Tselios.T. Tsiakopoulos, N. Tsortos, A. Tzakos, A. Tzougraki, C. Vakalopoulou, P. van Nuland, N. Vassis, S. Vlahakos, D. Voyiatzi, K. Zervou, M. Zoga, A. Zompra, A.A. Zoumpoulakis, P.
305 116,180,241 53 125 180 83 20 180 53 3 53 174 3 205 3,116,174