08.12.2012 Views

Trekking Through Receptor Chemistry - Università degli Studi di ...

Trekking Through Receptor Chemistry - Università degli Studi di ...

Trekking Through Receptor Chemistry - Università degli Studi di ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

28 th CAMERINO-CYPRUS-NOORDWIJKERHOUT SYMPOSIUM<br />

<strong>Trekking</strong> <strong>Through</strong> <strong>Receptor</strong> <strong>Chemistry</strong><br />

Camerino, May 16-20, 2010<br />

UNIVERSITÁ DEGLI STUDI DI CAMERINO<br />

Palazzo Ducale, Sala della Muta


Cover image<br />

Engraving of a view of Camerino taken from the book ”Theatrum Celebriorum Urbium”,<br />

Amsterdam 1652


28 th CAMERINO-CYPRUS-NOORDWIJKERHOUT<br />

SYMPOSIUM<br />

<strong>Trekking</strong> <strong>Through</strong> <strong>Receptor</strong> <strong>Chemistry</strong><br />

Organized by the<br />

University of Camerino<br />

under the sponsorship of the<br />

Me<strong>di</strong>cinal <strong>Chemistry</strong> Division of the Italian Chemical Society (SCI)<br />

and the<br />

European Federation for Me<strong>di</strong>cinal <strong>Chemistry</strong> (EFMC)<br />

Camerino, Italy<br />

May 16-20, 2010<br />

UNIVERSITÁ DEGLI STUDI DI CAMERINO<br />

Palazzo Ducale, Sala della Muta


Organizing Committee<br />

Ambrosini D.<br />

Angeli P.<br />

Del Bello F.<br />

Giannella M.<br />

Giar<strong>di</strong>nà D.<br />

Giorgioni G.<br />

Piergentili A.<br />

Pigini M.<br />

Quaglia W.<br />

Index<br />

History 3<br />

Programme 5<br />

Opening 15<br />

Introductions to the Sessions 19<br />

Lectures 29<br />

Short Communications 97<br />

Poster Session 111<br />

List of Participants 138<br />

Acknowledgments 143<br />

Scientific Committee<br />

Donati D. (Italy)<br />

Gaviraghi G. (Italy)<br />

Giannella M. (Italy)<br />

Glennon R.A. (USA)<br />

Gualtieri F. (Italy)<br />

Leurs R. (The Netherlands)<br />

Makriyannis A. (Cyprus)<br />

Melchiorre C. (Italy)<br />

Mosti L. (Italy)<br />

Pellicciari R. (Italy)<br />

Ratti E. (Italy)<br />

Timmerman H. (The Netherlands)<br />

Triggle D.J. (USA)


HISTORY<br />

The interplay between molecular structure and biological activity is central to contemporary biome<strong>di</strong>cal<br />

and translational research, relevant not only to structural biologists, but also to <strong>di</strong>verse scientists<br />

involved in the study of bioactive compounds and signaling me<strong>di</strong>ators and those working in drug<br />

<strong>di</strong>scovery and development. International meetings on this topic have been held over the past thirty<br />

years. In 1977, Dutch me<strong>di</strong>cinal chemists organized a symposium, “Trends in Me<strong>di</strong>cinal <strong>Chemistry</strong>,”<br />

which became known as the Noordwijkerhout Symposium. The symposium emphasized the<br />

increasingly inter<strong>di</strong>sciplinary nature of the then-emerging field of structural biology. The second and<br />

third Noordwijkerhout Symposia took place in 1981 and 1985, respectively. Contemporaneously, in<br />

1978, a team of me<strong>di</strong>cinal chemists from the University of Camerino, with the sponsorship of the Italian<br />

Chemical Society, organized a Camerino meeting on the synergy between the chemical and biological<br />

aspects of receptor research. This symposium became the first of a series known as the Camerino<br />

Symposia on <strong>Receptor</strong> <strong>Chemistry</strong>, the second of which convened in 1983.<br />

In 1987, the growing number of symposia risked redundancy. To eliminate this risk and enhance further<br />

the symposium content, the organizers of the Noordwijkerhout and Camerino symposia decided to<br />

cooperate closely. As a result of this concerted international effort between the Netherlands and Italy,<br />

every two years since 1987 a joint Camerino-Noordwijkerhout Symposium has been held under the<br />

auspices of the European Federation of Me<strong>di</strong>cinal <strong>Chemistry</strong> in either Camerino (1987, 1991, 1995,<br />

1999, 2003, and 2007) or Noordwijkerhout (1989, 1993, 1997, 2001, and 2005).<br />

Likewise under the auspices of the European Federation of Me<strong>di</strong>cinal <strong>Chemistry</strong>, the Cyprus<br />

Conference was first convened in Limassol, Cyprus, in 1983. All ten Cyprus conferences have been<br />

organized by a group of internationally renowned me<strong>di</strong>cinal chemists for the purpose of emphasizing<br />

state-of-the-art research and approaches in drug <strong>di</strong>scovery. These meetings covered a wide range of<br />

<strong>di</strong>sciplines, inclu<strong>di</strong>ng combinatorial, biophysical, and computational chemistry; molecular and structural<br />

biology; the “-omics” technologies; informatics; and new targets for me<strong>di</strong>cinal chemistry. The program<br />

was designed for scientists and research administrators who seek to exploit novel opportunities in drug<br />

<strong>di</strong>scovery. The conference format and seaside locale encourage interaction among all participants. The<br />

Cyprus Conference has now joined the Camerino-Noordwijkerhout group to constitute the first tripartite<br />

European meeting in me<strong>di</strong>cinal chemistry. In 2008 the 26th Cyprus-Noordwijkerhout-Camerino<br />

Symposium in this long lasting series was held in Limassol (Cyprus) and in 2009 the 27th e<strong>di</strong>tion in<br />

Noordwijkerhout (The Netherlands).<br />

3


4<br />

Agreement on a cooperation between the organizers of the series of symposia known as the<br />

The responsible organizers confirm that<br />

Camerino-Cyprus-Noordwijkerhout symposia.<br />

- they will each organize a me<strong>di</strong>cinal chemistry symposium in subsequent years, to start in 2007<br />

with a symposium in Camerino, 2008 a Cyprus-symposium, 2009 in Noordwijkerhout, 2010 in<br />

Camerino and so on (spring season);<br />

- each symposium will have its own specific character: Camerino focusing on Advances and<br />

Perspectives in <strong>Receptor</strong> Research, Cyprus on New Approaches and Methods in Drug Research,<br />

Noordwijkerhout on Trends in Me<strong>di</strong>cinal <strong>Chemistry</strong>;<br />

- the Organizing Committees of the in<strong>di</strong>vidual symposia will be responsible for the organization<br />

of ‘their’ symposia in all aspects, inclu<strong>di</strong>ng programme and finances;<br />

- the Organizing Committee for each conference will include at least one member from each of<br />

the other two;<br />

- the local Organizing Committees will exchange their mailing lists for the purpose of promoting<br />

the symposia;<br />

- the symposia will be announced sharing the same logo and the common name “Cyprus-<br />

Noordwijkerhout-Camerino”. The first name in<strong>di</strong>cates the location where the symposium takes<br />

place and the sequence of the series is in<strong>di</strong>cated by the order of the names;<br />

- the three organizing committees guarantee to each other they will organize a symposium in the<br />

in<strong>di</strong>cated year as follows from the sequence of the series;<br />

- the conference will continue seeking sponsorship from EFMC.<br />

Signed by<br />

Representative Camerino (Prof. Mario Giannella)<br />

Representative Cyprus (Prof. Alexandros Makriyannis)<br />

Representative Noordwijkerhout (Prof. Henk Timmerman)


PROGRAMME<br />

5


6<br />

Panorama of Camerino<br />

The shrine of Macereto


SUNDAY, MAY 16<br />

Sala della Muta, Palazzo Ducale<br />

18:45-19:00<br />

M. GIANNELLA<br />

Opening<br />

19:00-19:30<br />

U. HACKSELL<br />

Discovery and development of drugs for CNS <strong>di</strong>seases<br />

19:30-20:15<br />

Welcome of ANTONIO DAL SORDO TRIO<br />

OSTERIA DI MEZZO<br />

20:30 Welcome Dinner<br />

CORTE DEL PALAZZO DUCALE<br />

Ancient and Modern Emotions<br />

Book exhibition by Treccani<br />

Lighting effects by Guzzini<br />

7


8<br />

MONDAY, MAY 17<br />

Sala della Muta, Palazzo Ducale<br />

Starting on the right foot: selection criteria in lead <strong>di</strong>scovery<br />

9:00-9:10<br />

Chairman: R. J. Thomas<br />

9:10-9:40<br />

R. E. HUBBARD<br />

Too much choice: selecting which fragments to optimise<br />

9:40-10:10<br />

E. H. KERNS<br />

Integration of drug-like properties in selection, optimization and biological<br />

assessment in lead <strong>di</strong>scovery<br />

10:10-10:30 Coffee Break<br />

10:30-11:00<br />

C. GHIRON<br />

The design, management and maintenance of a screening collection<br />

11:00-11:30<br />

P. D. LEESON<br />

Can we improve “Compound Quality” ?<br />

11:30-11:45<br />

I. IJJAALI<br />

SAR knowledgebase approach for exploring target-ligand selectivity<br />

patterns: application to kinase target family<br />

13:00 Lunch (Hotel I Duchi)


MONDAY, MAY 17<br />

Sala della Muta, Palazzo Ducale<br />

New avenues to target G protein coupled receptors<br />

15:00-15:10<br />

Chairman: R. Leurs<br />

15:10-15:40<br />

M. BOUVIER<br />

Ligand-based GPCR signalling; potential implications for drug <strong>di</strong>scovery<br />

15.40-16:10<br />

G. J. R. ZAMAN<br />

New avenues to target Wnt/Frizzled receptor signaling<br />

16:10-16:40<br />

M. ALLEGRETTI<br />

Pharmacological characterization of functionally selective CXCR inhibitors<br />

16:40-17:00 Coffee Break<br />

17:00-17:30<br />

M. J. SMIT<br />

Nanobo<strong>di</strong>es as new GPCR modulators<br />

17:30-18:00<br />

S. SCHANN<br />

mGluR4 Positive allosteric modulator: new para<strong>di</strong>gm for CNS in<strong>di</strong>cation<br />

treatment<br />

18:00-18:30<br />

R. PELLICCIARI<br />

Prospects for a TGR5-me<strong>di</strong>ated control of glucose homeostasis: focus on the<br />

clinical can<strong>di</strong>date S-EMCA (INT-777)<br />

18:30-18:45<br />

S. COSTANZI<br />

Identification of GPCR ligands through docking-based virtual screening: not<br />

only crystal structures but also homology models<br />

19:30 Dinner (Hotel I Duchi)<br />

21:00 Offerta dei Ceri (Basilica S. Venanzio)<br />

9


10<br />

TUESDAY, MAY 18<br />

Sala della Muta, Palazzo Ducale<br />

G protein coupled receptors: old landscape with new eyes<br />

9:00-9:10<br />

Chairman: D. Donati<br />

9:10-9:40<br />

C. ENZENSPERGER<br />

The dopamine D1-family: steps towards selectivity<br />

9:40-10:10<br />

A. MAKRIYANNIS<br />

The endocannabinoid proteins as drug targets<br />

10:10-10:30 Coffee Break<br />

10:30-11:00<br />

D. PIOMELLI<br />

The endocannabinoid system and the regulation of pain and emotion<br />

11:00-11:30<br />

S. RONZONI<br />

Novel potent and selective ORL-1 antagonists with efficacy in animal models<br />

of Parkinson’s <strong>di</strong>sease and neuropathic pain<br />

11:30-12:00<br />

P. S. PORTOGHESE<br />

Are heteromeric G protein-coupled receptors relevant to in vivo<br />

pharmacology? Opioid receptors as a case in point<br />

12:00-12:15<br />

R. G. BOOTH<br />

Functionally-Selective Serotonin 5-HT2 GPCR Drugs:<br />

5-HT2C agonists with 5-HT2A/2B inverse agonist activity for neuropsychiatric<br />

<strong>di</strong>sorders<br />

12:15-12:30<br />

R. A. SMITS<br />

Me<strong>di</strong>cinal chemistry of novel histamine H4 receptor antagonists<br />

13:00 Lunch (Hotel I Duchi)


TUESDAY, MAY 18<br />

Sala della Muta, Palazzo Ducale<br />

14:45-15:00<br />

R. FORLANI<br />

The dark side of research: multiple data source management<br />

Application of computational methods in lead <strong>di</strong>scovery<br />

15:00-15:10<br />

Chairman: A. Martinelli<br />

15:10-15:40<br />

M. S. CONGREVE<br />

Stabilized GPCRs for structure based drug design<br />

15:40-16:10<br />

K.-H. BARINGHAUS<br />

Computer-assisted lead fin<strong>di</strong>ng of GPCR modulators<br />

16:10-16:25<br />

A. CARRIERI<br />

Novel α1-adrenoreceptor antagonists related to openphen<strong>di</strong>oxan: biological<br />

evaluation and α1d computational study<br />

16:25-16:40<br />

T. TUCCINARDI<br />

Protein kinases: docking and homology modeling reliability<br />

16:40-17:00 Coffee Break<br />

<strong>Receptor</strong>s and beyond<br />

17:00-17:10<br />

Chairman: R. A. Glennon<br />

17:10-17:40<br />

P. J. L. SVENNINGSSON<br />

Regulation of 5-HT receptor functions by p11<br />

17:40 18:10<br />

M. K. SCHULTE<br />

Allosteric modulation of neuronal nicotinic acetylcholine receptors<br />

18:10-18:40<br />

K. MOHR<br />

Dualsteric GPCR-targeting: fine-tuning bin<strong>di</strong>ng selectivity and signalling<br />

pathway activation<br />

18:40-18:55<br />

M. DUKAT<br />

A negative allosteric modulator of α7 nAChRs<br />

19:30 Typicality & Folklore (Castello <strong>di</strong> Lanciano)<br />

11


12<br />

WEDNESDAY, MAY 19<br />

Sala della Muta, Palazzo Ducale<br />

Polypharmacology: creating selective non selectivity<br />

9:00-9:10<br />

Chairmen: M. L. Bolognesi and C. Melchiorre<br />

9:10-9:40<br />

J. S. MASON<br />

Polypharmacology: using it to <strong>di</strong>fferentiate and as an advantage with<br />

selective non-selectivity<br />

9:40-10:10<br />

R. MORPHY<br />

Multi-target drug <strong>di</strong>scovery – past, present and future<br />

10:10-10:30 Coffee Break<br />

10:30-11:00<br />

C. J. VAN DER SCHYF<br />

Multiple mechanism drugs for neurodegenerative <strong>di</strong>seases and post-stroke<br />

neuroprotection<br />

11:00-11:30<br />

M. B. H. YOUDIM<br />

Novel therapeutic approaches constituting multifunctional neuroprotective<br />

and neurorestorative drugs for neurodegenerative <strong>di</strong>seases<br />

11:30-11:45<br />

X.-J. LI<br />

Chemoinformatics approaches for anti-dementia tra<strong>di</strong>tional Chinese<br />

me<strong>di</strong>cine research and implications for fin<strong>di</strong>ng polypharmacology drugs<br />

11:45-12:00<br />

E. SIMONI<br />

Curcumin analogs as multi-target antioxidants: focus on mithocondria<br />

13:00 Lunch (Hotel I Duchi)


WEDNESDAY, MAY 19<br />

Sala della Muta, Palazzo Ducale<br />

Orexin antagonists as novel treatment of insomnia<br />

15:00-15:10<br />

Chairman: H. Timmerman<br />

15:10-15:40<br />

E. MERLO PICH<br />

Central orexin peptidergic system as target for novel treatments of insomnia,<br />

anxiety and drug ad<strong>di</strong>ction: recent fin<strong>di</strong>ngs from the GSK Discovery<br />

Performance Unit<br />

15:40-16:10<br />

R. DI FABIO<br />

Bis-amido piperi<strong>di</strong>ne derivatives as in vitro and in vivo potent dual orexin<br />

receptor antagonists<br />

16:10-16:40<br />

A. J. ROECHER<br />

Discovery of potent, <strong>di</strong>azepane-containing dual orexin receptor antagonists<br />

for the treatment of insomnia<br />

16:40-17:00 Coffee break<br />

17:00-17:30<br />

C. BOSS<br />

Orexin receptor antagonists – A new therapeutic principle in CNS<br />

<strong>di</strong>sorders?<br />

17:30-17:45<br />

J. BENTLEY<br />

Identification of a novel OX1 antagonist series by high-throughput screening<br />

and SDM-<strong>di</strong>rected homology modeling of orexin receptors<br />

17:45-18:00<br />

J. SELENT<br />

So<strong>di</strong>um ions toggle the rotamer switch in so<strong>di</strong>um-sensitive dopaminergic Gprotein<br />

coupled receptors<br />

18:00-19:00<br />

POSTER SESSION<br />

19:30 Dinner (Hotel I Duchi)<br />

13


14<br />

THURSDAY, MAY 20<br />

Sala della Muta, Palazzo Ducale<br />

Application of stem cell technology to drug <strong>di</strong>scovery<br />

9:00-9:10<br />

Chairman: A. Makriyannis<br />

9:10-9:40<br />

F. FAROUZ<br />

Novel oxysterols activate the hedgehog pathway in vitro and stimulate spinal<br />

fusion in vivo<br />

9:40-10:10<br />

P. TUNICI<br />

Brain tumor stem cells as a tool to identify novel therapeutics<br />

10:10-10:30 Coffee Break<br />

10:30-11:00<br />

R. J. THOMAS<br />

Successful modulation of tumor stem cells with small molecule hedgehog<br />

pathway antagonists<br />

11:00-11:30<br />

G. GAVIRAGHI<br />

Therapeutic modulation of the Wnt signalling pathway, an innovative<br />

approach for oncology and neurodegeneration<br />

11:30-12:00<br />

Looking forward<br />

M. GIANNELLA, A. MAKRIYANNIS, H. TIMMERMAN<br />

12:00-12:30<br />

D. J. TRIGGLE<br />

The sustainability of pharmaceuticals in the 21 st century: the idle thoughts of<br />

an idle fellow<br />

13:00 Lunch (Hotel I Duchi)<br />

15:00 Social tour<br />

Matelica - Visit to “Museo Archeologico” and “Museo Piersanti”<br />

20:00 Social Dinner (Villa Fornari)


OPENING<br />

15


16<br />

Notes


DISCOVERY AND DEVELOPMENT OF DRUGS FOR CNS DISEASES<br />

Uli Hacksell<br />

ACADIA Pharmaceuticals Inc., San Diego, California, USA<br />

Current antipsychotic drugs suffer from various shortcomings: they are relatively effective in<br />

treating the positive symptoms of schizophrenia, but remain largely ineffective in treating the<br />

negative symptoms and, in particular, the cognitive deficits. In ad<strong>di</strong>tion, their side effect<br />

burden is a major problem. ACADIA is applying modern drug <strong>di</strong>scovery technologies<br />

integrated with tra<strong>di</strong>tional methods in attempts to <strong>di</strong>scover novel antipsychotic agents. Here I<br />

will <strong>di</strong>scuss our efforts to achieve both antipsychotic and procognitive activity by selective<br />

stimulation of M1 muscarinic receptors.<br />

We have access to a proprietary functional assay technology that allows us to study a broad<br />

range of GPCR interactions. Having enabled a functional ultra-HTS assay for M1 agonism, we<br />

screened our small-molecule library for M1 agonists and <strong>di</strong>scovered a novel type of muscarinic<br />

agonist, AC-42, that selectively stimulates M1 receptors. AC-42 does not bind to the orthosteric<br />

site of the M1 receptor, but binds to an allosteric region of the receptor that is non-conserved<br />

between the five muscarinic receptor subtypes. We have now developed several series of more<br />

potent and drug-like M1-selective agonists, inclu<strong>di</strong>ng AC-260584.<br />

As part of our effort to develop novel concepts in schizophrenia therapy, we profiled the GPCR<br />

interactions of known antipsychotic agents and their relevant metabolites and found that<br />

NDMC, the key-active metabolite of clozapine, is a partial M1 receptor agonist and that NDMC<br />

may be responsible for many of the uniquely positive effects of clozapine. We believe that the<br />

combination of D2 antagonism, 5-HT2A inverse agonism and selective M1 muscarinic agonism<br />

in the same molecule has the potential to provide superior antipsychotic activity. Several<br />

compounds with this profile have been <strong>di</strong>scovered. These pro-cognitive antipsychotic drug<br />

can<strong>di</strong>dates (PCAPs) are active in preclinical antipsychotic and cognition models.<br />

17


18<br />

<strong>Trekking</strong> through the “Cresta della Sibilla”<br />

Palazzo Borghese


INTRODUCTION TO THE SESSIONS<br />

19


20<br />

STARTING ON THE RIGHT FOOT: SELECTION CRITERIA IN LEAD<br />

DISCOVERY<br />

Russell J. Thomas<br />

Me<strong>di</strong>cinal <strong>Chemistry</strong> Department, Siena Biotech S.p.A., Strada del Petriccio e Belriguardo, 35, 53100, Siena, Italy<br />

rthomas@sienabiotech.it.<br />

The quality of structural starting points in any integrated drug <strong>di</strong>scovery programme will have an enormous impact<br />

on the activities, costs and ultimately chances of success of the project.<br />

The more time and money spent on ra<strong>di</strong>cal mo<strong>di</strong>fications of chemical starting points, attempting to transform<br />

target affinity into efficacy and selectivity, the lower the probability of finally arriving in the clinic with an<br />

effective, innovative and safe me<strong>di</strong>cine (1).<br />

This session looks at a number of complementary approaches to “starting on the right foot” with high quality<br />

compounds and data-driven decision tools as early as possible in a project.<br />

Optimizing compound affinity often results in an increase in molecular weight and lipophilicity. To avoid this<br />

tendency can be extremely challenging. Fragment based approaches accept this fact and work around it by<br />

carefully selecting significantly smaller ligands (2), albeit with often modest potency, and then attempt to improve<br />

the potency and properties through structure-based design.<br />

Irrespective of whether the initial lead structures are derived from fragments or larger compounds, research<br />

projects require clear vision of the importance of in vivo efficacy and safety as opposed to in vitro affinity (3).<br />

Ad<strong>di</strong>tionally, a suite of high quality in silico, in vitro and in vivo models (4), with appropriate tools to collate the<br />

data generated, are essential to build new hypotheses to test.<br />

Success depends not only upon careful selection of what to screen and synthesize but also on the way these<br />

compounds are characterized, organized, archived and <strong>di</strong>stributed to projects. Many aspects of how this is<br />

achieved will depend on the size and nature of the research organization however the basic scientific and<br />

organizational principles remain.<br />

(1) Proudfoot, J. R., Bioorg. Med. Chem. Lett., 2002, 12, 1647-1650.<br />

(2) Chen, I-J., Hubbard, R. E. J. Comput. Aided Mol. Des., 2009, 23, 603-620<br />

(3) Leeson, P. D., Springthorpe, B. Nature Rev. Drug Discov., 2007, 6, 881-890<br />

(4) Kerns, E., Di, L. Drug-like Properties: Concepts, Structure Design and Methods: from ADME to Toxicity Optimization,<br />

Academic Press, 2008.<br />

I-01


NEW AVENUES TO TARGET G-PROTEIN COUPLED RECEPTORS<br />

Rob Leurs<br />

Department of Me<strong>di</strong>cinal <strong>Chemistry</strong>, Leiden/Amsterdam Center for Drug Research, VU University Amsterdam, the<br />

Netherlands.<br />

G-protein coupled receptors have for many years been one of the most favorite drug targets for effective<br />

therapeutic intervention in many <strong>di</strong>sorders. Small molecules have for many years been very effective tools to<br />

target this important class of receptor proteins. The recent <strong>di</strong>scovery of many new concepts of G protein coupled<br />

receptor action (allosterism, ligand-biased signaling, hetero<strong>di</strong>merization) have greatly increased the possibilities to<br />

affect G protein coupled receptor function with small molecules. Moreover, research into GPCRs targeting has<br />

been growing at a fast pace and the range of approached that can be applied to target GPCRs continues to grow. In<br />

this session various new ways to target GPCRs will be <strong>di</strong>scussed.<br />

I-02<br />

21


22<br />

G PROTEIN COUPLED RECEPTORS: OLD LANDSCAPE WITH NEW<br />

EYES<br />

Daniele Donati<br />

Nerviano Me<strong>di</strong>cal Sciences srl, Oncology Business Unit, Viale Pasteur 10, 20014 Nerviano (MI), Italy<br />

The G-protein coupled receptors are seven trans-membrane domain (7TM) proteins and represent the largest<br />

family of proteins in the human genome. They are involved in the modulation of a large number of physiological<br />

and pathophysiological processes, and are the target for about 30% of prescription drugs (1-3). More in general,<br />

GPCRs represent the most explored and the successful target family in drug <strong>di</strong>scovery, covering a number of<br />

therapeutic areas, and many new drug can<strong>di</strong>dates are being reported as selective ligands for such receptors (4).<br />

With very few exceptions, all the currently available therapeutics based on the relevant receptors are targeting their<br />

orthosteric bin<strong>di</strong>ng sites and this reflects the nature of the assays (typically bin<strong>di</strong>ng assays) generally utilized as<br />

primary screening for novel chemical entities(5).<br />

In recent years, the need for more selective and safer therapeutic agents has triggered new research efforts aimed at<br />

the intimate understan<strong>di</strong>ng of the mechanism of action of GPCRs, with specific attention to the signal transduction<br />

mechanisms as well as to the nature of the effective receptor system responsible for the observed physiological<br />

action of both endogenous and exogenous ligands. A great deal of interest has indeed emerged for allosteric<br />

modulation (positive and negative) of both homo<strong>di</strong>meric and hetero<strong>di</strong>meric GPCR receptors (5, 6). The number of<br />

experimental evidences supporting the existence and the relevance of GPCR oligomerization on drug <strong>di</strong>scovery is<br />

growing and it is now evident the need for new strategies in the search of novel and more effective drugs targeting<br />

GPCR receptors. We need indeed to consider that <strong>di</strong>fferent and novel screening approaches have to be used (eg by<br />

co-expressing the protomers in the same cell line). As well, we need to consider that the oligomerization may be<br />

tissue specific and therefore we may need to explore more in deep the tissue selectivity for a new GPCR ligand.<br />

(1) Lagestrom, M.C.; Schioth, H.B. Nat. Rev. Drug Discov. 2008, 7, 339-357.<br />

(2) Overington, J.P. Nat. Rev. Drug Discov. 2006, 5, 993-996.<br />

(3) Jacoby, E.; Bouhelal, R.; Gerspacher, M.; Seuwen, K. Chem MedChem 2006, 1, 761-782<br />

(4) Gilchrist, A. Curr. Opin. Drug Discov. 2008, 3, 375-389<br />

(5) Milligan, G., Smith, N.J. Trends Pharmacol. Sci. 2007, 28, 615-620 and references cited therein<br />

(6) Panetta, R.; Greenwood, M.T., Drug Discov. Today 2008, 13, 1059-1066 and references cited therein<br />

I-03


APPLICATIONS OF COMPUTATIONAL METHODS IN LEAD<br />

DISCOVERY<br />

Adriano Martinelli<br />

Department of Pharmaceutical Sciences - University of Pisa.<br />

At present it is largely accepted that computational methods are an essential tool for <strong>di</strong>scovering new<br />

pharmacological leads.<br />

The lectures of this section concern computational stu<strong>di</strong>es of biological targets, which is the first step for a<br />

structural based <strong>di</strong>scovery strategy able to clarify the ligand-receptor interactions and to furnish the bases for the<br />

virtual screening of new ligands.<br />

Three lectures deal with G-protein-coupled receptors (GPCRs). The superfamily of GPCRs are single polypeptide<br />

chains possessing seven hydrophobic transmembrane-spanning segments that couple with an effector molecule<br />

through a trimeric G protein complex. At present they are the target of about 50% of the drugs in the market.<br />

Baringhaus et al. propose two approaches for fin<strong>di</strong>ng GPCR modulators, the first one is based on the quantitative<br />

similarity description of GPCR pairs, the latter on the definition of structure based pharmacophore models.<br />

A knowledge of the GPCR 3D structure could be of great help in the task of understan<strong>di</strong>ng their function and in<br />

the rational design of specific ligands, however high-resolution structural characterization is still an extremely<br />

<strong>di</strong>fficult task. For this reason, great importance has been placed on molecular modeling stu<strong>di</strong>es, and especially on<br />

homology modelling (HM) techniques which, however, request experimentally determined structures of suitable<br />

templates.<br />

Congreve describes a new technology able to stabilize GPCRs outside of the cell membrane therefore allowing<br />

their crystallization and X-ray structure determination.<br />

Carrieri et al. show how a computational study can help to elucidate the interaction with the receptor of novel α1-<br />

adrenoreceptor antagonists.<br />

The topic of the last lecture from Tuccinar<strong>di</strong> concerns an analysis of the reliability of molecular docking and<br />

homology modeling methods applied to another important class of drug targets, the protein kinases.<br />

I-04<br />

23


24<br />

RECEPTORS AND BEYOND<br />

Richard A. Glennon<br />

Department of Me<strong>di</strong>cinal <strong>Chemistry</strong>, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia 23298 USA<br />

Paul Ehrlich and John Langley, contemporaneously investigating the effects of chemical agents or plant extracts<br />

on physiological function in the late 1800s, suggested that specific interactions might be occurring. Ehrlich<br />

proposed the “side chain theory” (i.e., cells might have an affinity for certain chemicals or toxins because of their<br />

chemical composition), and in 1905 Langley introduced the term “receptive substance”. In the 1920s and 1930s<br />

Clarke and Gaddum introduced the concepts of log dose-response curves, competitive antagonism, and developed<br />

equations that might explain drug receptor interactions. Schild mathematically described, and demonstrated the<br />

concepts of dose-ratio and affinity. Subsequent contributions include those made by Ariens (partial agonists,<br />

intrinsic activity), Stephenson (efficacy), Furchgott (spare receptors, intrinsic efficacy), Paton (rate theory), del<br />

Castillo and Katz (interconvertible forms of receptors), Lefkowitz et al., (ternary complex model), Black and Leff<br />

(operational model), Costa and Herz (constitutive activity), Lefkowitz and Costa (allosteric ternary complex<br />

models), Weiss/Kenakin (cubic ternary complex model), and others. From its humble beginnings, the “receptive<br />

substance” has become more complex. At least four major superfamilies of receptors are recognized: ion channel,<br />

G-protein coupled (GPCRs), tyrosine kinase (Trk), and nuclear receptors. Furthermore, the >1,000 GPCRs are<br />

further <strong>di</strong>vided into six classes: Class A (rhodopsin-like), Class B (secretin family), Class C (metabotropic), Class<br />

D (fungal), Class E (cyclic AMP), and Class F (frizzled). And, many of these receptors have been targets of drug<br />

<strong>di</strong>scovery/development.<br />

Apart from <strong>di</strong>rect interactions with a receptor, there are other receptor-associated targets that are of interest<br />

because they can in<strong>di</strong>rectly influence the actions of a given neurotransmitter. Several such possibilities include:<br />

allosterism, modulation of post-receptor events, and agonist-<strong>di</strong>rected trafficking. i) An allosteric (or allotopic)<br />

effect results when an agent mo<strong>di</strong>fies the effect of another (i.e., orthosteric agent) by bin<strong>di</strong>ng to a protein at a site<br />

(i.e., allosteric site) topologically <strong>di</strong>fferent from that of the orthosteric ligand. Allosteric modulators may be<br />

positive or negative. Early stu<strong>di</strong>es focussed on allosteric regulation of ion channel receptors, but more recently are<br />

being <strong>di</strong>rected to GPCRs. ii) Dopamine (DA) is associated with a cAMP/PKA second messenger pathway that<br />

targets DA- and cAMP-regulated phosphoprotein (i.e., DARPP-32). The function of DARPP-32 is regulated by its<br />

phosphorylation state, and agents that target receptors other than DA receptors can influence the phosphorylation<br />

state of DARPP-32 to in<strong>di</strong>rectly influence its effects. iii) Certain receptors can be coupled to multiple second<br />

messenger systems. Hence, the possibility exists that <strong>di</strong>fferent agonists might stabilize <strong>di</strong>fferent agonist receptor<br />

conformations to result in selective activation of one second messenger system over another. These topics are the<br />

focus of the present session.<br />

I-05


POLYPHARMACOLOGY: CREATING SELECTIVE NON-<br />

SELECTIVITY<br />

Carlo Melchiorre and Maria Laura Bolognesi<br />

Department of Pharmaceutical Sciences, Alma Mater <strong>Stu<strong>di</strong></strong>orum, University of Bologna, Via Belmeloro 6, 40126 Bologna,<br />

Italy.<br />

Drug <strong>di</strong>scovery has contributed greatly to advancing the life sciences and the wellbeing of society in the past<br />

century. However, several <strong>di</strong>seases remain incurable and we are facing an unpre<strong>di</strong>ctable productivity crisis. In the<br />

last 20 years, while the costs of R&D have stea<strong>di</strong>ly increased, there has been a gradual decline in its productivity,<br />

as measured by the number of new approved molecular entities. This has been attributed to drug <strong>di</strong>scovery’s<br />

gradual move from an entirely human phenotype-based endeavor to the so-called ‘reductionist approach’. This<br />

approach attempts to reduce drug action to the level of in<strong>di</strong>vidual genes, single proteins, and one potential<br />

modulating molecule. Thus, the ‘one gene, one target, one drug’ para<strong>di</strong>gm seeks to generate potent and exquisitely<br />

selective ligands that could guard against unwanted side effects. 1<br />

However, it is now widely accepted that the majority of <strong>di</strong>seases that threaten humanity are multifactorial, with<br />

substantial environmental and genetic components. For this reason, a selective single-target drug might lack<br />

efficacy, while a treatment able to address this complexity will have a greater chance of success. 2 To this end,<br />

there has been growing recognition that polypharmacology might provide therapeutic benefits where tra<strong>di</strong>tional<br />

single-target drugs have failed. A combination of drugs, which offers the prospect of ad<strong>di</strong>tional benefits, is being<br />

used to treat several complex <strong>di</strong>seases such as HIV and hypertension. A more recent interpretation of<br />

polypharmacology considers single chemical entities able to simultaneously modulate several molecular targets. 3<br />

Although these concepts emerged less than 10 years ago, multitarget kinase inhibitors are already a reality in<br />

cancer therapy. 4 Their introduction into the market provided the pharmaceutical community with the proof of<br />

concept that a ligand with a rationally designed multimodal mechanism of action would not have intrinsically<br />

overwhelming toxicity. Other successful examples of multitarget drugs combining high efficacy with reasonable<br />

safety are emerging in the fields of neurodegeneration 5 and depression. It is highly conceivable that several others<br />

will be identified in the near future, bridging the conceptual gap between specific and nonselective drugs.<br />

(1) Hopkins, A. L.; Mason, J. S.; Overington, Curr. Opin. Struct. Biol. 2006, 16, 127-136.<br />

(2) Kong, D. X.; Li, X. J.; Zhang, H. Y. Drug Discov Today 2009, 14, 115-119.<br />

(3) Cavalli, A.; Bolognesi, M. L.; Minarini, A.; Rosini, M.; Tumiatti, V.; Recanatini, M.; Melchiorre, C. J. Med.<br />

Chem. 2008, 51, 347-372.<br />

(4) Morphy, R. J. Med. Chem. 2009, 53, 1413-1437.<br />

(5) Van der Schyf, C. J.; You<strong>di</strong>m, M. B. Neurotherapeutics 2009, 6, 1-3.<br />

I-06<br />

25


26<br />

OREXIN ANTAGONISTS AS NOVEL TREATMENT OF INSOMNIA<br />

Henk Timmerman<br />

VU University Amsterdam<br />

(henktim @ planet.nl)<br />

Ever since the first symposium of this series took place in Camerino the focus has been on receptors, their<br />

mechanisms, ligands, structure and role in pharmacotherapy. As a consequence certain classes of me<strong>di</strong>cines<br />

could in the “early” hardly get attention as their molecular targets were just unknown.; antiepileptics, anti obesity<br />

agents, sleep pills, to mention a few. In the 1987 symposium out of the twenty lectures four dealt with adrenergic<br />

receptors, five with cholinergic and three with histamine receptors.<br />

Much has changed since than. The introduction of molecular biology in me<strong>di</strong>cinal chemistry and pharmacology<br />

made it possible to identify the targets for several well known me<strong>di</strong>cines for which a target was unknown.<br />

Moreover, a number of new targets were found , new me<strong>di</strong>cines could be envisaged.<br />

An example of a new class of targetable receptors is the given by the receptors belonging to the orexin system.<br />

The orexins were first described in the late 1990ies and subsequently found to bind to a n organ GPCR. The<br />

receptors have been seen as implicated in the regulation of the intake of food. Currently however the focus is<br />

mainly on sleep <strong>di</strong>sorders,.<br />

During this symposium Merlo Pich will introduce the biology orexin system; subsequently ,three lectures will<br />

deal with new compounds, under investigation for use in the treatment of insomnia.<br />

I-07


APPLICATION OF STEM CELL TECHNOLOGY TO DRUG<br />

DISCOVERY<br />

Alexandros Makriyannis, Ph.D.<br />

George D. Behrakis Chair in Pharmaceutical Biotechnology<br />

Director, Center for Drug Discovery, Northeastern University<br />

Stem cell technologies have taken an important place in therapeutics. The most advanced efforts are in the fields<br />

of oncology and neurodegeneration. The three contributions to this symposium will <strong>di</strong>scuss current efforts in that<br />

<strong>di</strong>rection. Tumor stem cells can be used as tools for developing new me<strong>di</strong>cations. Ad<strong>di</strong>tionally, small drug-like<br />

molecules are designed to modulate stem cell-related signaling.<br />

I-08<br />

27


28<br />

Fiastra Lake<br />

Pilato’s Lake


LECTURES<br />

29


30<br />

Notes


TOO MUCH CHOICE: SELECTING WHICH FRAGMENTS TO<br />

OPTIMISE<br />

Roderick E. Hubbard<br />

University of York and Vernalis Ltd.<br />

Over the past eight years, we have developed and applied fragment-based ligand <strong>di</strong>scovery methods to identify<br />

compounds that bind to a range of protein targets. These approaches have been developed both in a drug<br />

<strong>di</strong>scovery (Vernalis) and an academic (York) context. Some of these compounds have been developed into<br />

effective inhibitors to probe the mechanism of action of certain protein targets. Others have been optimized to<br />

provide clinical can<strong>di</strong>dates, now in clinical trials. Fragments are just small, weak hits. The main challenges are<br />

design of the library, robust identification of which fragments bind and the need for structural information to<br />

decide how and which fragments to progress. The advantages are that a small library can sample a potentially<br />

large chemical <strong>di</strong>versity to generate novel lead compounds and that hits can be identified for new classes of target<br />

for which existing compound collections cannot provide a hit. Various approaches have been used to develop the<br />

fragment hits into useful leads. These include (a). linking fragments together, (b). growing by <strong>di</strong>rected limited<br />

library synthesis or searching for nearest neighbours in the accessible compound databases, or (c). merging where<br />

the structures of fragments, existing tool compounds and virtual screening hits provides guidance for how to<br />

merge features from <strong>di</strong>fferent compounds together. These experiences will be summarised.<br />

One of the big challenges is deci<strong>di</strong>ng which fragments to take forward. The methods have quite a high hit rate -<br />

for well-defined bin<strong>di</strong>ng sites, up to 100 hits can be generated from a library of 1200 fragments. In this<br />

presentation I will <strong>di</strong>scuss what properties could be used to decide between fragments, inclu<strong>di</strong>ng issues of<br />

chemical tractability, vectors, opportunities to achieve selectivity and how best to integrate this information with<br />

biophysical insights from bin<strong>di</strong>ng assays and measurements of the kinetics (SPR) and thermodynamics (ITC) of<br />

bin<strong>di</strong>ng. There is no simple answer - like most areas of drug <strong>di</strong>scovery, it can be <strong>di</strong>fficult to make the right<br />

decisions.<br />

References:<br />

(1) Hubbard, R.E. et al , (2007) "The SeeDs approach: Integrating Fragments into Drug Discovery", Current Topics<br />

Me<strong>di</strong>cinal <strong>Chemistry</strong>, 7, 1568-1581<br />

(2) Brough, P.A. et al (2009), “Combining hit identification strategies: Fragment-based and in silico approaches to orally<br />

active 2-aminothieno[2,3-d]pyrimi<strong>di</strong>ne inhibitors of the Hsp90 molecular chaperone”, J Med Chem, Epub 17th July<br />

(3) Chen, I and Hubbard, R.E. (2009), “Lessons for fragment library design: analysis of output from multiple screening<br />

campaigns”, J Comp Aided Mol Des, Epub Jun 3<br />

(4) Fisher, M and Hubbard, R.E. (2009), “Fragment-based ligand <strong>di</strong>scovery”, Mol Interventions, 9, 22-30<br />

(5) Schulz, M N and Hubbard RE (2009), “Recent progress in fragment-based lead <strong>di</strong>scovery “, Curr Opin<br />

Pharmacology, Epub May 27<br />

L-01<br />

31


32<br />

Notes


INTEGRATION OF DRUG-LIKE PROPERTIES IN SELECTION,<br />

OPTIMIZATION AND BIOLOGICAL ASSESSMENT IN LEAD<br />

DISCOVERY<br />

Edward H. Kerns<br />

Wyeth Research, CN8000, Princeton NJ08543-8000, U.S.A.<br />

ADME/Tox properties are a major focus of drug <strong>di</strong>scovery. They determine the ultimate delivery properties of the<br />

drug product from the dosage form to the therapeutic target and its toxicity. Thus, compound properties affect the<br />

efficacy and safety of the drug. For this reason, drug-like properties are integrated in lead selection and<br />

optimization with other major objectives (e.g., activity, selectivity, novelty). In ad<strong>di</strong>tion, properties affect<br />

biological experiments: solubility, chemical stability and permeability affect compound delivery to the target in<br />

HTS, enzyme/receptor and cell-based assays. This presentation will <strong>di</strong>scuss key ADME/Tox properties and their<br />

assessment, effects and improvement in Lead Discovery.<br />

L-02<br />

33


34<br />

Notes


THE DESIGN, MANAGEMENT AND MAINTENANCE OF A<br />

SCREENING COLLECTION<br />

Compound Management and Analytical Services in the Small Pharma<br />

Chiara Ghiron, Raffaella Bran<strong>di</strong>, Eva Genesio<br />

Siena Biotech SpA, Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy<br />

The organisation of compound acquisition, management, logistics and analytical services in the small pharma is<br />

deceptively simple. While the number of compounds in the compound collection may not be as large as in bigger<br />

companies, the issues related to compound collection creation, sample preparation and <strong>di</strong>stribution within the site,<br />

and analytical quality controls are essentially the same.<br />

In the presentation we will show how the process is organised in Siena Biotech, illustrating our compound<br />

acquisition and registration procedures, internal requests organisation, <strong>di</strong>stribution of assay-ready plates to<br />

screening units, and the analytical controls in place at the <strong>di</strong>fferent stages of the drug <strong>di</strong>scovery process.<br />

We will also illustrate how we improved the process over the years, after thorough analysis of past practices and<br />

through implementation of in-house developed software where necessary.<br />

L-03<br />

35


36<br />

Notes


CAN WE IMPROVE ‘COMPOUND QUALITY’?<br />

Paul D. Leeson<br />

AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, Leics, LE11 5RH, UK<br />

Following the ‘rule of 5’ publication in 1997 there have been further stu<strong>di</strong>es which have underlined the importance<br />

of fundamental physical properties in controlling permeability, metabolic stability, organ toxicity, bin<strong>di</strong>ng to the<br />

hERG channel, and receptor promiscuity. Exten<strong>di</strong>ng the ‘rule of 5’, these recent fin<strong>di</strong>ngs have introduced more<br />

stringent physical property ranges for risk reduction in drug <strong>di</strong>scovery (1-6).<br />

Comparisons of oral drugs launched before and after 1983 reveals that several drug-like properties, for example<br />

lipophilicity and % polar surface area, are not changing over time, whereas others, for example molecular weight<br />

and hydrogen bon<strong>di</strong>ng atoms, are increasing in newer drugs; time-constant properties are suggested to be more<br />

important success factors in drug development (7). The physical properties of current me<strong>di</strong>cinal chemistry are<br />

reflected by the patent literature, where increases in all molecular properties vs. current drugs are evident (1),<br />

suggesting significantly increased ADMET risk in pharmaceutical company portfolios. Moreover, there are<br />

meaningful <strong>di</strong>fferences between pharmaceutical company physical property profiles, in<strong>di</strong>cating that drug<br />

properties are influenced at least as much, or more, by local lead generation and optimisation strategy, culture and<br />

behaviour than by specific target <strong>di</strong>fficulty. This conclusion is supported by a comparative analysis of company<br />

target-specific patenting practices over time.<br />

Ligand lipophilicity efficiency (LLE = p(Activity) – LogP/D) is proposed as a key measure of compound quality.<br />

Increasing LLE to >5 (ie potency


38<br />

Notes


LIGAND-BASED GPCR SIGNALLING; POTENTIAL IMPLICATIONS<br />

FOR DRUG DISCOVERY<br />

Michel Bouvier<br />

Department of Biochemistry, Groupe de Recherche Universitaire sur les Mé<strong>di</strong>caments, Institute for Research in Immunology<br />

and Cancer, Université de Montréal, Montréal, Québec, Canada<br />

Tra<strong>di</strong>tionally known for their ability to selectively activate a unique hetero-trimeric G protein, in<strong>di</strong>vidual G<br />

protein-coupled receptors (GPCR) have since been shown to activate multiple G protein subtypes as well as G-<br />

protein independent signalling cascades. In ad<strong>di</strong>tion <strong>di</strong>fferent ligands were found to selectively promote the<br />

engagement of <strong>di</strong>stinct signalling partner subsets of a given receptor. Some compounds were also found to have<br />

clearly <strong>di</strong>stinct, some time, opposite efficacies toward <strong>di</strong>fferent pathways engaged by the same receptor. This<br />

phenomenon known as ligand-biased signalling offers interesting opportunities to develop compound with<br />

increased selectivity profiles but present important challenges for the drug <strong>di</strong>scovery process in particular in the<br />

context of high throughput screening. To better understand the molecular mechanisms <strong>di</strong>recting ligand-biased<br />

signalling we developed new assays based on luminescence and resonance energy transfer as well as label-free<br />

impedance measurements that allow monitoring multiple signalling pathways and to assess the structural<br />

determinants of ligand-biased signalling. Using the β1- and β2-adrenergic receptors as study models, we <strong>di</strong>ssected<br />

the signalling cascades engaged by ligands that have biased efficacy toward the adenylyl cyclase, mitogen-<br />

activated protein kinase and calcium pathways and revealed <strong>di</strong>stinct conformational rearrangements of the<br />

signalling modules involved. Combined with molecular modelling of the recently solved 3D structures of the<br />

βARs, these stu<strong>di</strong>es should provide the basis for the rational design of drugs with predetermined biased signalling<br />

profiles and improved therapeutic activities.<br />

L-05<br />

39


40<br />

Notes


NEW AVENUES TO TARGET WNT/FRIZZLED RECEPTOR<br />

SIGNALING<br />

Guido. J. R. Zaman<br />

Merck Research Laboratories, MSD, Molenstraat 110, 5340 BH, Oss, The Netherlands<br />

Wnt/β-catenin signaling is an important regulator of cell polarity, proliferation and stem cell maintenance during<br />

development and adulthood. Signaling through this pathway is initiated by bin<strong>di</strong>ng of Wnt proteins to seven-<br />

transmembrane Frizzled receptors, which have structural homology to GPCRs. Aberrant Wnt/β-catenin signaling<br />

has been implicated in a plethora of <strong>di</strong>seases, most notably colon cancer. We have applied β-galactosidase enzyme<br />

fragment complementation to measure the nuclear translocation of β-catenin, the central transcriptional modulator<br />

in the Wnt/β-catenin pathway. To this end, β-catenin was tagged with a peptide fragment of β-galactosidase and<br />

transfected into cells expressing a correspon<strong>di</strong>ng deletion mutant of the enzyme exclusively in the nucleus.<br />

Stimulation of the cells with the Frizzled ligand Wnt-3a increased β-galactosidase activity in a dose-dependent<br />

manner with nanomolar potency. We tested a library of over 2000 synthetic chemical compounds for their ability<br />

to induce or inhibit β-catenin nuclear accumulation. The immunosuppressive protein kinase C inhibitor<br />

sotrastaurin (AEB-071) was identified as an activator of Wnt/β-catenin signaling through its inhibition of glycogen<br />

synthase kinase 3. Furthermore, we identified a class of compounds that activated Wnt/β-catenin signaling with<br />

remarkable cell-specificity and high efficacy. These data show that the β-catenin nuclear accumulation assay can<br />

be used to find new therapeutics targeting the Wnt/β-catenin pathway.<br />

L-06<br />

41


42<br />

Notes


PHARMACOLOGICAL CHARACTERIZATION OF FUNCTIONALLY<br />

SELECTIVE CXCR INHIBITORS<br />

Marcello Allegretti<br />

Dompé S.p.A. Research Center, Via Campo <strong>di</strong> Pile, snc 67100 – L’Aquila, Italy<br />

The inhibition of CXCL8 is considered as a valid target for the development of innovative treatments for a variety<br />

of severe clinical con<strong>di</strong>tions. The biological activity of CXCL8 is me<strong>di</strong>ated by the interaction with CXCR1 and<br />

CXCR2 membrane receptors belonging to the family of 7TM-GPCRs, expressed on the surface of human<br />

polimorphonuclear cells (PMNs) and of some types of T-cells.<br />

Along the last few years we developed a me<strong>di</strong>cinal chemistry program that led to the characterization of SMW<br />

inhibitors of CXCL8-induced biological activity. Novel classes of potent and selective inhibitors of CXCL8 were<br />

selected and reparixin, the first clinical can<strong>di</strong>date, is active in the low nanomolar range in inhibition of human<br />

PMN migration induced by CXCL8. Mechanism of action stu<strong>di</strong>es have clarified that reparixin acts as a non-<br />

competitive allosteric inhibitor of both CXCL8 receptors, CXCR1 and CXCR2. Further extension of the<br />

MedChem program led to the generation of novel classes of analogues with optimized pharmacokinetic profile<br />

that supported the reliability of the model.<br />

The characterization of the effects of these molecules on signalling led to a more in-depth comprehension of the<br />

“functional selectivity” of this specific class, that is the ability of allosteric modulators to inhibit some of the<br />

effects induced by the endogenous agonist without affecting others. In particular, as chemokine receptors are also<br />

able to scavenge ligands by internalization, the observation that this class of CXCR1/CXCR2 inhibitors does not<br />

interfere with receptor internalization paves the way to a highly selective inhibition strategy that preserves receptor<br />

ability to target the ligand to degradation, possibly avoi<strong>di</strong>ng ligand accumulation during a repeated treatment<br />

regimen.<br />

The pharmacological characterization of CXCR1/2 allosteric modulators in I/R injury models has been thoroughly<br />

investigated by preclinical and clinical stu<strong>di</strong>es but, in the last period, the availability of the novel molecules has<br />

allowed both the exploration of the therapeutic potential of the class in acute and chronic inflammatory con<strong>di</strong>tions<br />

and the investigation on the physiopathological role of CXCL8 in new therapeutic areas with still high me<strong>di</strong>cal<br />

need. In this talk the most recent results from our stu<strong>di</strong>es on the pharmacological preclinical characterization of<br />

functionally selective CXCR inhibitors will be <strong>di</strong>scussed.<br />

L-07<br />

43


44<br />

Notes


NANOBODIES AS NEW GPCR MODULATORS<br />

Sven Jähnichen 1 , Christophe Blanchetot 2 , Maria Gonzalez-Pajuelo 3 , David Maussang 1 , Ken Y. Chow 1 , Leontien<br />

Bosch 1 , Sin<strong>di</strong> De Vrieze 3 , Bene<strong>di</strong>kte Serruys 3 , Hans Ulrichts 3 , Wesly Vandevelde 3 , Michael Saunders 3 , Hans J. De<br />

Haard 2,3 , Dominique Schols 4 , Rob Leurs 1 , Peter Vanlandschoot 3 , Theo Verrips 2 , Martine J. Smit 1<br />

1 VU University Amsterdam, The Netherlands. 2 Utrecht University, The Netherlands, 3 Ablynx N.V., Ghent, Belgium, 4 Rega<br />

Institute, Belgium.<br />

G-protein-coupled receptors have so far not been targeted very successfully with conventional monoclonal<br />

antibo<strong>di</strong>es. Here we report the isolation and characterization of the first functional single domain antibo<strong>di</strong>es<br />

(nanobo<strong>di</strong>es) against the chemokine receptor, CXCR4. Two potent nanobo<strong>di</strong>es, were obtained using a time-<br />

efficient whole cell immunization, phage <strong>di</strong>splay and counter selection method. The highly selective nanobo<strong>di</strong>es<br />

competitively inhibited the CXCR4-me<strong>di</strong>ated activation of signal transduction cascades and antagonized the<br />

chemo-attractant effect of the CXCR4 ligand CXCL12. Epitope mapping showed that the two nanobo<strong>di</strong>es bind to<br />

<strong>di</strong>stinct but partially overlapping sites in the extracellular loops. Interestingly, one behaves as a neutral antagonist,<br />

while the other as a partial inverse agonist at the constitutively active CXCR4. Short peptide linkage of the two<br />

nanobo<strong>di</strong>es resulted in significantly increased affinity for CXCR4, full inverse agonism and picomolar activity in<br />

anti-chemotactic assays. Both monovalent and particularly the peptide-linked nanobo<strong>di</strong>es <strong>di</strong>splayed strong anti-<br />

retroviral activity against T cell- and dual-tropic human immunodeficiency virus-1 (HIV-1). Thus, the nanobody<br />

platform is highly effective in generating extremely potent and selective modulators of GPCRs.<br />

L-08<br />

45


46<br />

Notes


mGluR4 POSITIVE ALLOSTERIC MODULATOR: NEW PARADIGM<br />

FOR CNS INDICATION TREATMENT<br />

Stephan Schann, 1 Bruno Giethlen, 2 Christophe Morice, 2 Stanislas Mayer, 1 Christel Franchet, 1 Mélanie Frauli, 1<br />

Rachel Ru<strong>di</strong>gier, 2 Delphine Thiarc, 2 E<strong>di</strong>th Steinberg, 2 Camille G Wermuth, 2 Thierry Langer, 2 Pascal Neuville 1<br />

1 Domain Therapeutics, BioParc, Boulevard Sebastien Brant, F-67400 Illkirch, France<br />

2 Prestwick Chemical, Parc d’innovation, Boulevard Gonthier d’Andernach, F-67400 Illkirch, France<br />

Glutamate, the most commonly occurring neurotransmitter in the brain, is involved in the main neurological<br />

functions either <strong>di</strong>rectly or through interactions with other neurotransmitters. In excess, it can also induce cell<br />

death, a phenomenon known as excitotoxicity. Therefore, acting on glutamatergic transmission can reduce<br />

symptoms of neurological <strong>di</strong>seases but also prevent neuronal cell death.<br />

Glutamate operates through two families of receptors, the ionotropic and the metabotropic glutamate receptors<br />

(respectively iGluRs and mGluRs). This second family and more particularly Allosteric Modulators of mGluRs<br />

are now considered as better therapeutics approach for the treatment of CNS <strong>di</strong>sorders such as schizophrenia,<br />

Parkinson Disease (PD) or anxiety. 1 mGluRs have been <strong>di</strong>vided in three groups accor<strong>di</strong>ng to their sequence<br />

homologies, pharmacological properties and signal transduction pathways. mGluR4, a Gi-coupled group III<br />

mGluR, is mostly presynaptic and has been shown to be involved in regulation of both glutamatergic and<br />

GABAergic transmissions. Its specific localization in the Basal Ganglia, the CNS structure controlling<br />

movements, makes it a highly promising target can<strong>di</strong>date for the treatment of both symptomatic and<br />

neurodegenerative aspects of PD. 2<br />

Domain Therapeutics and Prestwick Chemical jointly work on the identification and early development of<br />

mGluR4 Positive Allosteric Modulators (PAMs). In the course of this program, new chemical entities were<br />

generated that <strong>di</strong>splay nanomolar efficacy for mGluR4, leftward shift of glutamate EC50, increase in glutamate<br />

Emax, subtype selectivity over the other mGluR subtypes and high water solubility. Moreover, these optimized<br />

leads showed efficacies after peripheral administration in two animal models of PD, the reserpine-induced<br />

dyskinesia and the haloperidol-induced catalepsy in mice. This presentation will summarized the scientific<br />

rationale of targeting mGluR4 for PD treatment as well as key aspects of Domain-Prestwick’s programs.<br />

References:<br />

(1) Conn, PJ.; Christopoulos, A.; Lindsley, CW. Nat. Rev. Drug Disco. 2009, 8, 41-54.<br />

(2) Lindsley, CW.; Niswender, CM.; Engers, DW.; Hopkins CR. Curr. Top. Med. Chem. 2009, 9, 949-963.<br />

L-09<br />

47


48<br />

Notes


PROSPECTS FOR A TGR5-MEDIATED CONTROL OF GLUCOSE<br />

HOMEOSTASIS: FOCUS ON THE CLINICAL CANDIDATE<br />

S-EMCA (INT-777)<br />

Roberto Pellicciari<br />

Dipartimento <strong>di</strong> Chimica e Tecnologia del Farmaco, <strong>Università</strong> <strong>di</strong> Perugia, Via del Liceo 1, 06123 Perugia (Italy)<br />

Beyond the regulation of lipid, glucose and their own metabolism by the nuclear receptor FXR, Bile Acids (BAs)<br />

have also been reported to activate TGR5, a GPCR receptor detected in macrophages, gallbladder and intestine,<br />

also present in lower levels in muscle and brown a<strong>di</strong>pose tissue (BAT). <strong>Through</strong> TGR5, BAs have been shown to<br />

induce energy expen<strong>di</strong>ture by controlling the activity of. Type 2 deio<strong>di</strong>nase and the consequent activation of the<br />

thyroid hormone in BAT and muscle. TGR5 has also been shown to be expressed in enteroendocrine L-cells<br />

playing a critical role in the control of the incretin glucagon-like peptide-1 (GLP-1) release and in glucose<br />

homeostasis.<br />

We have developed 6-alpha-ethy-23(S)-methyl-cholic acid (EMCA,INT-777), a semisynthetic cholic acid<br />

derivative as a potent TGR5 agonist with no FXR activity and explored its therapeutic properties. Thus, INT-777<br />

was shown to normalize glucose tolerance in obese, insulin resistant (DIO) and <strong>di</strong>abetic mice, to prevent weight<br />

gain and fat accumulation in mice on a HFD by increasing energy expen<strong>di</strong>ture and fat burning , to induce the<br />

release of GLP-1 in the gut in a TGR5-dependent manner Altogether, the data obtained confirm that TGR5<br />

represents a novel, promising therapeutic target for the treatment of metabolic <strong>di</strong>sorders, and <strong>di</strong>abesity and<br />

in<strong>di</strong>cate INT-777 as a promising can<strong>di</strong>date for the treatment of <strong>di</strong>abesity.<br />

(1) Pellicciari, R.; Gioiello, A.; Macchiarulo, A.; Thomas, C.; Rosatelli, E.; Natalini, B.; Sardella, R.; Pruzanski, M.; Roda, A.;<br />

Pastorini, E.; Schoonjans, K.; Auwerx, J. Med. Chem., 2009, 52(24), 7958-7961. (2) Thomas, C.; Gioiello, A.; Noriega, L.;<br />

Strehle, A.; Outy, J.; Rizzo, G.; Macchiarulo, A.; Yamamoto, H.; Mataki, C.; Pruzanski, M.; Pellicciari, R.; Auwerx, J.;<br />

Schoonjans, K., Cell. Metabolism, 2009, 10(3), 167-177. (3) Macchiarulo, A.; Gioiello, A.; Thomas, C.; Massarotti, A.; Nuti,<br />

R.; Rosatelli, E.; Sabbatini, P.; Schoonjans, K.; Auwerx, J.; Pellicciari, R., J. Chem. Inf. Model., 2008, 48, 1792-180. (4)<br />

Thomas, C.; Pellicciari, R.; Pruzanski, M.; Schoonjans, K.; Auwerx, J., Nat. Rev. Drug Discov., 2008, 7, 678-693.<br />

L-10<br />

49


50<br />

Notes


THE DOPAMINE D1-FAMILY: STEPS TOWARDS SELECTIVITY<br />

Christoph Enzensperger, Dina Robaa, Maria Schulze, Franziska Müller and Jochen Lehmann<br />

Institut für Pharmazie, Lehrstuhl für Pharmazeutische/Me<strong>di</strong>zinische Chemie, Friedrich-Schiller-Universität Jena,<br />

Philosophenweg 14, D-07743 Jena, Germany<br />

The dopamine D1 and the D5 receptor belong to the D1-family, are very similar and at present there are no really<br />

selective compounds available. 1 Relying on the bis-annulated mid-sized azecine-like core we synthesized and<br />

screened a set of ~300 congeners with more or less affinity and selectivity. Here we selected structures (1-4) with<br />

more <strong>di</strong>stinct selectivity further derivatized them in order to improve/work out the selectivity.<br />

H 3 CO<br />

1 2<br />

O<br />

N CH 3<br />

HO<br />

Cl<br />

2 3<br />

CH3 N<br />

H 3CO<br />

N<br />

CH3 3 4<br />

CH3 N<br />

D 1 Affinity [nM]: 35.5 0.83 2.00 3019<br />

D 5 Affinity [nM]: 1.8 0.075 0.23 >10000<br />

D 1 /D 5 Selectivity: 19.7 11 8.7


52<br />

Notes


THE ENDOCANNABINOID PROTEINS AS DRUG TARGETS<br />

Alexandros Makriyannis, Ph.D.<br />

George D. Behrakis Chair in Pharmaceutical Biotechnology<br />

Director, Center for Drug Discovery, Northeastern University<br />

The endocannabinoid biochemical system provides very promising opportunities for therapeutic intervention.<br />

Currently, the CB1 and CB2 cannabinoid receptors, as well as the two critical enzymes involved in<br />

endocannabinoid deactivation, fatty acid amide hydrolase (FAAH) and monoacyglycerol lipase (MGL), are being<br />

seriously explored as therapeutic targets. Our laboratory has been developing biochemical and biophysical<br />

methods to obtain structural and functional information on the interaction of ligands with the target proteins. Such<br />

information is used in the design and development of novel me<strong>di</strong>cations. I shall <strong>di</strong>scuss Ligand Assisted Protein<br />

Structure (LAPS), a recently developed approach combining molecular biology and mass spectroscopy-based<br />

focused proteomics. The method is currently being used to study ligand-protein interactions with both GPCRs and<br />

enzymes. Also to be <strong>di</strong>scussed are NMR methods as aids for the design of enzyme inhibitors.<br />

1 (Supported by NIH grants DA3801, DA7215, DA00493)<br />

L-12<br />

53


54<br />

Notes


THE ENDOCANNABINOID SYSTEM AND THE REGULATION OF PAIN<br />

AND EMOTION<br />

Daniele Piomelli<br />

Drug Discovery and Development, Italian Institute of Technology, Genoa, Italy<br />

Departments of Pharmacology and Biological <strong>Chemistry</strong>, University of California, Irvine, CA, USA<br />

The major psychoactive constituent of cannabis, ∆ 9 -tetrahydrocannabinol, influences pain perception and emotions<br />

in humans and laboratory animals by activating brain CB1-type cannabinoid receptors. Two primary endogenous<br />

ligands of these receptors are anandamide, the amide of arachidonic acid with ethanolamine, and 2-<br />

arachidonoylglycerol (2-AG), the ester of arachidonic acid with glycerol. Anandamide and 2-AG are released in<br />

select regions of the brain and are deactivated through a two-step process consisting of transport into cells<br />

followed by intracellular hydrolysis. Selective pharmacological inhibition of anandamide deactivation – by<br />

inhibiting either anandamide transport into cells or its intracellular hydrolysis catalyzed by fatty-acid amide<br />

hydrolase (FAAH) – produces analgesic, anxiolytic-like and antidepressant-like effects in rats. These actions are<br />

not associated with behavioral responses typical of <strong>di</strong>rect-acting cannabinoid agonists and are accompanied by<br />

profound changes in serotonergic adrenergic transmission. On the other hand, selective blockade of intracellular 2-<br />

AG hydrolysis – catalyzed by monoacylglycerol lipase – enhances stress-induced analgesia. These fin<strong>di</strong>ngs<br />

suggest that anandamide and 2-AG contribute to the regulation of pain and emotion, and that the deactivation of<br />

these endocannabinoid lipids might be the target for novel analgesic, anxiolytic and antidepressant drugs.<br />

L-13<br />

55


56<br />

Notes


NOVEL POTENT AND SELECTIVE ORL-1 ANTAGONISTS WITH<br />

EFFICACY IN ANIMAL MODELS OF PARKINSON’S DISEASE AND<br />

NEUROPATHIC PAIN<br />

Silvano Ronzoni<br />

NiKem Research srl, Via Zambeletti 25, 20021 Baranzate (MI), Italy.<br />

Since the <strong>di</strong>scovery of the NOP/ORL-1 receptor in 1994, many research groups have devoted a considerable effort<br />

in the identification of selective non-peptide ligands to better elucidate the biological role of this receptor. ORL-1<br />

antagonists in particular have been suggested to be useful, amongst other therapeutic applications, in neuropathic<br />

pain, depression, Parkinson’s <strong>di</strong>sease, food intake inhibition and learning and memory.<br />

During the course of our previous stu<strong>di</strong>es we identified a novel ORL-1 antagonist, SB-612111/NiK-10001 (1)<br />

which, notwithstan<strong>di</strong>ng a promising preliminary profile (2), was endowed with significant liabilities (mainly high<br />

hERG bin<strong>di</strong>ng affinity and low oral bioavailability) which hampered its potential as a preclinical can<strong>di</strong>date. A<br />

me<strong>di</strong>cinal chemistry programme focused at the identification of novel chemical scaffolds was therefore<br />

undertaken, resulting in the <strong>di</strong>scovery of indole derivatives possessing high affinity and selectivity for the ORL-1<br />

receptor.<br />

Detailed structure-activity relationship stu<strong>di</strong>es aimed at the optimisation of this new chemical class, with particular<br />

focus on early ADME parameters, will be presented. This investigation led to the identification of NiK-21273, a<br />

potent and selective ORL-1 antagonist with efficacy in animal models of neuropathic pain and Parkinson’s<br />

<strong>di</strong>sease. These results confirmed that ORL-1 antagonists could be of clinical relevance for the treatment of<br />

intractable neuropathic pain and for the symptomatic therapy of parkinsonism.<br />

(1) Zaratin, P.F.et al. J.Pharmacol. Exp. Ther. 2004, 308, 454-461.<br />

(2) Rizzi, A.et al. J.Pharmacol. Exp. Ther. 2007, 321, 968-974.<br />

L-14<br />

57


58<br />

Notes


ARE HETEROMERIC G PROTEIN-COUPLED RECEPTORS<br />

RELEVANT TO IN VIVO PHARMACOLOGY?<br />

OPIOID RECEPTORS AS A CASE IN POINT<br />

Philip S. Portoghese<br />

University of Minnesota, Department of Me<strong>di</strong>cinal <strong>Chemistry</strong>, Minneapolis, MN 55455<br />

Burgeoning evidence for <strong>di</strong>mers of G protein-coupled receptors (GPCRs) in cultured cells together with the<br />

fin<strong>di</strong>ng that signaling and trafficking may be mo<strong>di</strong>fied by such association, raises the likelihood that homomers<br />

and heteromers and may generally be the targets of most drugs that activate or antagonize GPCRs. Until relatively<br />

recently the in vivo relevance of heteromers in the class A GPCR family was not established because both the<br />

assay technology and selective ligands for heteromers were not available. One such class A GPCR family is the<br />

opioid receptors and, in this regard, over a dozen heteromers have been reported in cultured cells. In an effort to<br />

bridge the gap between cultured cells, experimental animals, and humans, we have designed a number of ligands<br />

that potently and selectively activate <strong>di</strong>fferent heteromeric opioid receptors. Support for the involvement of<br />

heteromers in the action of clinically employed opioid analgesics such as morphine, methadone, fentanyl,<br />

buprenorphine, and nalbuphine has been obtained in culutured cells that contain heteromeric or homomeric opioid<br />

receptors. The fin<strong>di</strong>ng that some of these clinically employed ligands are devoid of physical dependence while<br />

others produce dependence is related to the specific heteromers that are activated. The screening of ligands on both<br />

heteromeric and homomeric GPCRs in cultured cells offers an approach to develop potent analgesics without<br />

significant side effects.<br />

L-15<br />

59


60<br />

Notes


STABILIZED GPCRs FOR STRUCTURE BASED DRUG DESIGN<br />

Miles Congreve<br />

Heptares Therapeutics, BioPark, Welwyn Garden City, UK<br />

Heptares has developed a technology that facilitates the study of G-protein-coupled receptors (GPCRs) by<br />

dramatically stabilizing these important proteins outside of the cell membrane. The new stabilized human<br />

receptors (StaRs) are much more robust than the correspon<strong>di</strong>ng wild type proteins; they are amenable to<br />

crystallography, biophysical/fragment screening and for raising monoclonal antibo<strong>di</strong>es. The process whereby the<br />

StaRs are first engineered will be explained. The potential of this new technology for structure-based drug design<br />

and biophysical screening of GPCR targets will be presented. The Heptares virtual and fragment screening hit<br />

identification approaches will be outlined with some example results against the A2A receptor target illustrated.<br />

A2A is a validated therapeutic target for treatment of Parkinson’s <strong>di</strong>sease and also of interest for<br />

neurodegeneration. Leads with good ligand efficiency and drug-like properties have been identified from virtual<br />

screening and have been progressed to can<strong>di</strong>date selection stu<strong>di</strong>es in less than 12 months.<br />

L-16<br />

61


62<br />

Notes


COMPUTER-ASSISTED LEAD FINDING OF GPCR MODULATORS<br />

Karl-Heinz Baringhaus, Andreas Evers, Clemens Giegerich, Robert Jaeger, Thomas Klabunde<br />

Sanofi-Aventis Deutschland GmbH, CAS Drug Design, Buil<strong>di</strong>ng G 878, 65926 Frankfurt am Main, Germany<br />

Computer-assisted lead fin<strong>di</strong>ng is a knowledge-driven strategy taking into account public and proprietary chemical<br />

and biological data (e.g. compounds, 3D target structures, biological activities). Buil<strong>di</strong>ng a target family related<br />

chemical and biological space enables lead fin<strong>di</strong>ng by linking chemotypes to biological targets.<br />

The chemical space of a target family is generated through buil<strong>di</strong>ng databases of biologically active compounds,<br />

privileged fragments, frameworks and pharmacophores. The biological space is comprised through protein<br />

sequences, ligand recognition motifs, 3D structures, 3D pharmacophores and biological activities.<br />

This presentation contains two <strong>di</strong>fferent methods for GPCR lead fin<strong>di</strong>ng: 1. All family A GPCRs are described via<br />

three physicochemical properties (Wold scales) of 39 amino acids lining the retinol-like bin<strong>di</strong>ng site. The resulting<br />

matrix of 39*3 descriptors for each of the 270 GPCRs can be <strong>di</strong>splayed in an in-house tool termed GPCR-<br />

Navigator and gives access to a quantitative similarity description of GPCR pairs. In ad<strong>di</strong>tion, the tool provides<br />

information on in-house and external GPCR ligands allowing the identification of ligated GPCRs for orphan<br />

receptors as starting point for chemogenomics-based virtual screening. Validation of this GPCR bin<strong>di</strong>ng site<br />

classification was achieved by a retrospective study revealing its applicability for GPCR lead fin<strong>di</strong>ng. The<br />

likelihood of success is linked to the GPCR-Navigator defined similarity and this will be exemplified by a recent<br />

successful identification of a nanomolar GPCR agonist. 2. The second approach, termed Pharma 3D provides<br />

sequence-derived 3D-pharmacophore models for GPCRs suitable for virtual screening. This method captures<br />

pharmacophoric features for ligand recognition identified from 13 <strong>di</strong>fferent GPCR-ligand structures (3 X-ray<br />

structures, 10 homology models). In total, 35 molecular interaction partners (chemoprints) were identified each<br />

enco<strong>di</strong>ng the location and identity of a single-feature pharmacophore. This information together with the link to<br />

the correspon<strong>di</strong>ng amino acid sequence motif is stored in a pharmacophore buil<strong>di</strong>ng block database. A family A<br />

GPCR can be searched for the presence of such chemoprints in our database and all identified single-feature<br />

pharmacophores are then automatically assembled to a sequence-derived 3D-pharmacophore model. A successful<br />

application of this Pharma 3D approach yiel<strong>di</strong>ng the <strong>di</strong>scovery of potent agonists of the complement component 3a<br />

receptor 1 (C3AR1) underpins the broad applicability of this concept (1).<br />

(1) Klabunde, T. ; Giegerich, C.; Evers, A. J. Med. Chem. 2009, 52, 2923-2932.<br />

L-17<br />

63


64<br />

Notes


REGULATION OF 5-HT RECEPTOR FUNCTIONS BY P11<br />

Per Svenningsson 1 , Paul Greengard 2 , Jennifer Warner-Schmidt 2 , Xiaoqun Zhang 1 , Karl Björk 1<br />

1 Karolinska Institutet, Stockholm, Sweden; 2 Rockefeller University, New York, USA<br />

P11 (S100A10) is a member of a large family of S100 proteins that interacts with serotonin receptor 1B<br />

(5-HTR1B) and serotonin receptor 4 (5-HTR4), modulates signal transduction via 5-HT1B and 5-HTR4 receptors<br />

(1, 2). p11 increases 5-HT1B and 5-HTR4 surface expression and facilitates signaling via these receptors. p11 and<br />

5-HTR1B/5-HTR4 mRNAs and proteins are co-expressed in several brain regions. p11 is down-regulated in post-<br />

mortem tissue from depressed in<strong>di</strong>viduals, whereas at least two classes of antidepressants (imipramine and<br />

electroconvulsive seizures) increase p11 . P11 knockout mice have a depression-like phenotype and p11 is<br />

required for the behavioral antidepressant responses to 5-HT1B and 5-HTR4 stimulation as well as citalopram and<br />

imipramine in vivo. Likewise, transgenic over-expression of p11 in mice produces antidepressant-like effects in<br />

rodent models of depression. P11 is also involved in L-DOPA-me<strong>di</strong>ated actions in Parkinsonian animals (3).<br />

References<br />

1. Svenningsson P, Chergui K, Rachleff I, Flajolet M, Zhang X, El Yacoubi M, Vaugeois JM, Nomikos GG, Greengard<br />

P. Alterations in 5-HT1B receptor function by p11 in depression-like states. Science. 2006 Jan 6;311(5757):77-80.<br />

2. Warner-Schmidt JL, Flajolet M, Maller A, Chen EY, Qi H, Svenningsson P, Greengard P. Role of p11 in cellular and<br />

behavioral effects of 5-HT4 receptor stimulation. J Neurosci. 2009 Feb 11;29(6):1937-46.<br />

3. Zhang X, Andren PE, Greengard P, Svenningsson P. Evidence for a role of the 5-HT1B receptor and its adaptor<br />

protein, p11, in L-DOPA treatment of an animal model of Parkinsonism. Proc Natl Acad Sci U S A. 2008 Feb<br />

12;105(6):2163-8.<br />

L-18<br />

65


66<br />

Notes


ALLOSTERIC MODULATION OF NEURONAL NICOTINIC<br />

ACETYLCHOLINE RECEPTORS<br />

Marvin K. Schulte<br />

University of Alaska Fairbanks, Department of <strong>Chemistry</strong> and Biochemistry, Fairbanks, AK 99775<br />

Nicotinic acetylcholine receptors (nAChR) are involved in me<strong>di</strong>ating acetylcholine based signal transmission in<br />

the CNS and have been implicated in a broad range of CNS <strong>di</strong>sorders inclu<strong>di</strong>ng Alzheimer’s <strong>di</strong>sease (1, 2), Autism<br />

(3, 4), Parkinson’s <strong>di</strong>sease (5) and Schizophrenia (6-8). Treatment strategies aimed at increasing activity of<br />

cholinergic systems have until recently focused on acetylcholinesterase inhibitors and partial agonists. Efforts to<br />

develop novel means of modulating nicotinic receptors through allosteric effectors have led to the development of<br />

new classes of nicotinic agents termed positive allosteric modulators (PAMs.) PAMs represent an alternative<br />

treatment strategy since they typically act to enhance responses to endogenous agonist but do not by themselves<br />

elicit responses. Much recent work has focused on α7 nAChR PAMs with a lesser number of stu<strong>di</strong>es reporting<br />

compounds that selectively modulate other nAChR subtypes. We have reported the synthesis of a novel type II<br />

PAM selective for the α4β2 nAChR subtype (9, 10). Co-application of dFBr with acetylcholine produces a<br />

biphasic dose response curve with a peak potentiation of over 265% at dFBr concentrations 10 µM. While dFBr does not appear to alter the action of nAChR antagonists, it<br />

is capable of significantly altering the behavior of partial agonists inclu<strong>di</strong>ng nicotine. The evaluation of dFBr, its<br />

putative mechanism of action and its potential use as a therapeutic agent in the treatment of <strong>di</strong>sorders involving<br />

nAChR me<strong>di</strong>ated synaptic transmission will be <strong>di</strong>scussed.<br />

(1) Nordberg A. 2001. Biol Psychiatry 49: 200-10<br />

(2) Court J, Martin-Ruiz C, Piggott M, Spurden D, Griffiths M, Perry E. 2001. Biol Psychiatry 49: 175-84<br />

(3) Martin-Ruiz CM, Lee M, Perry RH, Baumann M, Court JA, Perry EK. 2004. Brain Res Mol Brain Res 123:<br />

81-90<br />

(4) Lippiello PM. 2006. Med Hypotheses 66: 985-90<br />

(5) Aubert I, Araujo DM, Cecyre D, Robitaille Y, Gauthier S, Quirion R. 1992. J Neurochem 58: 529-41<br />

(6) Adams CE, Stevens KE. 2007. Front Biosci 12: 4755-72<br />

(7) Friedman JI. 2004. Psychopharmacology (Berl) 174: 45-53<br />

(8) Woodruff-Pak DS, Gould TJ. 2002. Behav Cogn Neurosci Rev 1: 5-20<br />

(9) Sala F, Mulet J, Reddy KP, Bernal JA, Wikman P, et al. 2005. Neurosci Lett 373: 144-9<br />

(10) Kim JS, Padnya A, Weltzin M, Edmonds BW, Schulte MK, Glennon RA. 2007. Bioorg Med Chem Lett 17:<br />

4855-60<br />

L-19<br />

67


68<br />

Notes


DUALSTERIC GPCR-TARGETING: FINE-TUNING BINDING<br />

SELECTIVITY AND SIGNALLING PATHWAY ACTIVATION<br />

Marco De Amici 1 , Elisabetta Barocelli 2 , Ulrike Holzgrabe 3 , Evi Kostenis 4 , Christian Tränkle 5 , Klaus Mohr 5<br />

1 Dipartimento <strong>di</strong> Scienze Farmaceutiche “Pietro Pratesi”, <strong>Università</strong> <strong>degli</strong> <strong>Stu<strong>di</strong></strong> <strong>di</strong> Milano, 20133 Milano, Italy;<br />

2 Department of Pharmacological, Biological and Applied Chemical Sciences, University of Parma, Parma 43100, Italy<br />

3 Department of Pharmaceutical <strong>Chemistry</strong>, Institute of Pharmacy, Julius-Maximilians-University, D-97074 Würzburg,<br />

Germany;<br />

4 Institute of Pharmaceutical Biology and 5 Pharmacology & Toxicology Section, Institute of Pharmacy, Rheinische Friedrich-<br />

Wilhelms-University, D-53121 Bonn, Germany.<br />

An increasing number of G protein-coupled receptors (GPCRs) is recognized to harbor an allosteric bin<strong>di</strong>ng area<br />

in ad<strong>di</strong>tion to the orthosteric site for bin<strong>di</strong>ng of the endogenous messenger substance. If both sites are located in<br />

spatial proximity, ligand molecules should be achievable that bind in an overlap-fashion to both sites<br />

simultaneously. The muscarinic acetylcholine receptor subtypes (M subtypes) are prototypal for the exploration of<br />

allosteric interactions at GPCRs and rather deep insight has been gained in the recent years into the topography of<br />

orthosteric and allosteric ligand bin<strong>di</strong>ng. To explore the feasibility of the dualsteric (allosteric/orthosteric) design<br />

concept, we fused M2-preferring alkane-bis-ammonio-type allosteric buil<strong>di</strong>ng blocks, which are devoid of<br />

agonistic activity, with orthosteric oxotremorine-like non-selective agonists. <strong>Receptor</strong> bin<strong>di</strong>ng and functional<br />

stu<strong>di</strong>es revealed M2-preferring agonism of some of the hybrid molecules and proved true dualsteric receptor<br />

bin<strong>di</strong>ng. Noteworthy, selectivity for the Gi-signaling pathway is achieved by these dualsteric agonists, whereas<br />

conventional orthosteric agonists such as acetylcholine also activate Gs in ad<strong>di</strong>tion to Gi ((1) and references<br />

therein, (2) for review). Taken together, the design concept of dualsteric agonists is a promising new avenue to<br />

adjust receptor subtype selectivity, efficacy and signaling pathway specificity of GPCR-activating drugs.<br />

(1) Antony, J.; Kellershohn, K.; Mohr-Andrä, M. et al. FASEB J. 2009, 23, 442-450.<br />

(2) Mohr, K.; Tränkle, C.; Kostenis, E.; Barocelli, E.; De Amici, M.; Holzgrabe, U.; Brit. J. Pharmacol., in press.<br />

L-20<br />

69


70<br />

Notes


POLYPHARMACOLOGY: USING IT TO DIFFERENTIATE AND AS AN<br />

ADVANTAGE WITH SELECTIVE NON-SELECTIVITY<br />

Jonathan S. Mason 1,2<br />

1 Heptares Therapeutics Ltd, BioPark, Broadwater Road, Welwyn Garden City, Herts, AL7 3AX, UK; 2 Lundbeck Research,<br />

Ottiliavej 9, Valby, Copenhagen, DK-2500, Denmark<br />

Polypharmacology is a key concept in the drug <strong>di</strong>scovery process. It is a two-edged sword, where it may be<br />

needed for the effective action of some drugs, but undesirable activities can lead to side effects and toxicity that<br />

limit the use of, or cause the attrition of, an otherwise effective me<strong>di</strong>cine. It does however offer a unique way to<br />

describe and <strong>di</strong>fferentiate molecules, based not on the chemical structure or any derived properties, but on how<br />

biological targets see the molecule, by using for example a biological “fingerprint” created by measuring the<br />

bin<strong>di</strong>ng affinity of a molecule to a large <strong>di</strong>verse set of relevant biological targets.<br />

In recent years it has become possible to understand much better the extent of polypharmacology present in<br />

me<strong>di</strong>cinal chemistry compounds and the resultant drug can<strong>di</strong>dates. Two major efforts that will be <strong>di</strong>scussed,<br />

highlighting some key fin<strong>di</strong>ngs, are: 1) The Cerep BioPrint® initiative wherein drugs, related reference<br />

compounds, project compounds and attrited compounds (those that failed during development or after launch)<br />

were profiled in a full-matrix manner against 180 pharmacological and ADME targets, with full dose response<br />

measurements for all compounds that had >30% inhibition at 10µM (~4000 compound dataset) and 2) Organized<br />

and integrated databases of structure-activity data (e.g. published, in-house, BioPrint®) that cover much larger<br />

numbers of compounds and targets (e.g. the ChEMBL database has >0.6 million compounds, 7.2 thousand targets<br />

and 2.4 million activities).<br />

Polypharmacology is of particular importance for many GPCR (G-protein-coupled receptor) drugs, and now the<br />

action of ligands can be probed and analyzed at the molecular level following the recent <strong>di</strong>scovery of methods that<br />

enable the stabilization of GPCRs (StaR- Stabilized <strong>Receptor</strong> technology) and thence two key approaches: 1)<br />

crystallization to yield high resolution X-ray structures with both high and low affinity ligands and 2) biophysical<br />

screening (e.g. Biacore/SPR, TINS/NMR) for kinetics, fragment screening and using site-<strong>di</strong>rected mutants<br />

Biophysical Mapping (Heptares) to probe bin<strong>di</strong>ng modes/interactions. This stabilization of the receptor is<br />

transformational for GPCR drug research, and enabled the pivotal X-ray structure of the beta-1 adrenergic receptor<br />

with cyanopindolol. The application of the StaR technology platform enables <strong>di</strong>fferent conformations (agonist,<br />

antagonist…) to be stabilized. Rational approaches to addressing polypharmacology (desired and undesired) are<br />

now possible based upon the target protein bin<strong>di</strong>ng site and not just ligand-based activity models.<br />

L-21<br />

71


72<br />

Notes


MULTI-TARGET DRUG DISCOVERY - PAST, PRESENT AND FUTURE<br />

Richard Morphy<br />

Schering-Plough, a part of MSD, Newhouse, Lanarkshire, ML1 5SH. UK.<br />

Currently, drug <strong>di</strong>scovery is largely based upon a “one molecule - one target - one <strong>di</strong>sease” philosophy and many<br />

successful drugs, that are selective for a single target (“magic bullets”), have emerged from this strategy. There is<br />

however an increasing interest in and awareness of multi-target drug <strong>di</strong>scovery (MTDD) within the me<strong>di</strong>cinal<br />

chemistry community as a means of provi<strong>di</strong>ng a superior therapeutic effect and side effect profile compared to the<br />

modulation of a single target. The inherent redundancy and robustness that exists within biological networks and<br />

pathways means that modulating single proteins may not be sufficient to produce the desired efficacy, whilst at the<br />

same time minimising adverse side effects. It has been demonstrated that a balanced modulation of multiple<br />

targets in parallel can be beneficial for treating <strong>di</strong>seases with complex etiologies, such as using multi-kinase<br />

inhibitors for treating cancer and multi-target monoaminergic ligands for treating CNS <strong>di</strong>seases.<br />

Although many currently marketed drugs act via multiple targets, the <strong>di</strong>scovery of their polypharmacological<br />

mechanism of action was usually seren<strong>di</strong>pitous and retrospective. The preme<strong>di</strong>tated design of compounds with a<br />

predefined multi-target profile (i.e. “designed” multiple ligands or DMLs) is a relatively recent development. The<br />

combination of the desirability of <strong>di</strong>scovering multiple ligands on the one hand, and the increasing tractability of a<br />

rational approach on the other, has lead to a strengthening of interest in this area in recent years. Designing<br />

multiple ligands presents me<strong>di</strong>cinal chemists with many challenges and an early assessment of the feasibility of<br />

the approach for a particular target combination is essential. A key objective is to achieve a balanced activity for<br />

each target of interest, whilst obtaining wider selectivity and a suitable oral pharmacokinetic profile.<br />

Historically, there have been two quite <strong>di</strong>fferent methods of generating lead compounds, screening approaches that<br />

rely largely upon seren<strong>di</strong>pity and knowledge-based approaches that exploit information either from the general<br />

literature or proprietary information from within an organization. New strategies to help me<strong>di</strong>cinal chemists<br />

<strong>di</strong>scover the next generation of DMLs will also be described.<br />

L-22<br />

73


74<br />

Notes


MULTIPLE MECHANISM DRUGS FOR NEURODEGENERATIVE<br />

DISEASES AND POST-STROKE NEUROPROTECTION<br />

Cornelis J. (Neels) Van der Schyf<br />

Departments of Pharmaceutical Sciences, and Neurobiology, Northeastern Ohio Universities Colleges of Me<strong>di</strong>cine and<br />

Pharmacy, Rootstown, OH, 44313 USA.<br />

Transcriptomics and proteomics profiling suggest that the <strong>di</strong>sease etiologies of neurodegenerative <strong>di</strong>sorders and<br />

post-stroke neurodegeneration are multifactorial in origin. As such, these pathologies offer complex challenges to<br />

drug <strong>di</strong>scovery efforts aimed at preventing or even reversing their progression. In response to this complexity, a<br />

new para<strong>di</strong>gm has emerged which challenges the widely held assumption that ‘silver bullet’ agents are superior to<br />

‘<strong>di</strong>rty drugs’ in therapeutic approaches aimed at the prevention or treatment of these <strong>di</strong>seases. In stroke,<br />

neuroprotective strategies have typically focused on the neurotoxic ischemic environment that threatens neuronal<br />

cells. It has now become clear that the complex processes that occur post-stroke call for the targeting of multiple<br />

pathobiological events (apoptotic and necrotic cell death and inflammatory processes among others) and a<br />

<strong>di</strong>versity of cells that include glia, vascular epithelial, and inflammatory cells. While the core or umbral area of<br />

ischemic damage in stroke likely cannot be saved from cellular death, the penumbral area surroun<strong>di</strong>ng the core,<br />

despite being burdened with severely compromised blood flow, may be preserved either by reperfusion or by the<br />

use of neuroprotective agents. A neuroprotective approach is attractive clinically because, if left untreated, the<br />

penumbral region ultimately will be incorporated in the umbral core, and the lesion will enlarge over time. In this<br />

presentation, I will report on new multimodal drug can<strong>di</strong>dates developed in our laboratory that target dual calcium<br />

entry pathways, i.e. both the L-type calcium channel and the NMDA receptor channel. The effect of one can<strong>di</strong>date<br />

compound was measured after permanent, and transient (1 h) focal cerebral ischemia in the murine middle cerebral<br />

artery occlusion (MCAO) model. In the permanent MCAO model, infarct area (visualized by TTC staining) was<br />

reduced by 42.6% (P < 0.05) compared to vehicle (DMSO)-treated controls, and brain swelling by 78.3% (P <<br />

0.001), while also in the transient MCAO model, significantly less brain damage (visualized by Fluoro-Jade<br />

staining, 13±7 mm 3 , P < 0.01) was seen in treated animals compared with two control groups (no treatment: 47±4<br />

mm 3 ; vehicle, DMSO: 50±10 mm 3 ). These results, hitherto unexplored drug targets for post-stroke<br />

neuroprotection, and future <strong>di</strong>rections in multiple mechanism drug <strong>di</strong>scovery for neurodegenerative <strong>di</strong>seases will<br />

be <strong>di</strong>scussed.<br />

L-23<br />

75


76<br />

Notes


NOVEL THERAPEUTIC APPROACHES CONSTITUTING<br />

MULTIFUNCTIONAL NEUROPROTECTIVE AND<br />

NEURORESTORATIVE DRUGS FOR NEURODEGENERATIVE<br />

DISEASES<br />

Moussa B.H. You<strong>di</strong>m<br />

Eve Topf and US National Parkinson Foundations Centers of Excellence for Neurodegenerative Diseases<br />

Research and Teaching Technion-Rappaport Faculty of Me<strong>di</strong>cine, Haifa; Israel and Varinel Inc. and Varinel<br />

LCD (USA-Israel)<br />

Alzheimer’s <strong>di</strong>sease (AD) and Parkinson’s <strong>di</strong>sease (PD), Huntington <strong>di</strong>sease (HD) , amyotrophic lateral sclerosis<br />

(ALS) and multiple Sclerosis (MS) are initiated by cascade of neurotoxic events, that includes oxidative stress,<br />

brain iron dysregulation, glutamate excitotoxicity, nitric oxide, inflammatory process, neurotxic processing<br />

misfol<strong>di</strong>ng and aggregation of abeta-peptide, alpha-synuclein resulting from the possible demise of ubiquitin-<br />

proteasome system (UPS) as demonstrated neurochemically and by transcriptomics and proteomic profiling. AD<br />

and PD subjects are benefiting from symptomatic effects of cholinesterase inhibitors, memantine, monoamine<br />

oxidase A and B inhibitors, L-dopa, dopamine receptor agonists, COMT inhibitors and dopamine agonists,<br />

glutame antagonist (memantine),anti viral drug amanta<strong>di</strong>ne developed to act on a single molecular target. For HD<br />

and ALS there are no effective drugs. Such drugs have limited symptomatic activities and current<br />

pharmacological approaches have sever limitation in their ability to mo<strong>di</strong>fy the course of the <strong>di</strong>sease, offering<br />

incomplete and transient benefit to patients. However, the new therapeutic strategies for neurodegenerative<br />

<strong>di</strong>seases are those in which drug can<strong>di</strong>dates are designed expressly to act on multiple neural and biochemical<br />

targets involved in the neurodegenerative process and to possesses neuropotective and neurorestorative activities.<br />

Thus we have hypothesized and developed a series of innovative novel multifunctional drugs from our successful<br />

neuroprotectiev <strong>di</strong>sease mo<strong>di</strong>fying anti Parkinson drug, rasagiline (Azilect, FDA approved 2006), called ladostigil<br />

(phase II clinical stu<strong>di</strong>es), M30 and HLA-20. These drugs possess neuroprotective (anti apoptotic) and<br />

neurorestorative activities for AD and PD. There were achieved with the use of multifunctional moieties that<br />

target an array of pathological pathways, each of which is believed to contribute to the cascade that ultimately<br />

leads to neuronal cell death. These moieties include cholinesterase and monoamine oxidase inhibitory activities,<br />

iron chelating , ra<strong>di</strong>cal scavenging, APP processing to non amyloigenic neuroprotective neurotrophic, soluble<br />

APP alpha, inductions of GDNF, BDNF and HIF (hypoxia inducing factor),VEGF (vascular endothelial growth<br />

factor) and prevention entry into cell cycle via inhibition of G o / G 1 and cycline D. These drugs convert more<br />

than 85% of human embryonic stem cells in to neurons and <strong>di</strong>fferentiate several other cells into neurons in<br />

culture. Multifunctional drugs have advantage over the problem of so called monomodal “Magic Bullets” that act<br />

on one neurotoxic modality of neurodegenerative <strong>di</strong>seases, where there are cascade of neurotoxic event and<br />

pathologies (You<strong>di</strong>m and Van Der Schyf, 2009).<br />

You<strong>di</strong>m, MBH and Van Der Schyf, C (E<strong>di</strong>tors) (2009) Multifunctional Drugs as Neurotherapeutics, Neurotherapeutics 6; 1-<br />

211.<br />

L-24<br />

77


78<br />

Notes


CENTRAL OREXIN PEPTIDERGIC SYSTEM AS TARGET FOR NOVEL<br />

TREATMENTS OF INSOMNIA, ANXIETY AND DRUG ADDICTION:<br />

RECENT FINDINGS FROM THE GSK DISCOVERY PERFORMANCE<br />

UNIT<br />

E. Merlo Pich, A. Gozzi, A. Bifone, L. Piccoli, M. Massagrande, P. Martinelli, D Amantini, D. Montanari, S.<br />

Zamuner, P. Bettica, R. Di Fabio, M. Corsi, E. Ratti.<br />

Ad<strong>di</strong>ction & Sleep Disorder DPU, Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline Me<strong>di</strong>cines<br />

Research Centre, Verona, Italy.<br />

Orexin/hypocretin neuropeptides (A and B) bind to two G-protein coupled receptors, OX1 and OX2 receptors that<br />

are widely <strong>di</strong>stributed in the mammalian brain with only partially overlapping patterns. A large body of pre-<br />

clinical and clinical evidence suggest that the orexin system is deeply implicated in the sleep <strong>di</strong>sorders, in<br />

particular insomnia, as well as in modulating the reward pathways and anxiety. As part of our internal drug<br />

<strong>di</strong>scovery program aimed towards the identification of potent, selective and bioavailable orexin receptor<br />

antagonists, several preclinical tests and innovative compounds were developed to profile the novel molecules and<br />

to pre<strong>di</strong>ct possible therapeutic roles. These investigations led to the filing of several patents and assessment of<br />

pre-can<strong>di</strong>date compounds to identify high quality molecules to be progressed in human stu<strong>di</strong>es. The use of some of<br />

these compounds as exploratory tools produced evidence of a <strong>di</strong>fferential regulatory role for OX1 receptors vs.<br />

OX2 receptors on behavioural and neurobiologic outcomes. A particular case of translational approach linking<br />

preclinical and clinical data on sleep control will be presented for SB-649868, currently in Phase II, and<br />

GSK1059865, a novel OX1 selective tool compound.<br />

L-25<br />

79


80<br />

Notes


BIS-AMIDO PIPERIDINE DERIVATIVES AS IN VITRO AND IN VIVO<br />

POTENT DUAL OREXIN RECEPTOR ANTAGONISTS<br />

R. Di Fabio, 1 P. Gerrard, 1 R. Porter, 2 G. Stemp, 2 D. Nash, 2 C. Branch, 2 M. Massagrande, 1 A. Poffe, 1 L. Piccoli, 1<br />

S. Braggio, 1 E. Ratti. 1<br />

1 Neurosciences Centre of Excellence for Drug Discovery, GlaxoSmithKline Me<strong>di</strong>cines Research Centre, Verona, Italy;<br />

2 Neurosciences Centre of Excellence for Drug Discovery, New Frontiers Science Park, Harlow, UK.<br />

The hypothalamic peptides orexin A and orexin B are potent agonists of two G-protein coupled receptors, namely<br />

OX1R and OX2R. These receptors are widely <strong>di</strong>stributed, though <strong>di</strong>fferentially, in the rat brain. In particular,<br />

OX1R was found to be highly present throughout the hypothalamus, whilst OX2R was found mainly in the VPN.<br />

A large body of compelling pre-clinical and clinical evidence suggest that the orexin system is deeply implicated<br />

in the sleep <strong>di</strong>sorders. In particular, modulation of the orexin receptors by appropriate antagonists was proven to<br />

be an efficacious strategy for treatment of insomnia in humans.<br />

As part of our internal drug <strong>di</strong>scovery program aimed towards the <strong>di</strong>scovery potent and selective druggable orexin<br />

receptor antagonists, novel bis-amido piperi<strong>di</strong>ne derivatives were identified as in vitro potent dual orexin receptor<br />

antagonists showing hypnotic properties in multiple pre-clinical models. The structure-activity relationship of this<br />

new chemical series as well as the profile of the best exemplars selected will be <strong>di</strong>scussed.<br />

L-26<br />

81


82<br />

Notes


DISCOVERY OF POTENT, DIAZEPANE-CONTAINING DUAL OREXIN<br />

RECEPTOR ANTAGONISTS FOR THE TREATMENT OF INSOMNIA<br />

Anthony J. Roecker 1 ,* Christopher D. Cox 1 , John D. Schreier 1 , Swati P. Mercer 1 , Michael J. Breslin 1 , David B.<br />

Whitman 1 , Jeffrey M. Bergman 1 , Karen M. Brashear 1 , Georgia McGaughey 1 , Michael Bogusky 1 , Scott M. Doran 2 ,<br />

Christopher J. Winrow 2 , Anthony Gotter 2 , Susan L. Garson 2 , Steve V. Fox 2 , Richard L. Kraus 2 , C. Meacham<br />

Harrell 2 , Duane R. Reiss 2 , Kristina Groover 2 , Rodney A. Bednar 4 , Wei Lemaire 4 , Joseph G. Bruno 4 , Kathy L.<br />

Murphy 2 , Chunze Li 3 , Cuyue Tang 3 , Dan Cui 3 , Tamara Cabalu 3 , Thomayant Prueksaritanont 3 , Ken S. Koblan 2 ,<br />

Steven D. Young 1 , George D. Hartman 1 , John J. Renger 2 , and Paul J. Coleman 1<br />

Departments of 1 Me<strong>di</strong>cinal <strong>Chemistry</strong>, 2 Neuroscience Research, 3 Drug Metabolism, and 4 The West Point Facility for<br />

Automation and Screening Technology (WP FAST), Merck Research Laboratories, WP14-2, P.O. Box 4, Sumneytown Pike,<br />

West Point, Pennsylvania 19486<br />

Orexin antagonists have been identified as possible treatments for a number of<br />

in<strong>di</strong>cations inclu<strong>di</strong>ng ad<strong>di</strong>ction, psychiatric in<strong>di</strong>cations, and insomnia. Clinical proof-of-<br />

concept has been achieved using dual orexin receptor antagonists (DORAs) for the treatment of<br />

insomnia. This milestone has piqued interest in orexin antagonist research from academic and<br />

industrial laboratories worldwide. Merck has recently <strong>di</strong>sclosed the <strong>di</strong>scovery and preliminary<br />

optimization of DORAs with a 1,4-<strong>di</strong>azepane central constraint. This presentation will focus on<br />

the optimization of the metabolic profile and physical properties of this series of compounds<br />

though core and peripheral heterocycle replacements. The in vivo activity of optimized<br />

compounds in mouse and rat sleep experiments will also be <strong>di</strong>scussed.<br />

L-27<br />

83


84<br />

Notes


OREXIN RECEPTOR ANTAGONISTS – A NEW THERAPEUTIC<br />

PRINCIPLE IN CNS DISORDERS?<br />

Christoph Boss, Ralf Koberstein, Hamed Aissaoui, Thierry Sifferlen, Markus Gude, Romain Siegrist, Jo<strong>di</strong><br />

Williams, Christine Brotschi, Martin Bolli, Thomas Weller, Catherine Brisbare-Roch, Francois Jenck, Alexander<br />

Treiber, John Gatfield<br />

Actelion Pharmaceuticals Ltd, Gewerbestrasse 16, CH-4123 Allschwil, Switzerland<br />

Orexins/hypocretins are hypothalamic neuropeptides that play a role in regulating vigilance in mammals, as<br />

suggested by the deficits in orexinergic function that are associated to rodent, canine and human narcolepsy.<br />

However, orexinergic fibers widely <strong>di</strong>stribute in the brain, suggesting that orexins exert broad homeostatic<br />

functions, with a particular role as regulators of behavioral arousal and wake states relating to stress and reward.<br />

Manipulation of the orexin system using brain-penetrant orexin receptor antagonists, or partial, or inverse agonists<br />

may prove therapeutically useful in the treatment of me<strong>di</strong>cal and psychiatric con<strong>di</strong>tions associated with <strong>di</strong>sturbed<br />

orexin circuitry in the brain. Selective or dual orexin1-receptor and/or orexin2-receptor ligands (examples given in<br />

Scheme 1 below) that cross the blood-brain-barrier and transiently mo<strong>di</strong>fy orexinergic function are of key<br />

pharmacological interest for scientific and me<strong>di</strong>cal investigations. This presentation summarizes research to<br />

identify and characterize orexin receptor ligands and their biome<strong>di</strong>cal potential for understan<strong>di</strong>ng the physiological<br />

and pathophysiological roles of endogenous orexins.<br />

O<br />

O<br />

N<br />

O<br />

N<br />

H<br />

CF3 Almorexant [1]<br />

N<br />

SB-334867<br />

(dual Ox1/Ox2-receptor antagonist;<br />

Clinical Phase 3; Actelion Phramaceuticals Ltd)<br />

[2]<br />

(selective Ox1-receptor antagonist)<br />

N<br />

HN<br />

HN O<br />

Scheme 1: Examples of orexin receptor antagonists with <strong>di</strong>fferent selectivity profiles.<br />

N<br />

O<br />

Br<br />

Br<br />

N<br />

H<br />

O<br />

N<br />

H<br />

JNJ-10397049 [3]<br />

selective Ox2-receptor antagonist)<br />

[1] Dingemanse, J.; Dorffner, G.; Hajak, G.; Benes, H.; Danker-Hopfe, H.; Polo, O.; Saletu, B.; Barbanoj, M. J.; Pillar, G.;<br />

Penzel, T.; Chiossi, E.; Hoever, P. Proof-of-concept study in primary insomnia patients with almorexant (ACT-078573), a dual<br />

orexin receptor antagonist. Sleep and Biological rhythms 2007, 5, A-194.<br />

[2] Smart, D.; Sabido-David, C.; Brough, S. J.; Jewitt, F.; Johns, A.; Porter, R. A.; Jerman, J. C. SB-334867-A: the first<br />

selective orexin-1 receptor antagonist. Br J Pharmacol 2001, 132, 1179-1182.<br />

[3] Dugovic, C.; Shelton, J.; Sutton, S.; Yun, S.; Li, X.; Dvorak, C.; Carruthers, N.; Atack, J.; Lovenberg, T. Sleep-inducing<br />

effects me<strong>di</strong>ated by selective blockade of orexin OX2 receptors during the light phase in the rat. Sleep 2008, 31, A33.<br />

L-28<br />

O<br />

O<br />

85


86<br />

Notes


NOVEL OXYSTEROLS ACTIVATE THE HEDGEHOG PATHWAY IN<br />

VITRO AND STIMULATE SPINAL FUSION IN VIVO<br />

Wei Xiao 1 , Matthew Epperson 1 , Frank Stappenbeck 1 , Francine Farouz 1 , Mariko Reilly 1 , David Jenkins 1 , Scott<br />

Thies 1 , Farhad Parhami 2 , Jeffrey Wang 3 , Michael Jung 4 , Khanhlinh Nguyen 4<br />

1 Fate Therapeutics, 3535 General Atomics Court, San Diego, CA, 92121<br />

2 Department of Me<strong>di</strong>cine, David Geffen School of Me<strong>di</strong>cine, UCLA, Los Angeles, California, USA<br />

3 Department of Orthope<strong>di</strong>c Surgery, UCLA, Los Angeles, California, USA<br />

4 Department of <strong>Chemistry</strong> and Biochemistry, UCLA, Los Angeles, California, USA<br />

Bone morphogenic proteins (BMPs) that induce osteoblast <strong>di</strong>fferentiation have been used successfully in humans<br />

in spinal fusion procedures and non-union fracture treatments. However, BMPs are restricted in clinical use due to<br />

safety risks and high costs. Therefore, the identification of small molecules acting as anabolic agents represents a<br />

need for orthope<strong>di</strong>c me<strong>di</strong>cine, in particular for spinal fusion and fracture repair. In our search for new small<br />

molecule anabolic agents, we found that several naturally occurring oxysterols play a critical role during the<br />

<strong>di</strong>fferentiation of pluripotent mesenchymal stem cells to osteoblasts through activation of the Hedgehog signaling 1<br />

pathway. Here, we report on the synthesis and characterization of several novel structural analogs of 20(S)-<br />

hydroxy cholesterol and confirm that these novel oxysterols can induce in vivo osteogenic <strong>di</strong>fferentiation of<br />

mesenchymal stem cells in a spinal fusion model.<br />

1. Journal of Biol.Chem., 2008, 282, 12, 8959.<br />

L-29<br />

87


88<br />

Notes


BRAIN TUMOR STEM CELLS AS A TOOL TO IDENTIFY NOVEL<br />

THERAPEUTICS<br />

Patrizia Tunici and Annette Bakker<br />

Oncology Department, Siena Biotech S.p.A., Strada del Petriccio e Belriguardo, 35. 53100, Siena, Italy<br />

ptunici@sienabiotech.it.<br />

Primary brain tumours often represent the most devastating and <strong>di</strong>fficult to treat tumours. Although enormous<br />

advantages in treating other solid tumours highlighted the last decade, the me<strong>di</strong>an survival of glioblastoma<br />

remained the same over the last decades, averaging approximately 1 year. Our paramount inability to successfully<br />

treat brain cancer mostly stems from the lack of full understan<strong>di</strong>ng of the underlying brain tumour biology and the<br />

limited success of these strategies is largely due to a high incidence of tumour recurrence after treatment.<br />

Recent evidences suggested that a rare population of stem-like cells (or tumour initiating cells) present in brain<br />

tumours may be responsible for development of highly invasive and chemoresistant recurrence tumours. It is<br />

suggested that not all cells from a given brain tumour have the same ability to proliferate and maintain the growth<br />

of the tumour. Only a relatively small fraction of cells in the tumour, termed BTSC, possess the ability to<br />

proliferate, self-renew extensively and adapt to very <strong>di</strong>fferent micro-environments. Further investigations have<br />

demonstrated that brain tumour stem-like cells also more closely mirror the phenotype and genotype of the<br />

primary tumors and when injected in vivo they are able to give a tumour that recapitulates the characteristics of the<br />

tumour in the patient. Therefore, it can be suggested that to effectively treat brain tumour, BTSCs need to be<br />

eliminated. If the therapy eliminates only the <strong>di</strong>fferentiated cancer cells but spares a significant population of the<br />

BTSCs, the tumour will rapidly reform and the patient will have a clinical relapse.<br />

Since brain tumour stem-like cells (BTSC), share many properties with normal neural stem cells, it was shown that<br />

they can either be isolated from the tumour by making use of certain membrane markers (although which marker<br />

to use is under heavy debate) or can be cultured in stem cell like me<strong>di</strong>um con<strong>di</strong>tions in which they grow as<br />

neurospheres.<br />

In Siena Biotech, we have developed, characterized and validated reliable and reproducible 3D-culture sphere<br />

models derived from human biopsies originating from glioblastoma. We use such them for both target<br />

identification and validation as well as to for in vitro screening purposes and in vivo models.<br />

In the presentation, few application examples of the neurosphere model in neuro-oncology will be presented.<br />

L-30<br />

89


90<br />

Notes


SUCCESSFUL MODULATION OF TUMOR STEM CELLS WITH<br />

SMALL MOLECULE HEDGEHOG PATHWAY ANTAGONISTS<br />

Russell J. Thomas<br />

Me<strong>di</strong>cinal <strong>Chemistry</strong> Department, Siena Biotech S.p.A., Strada del Petriccio e Belriguardo, 35, 53100, Siena, Italy<br />

rthomas@sienabiotech.it.<br />

The hedgehog pathway, initially <strong>di</strong>scovered in Drosophila by two Nobel laureates, C Nusslein-Volhard and E<br />

Wieschaus (1), is a major regulator for cell <strong>di</strong>fferentiation, tissue polarity and cell proliferation. <strong>Stu<strong>di</strong></strong>es from many<br />

laboratories reveal activation of this pathway in a variety of human cancers (2). It is thus hoped that targeted<br />

inhibition of hedgehog signaling may be effective in treatment of human cancer.<br />

A number of research and development groups are actively investigating ways of blocking the hedgehog pathway<br />

(3). The most successful approach to date is to antagonize the seven-transmembrane domain receptor Smoothened<br />

(Smo), a key component in transforming hedgehog ligand signaling into growth factor expression.<br />

This presentation describes some of the results achieved in the laboratories of Siena Biotech. The approach has<br />

been to develop ligand-based pharmacophore models using reported antagonists of the Smo receptor and use these<br />

to design new structural classes. This approach, incorporating careful application of physicochemical property<br />

design principles, has led to the identification of a number of new scaffolds with promising levels of activity in a<br />

number of model systems.<br />

(1) Nusslein-Volhard C.; Wieschaus E. Nature, 1980, 287, 795–801.<br />

(2) Yang L., Xie G., Fan Q, Xie J. Oncogene, 2010, 29, 469-481.<br />

(3) Stanton B.Z.; Peng L.F.; Molecular BioSystems, 2010, 6, 44-54.<br />

L-31<br />

91


92<br />

Notes


THERAPEUTIC MODULATION OF THE WNT SIGNALLING<br />

PATHWAY, AN INNOVATIVE APPROACH FOR ONCOLOGY AND<br />

NEURODEGENERATION<br />

Giovanni Gaviraghi<br />

Siena Biotech S.p.A., Strada del Petriccio e Belriguardo, 35, 53100, Siena, Italy<br />

ggaviraghi@sienabiotech.it.<br />

Inhibition of aberrant Wnt signalling represents an important strategic approach to cancer therapies. A desperate<br />

need for agents able to modulate the pathway is also supported by the lack of chemicals specifically targeting the<br />

Wnt signalling in clinical testing.<br />

The pleiothropic effects of the Wnt pathway activate a broad number of target genes strongly linked to cancer and<br />

involved in all the steps of the <strong>di</strong>sease (insensitivity to anti-growth signals, limitless replicative potential, evasion<br />

of apoptosis, motility and tissue invasion and sustained angiogenesis) in a range of <strong>di</strong>fferent tumour types;<br />

consequently a pathway-based approach coupled to oncology-relevant phenotypic assays has the potential to<br />

identify inhibitors with <strong>di</strong>fferent therapeutic applications.<br />

Due to the <strong>di</strong>fferent mechanisms underlying Wnt pathway activation and the <strong>di</strong>fficulty a priori to identify relevant<br />

therapeutic entry points we decided to focus on a pathway or phenotype screening approach rather than target-<br />

based approaches (e.g. targeting β-catenin or components of the signalling downstream β-catenin). Our strategy is<br />

based on a pathway screening approach with the aim of identifying molecules able to affect <strong>di</strong>fferent nodes of the<br />

Wnt signalling network. This approach has the advantage of selecting compounds able to interfere with the<br />

signalling pathway and therefore affecting the phenotype and the need of identifying the target later on.<br />

The screening system based on a relevant cellular model (Wnt responsive cell line) with a wild-type genetic<br />

profile (free from mutations in the key Wnt molecules (Axin, APC and β-catenin)) and designed with a “three<br />

step” cascade to suit a primary high-throughput screening campaign delivered <strong>di</strong>fferent chemotypes (hit series)<br />

with potential <strong>di</strong>fferent mechanisms of action. The initial hit series selection based on cluster analysis and<br />

chemical structure, physico-chemical properties, biological activity and IP position was further prioritized on the<br />

basis of a minimal SAR (achieved through screening of chemical analogues) and IC50 values. The hit series<br />

prioritized showed in-vitro growth and motility inhibition property in the low micromolar range in <strong>di</strong>fferent cancer<br />

types. The confidence in the project is also based on the recent <strong>di</strong>scovery of the mechanism of action for one of<br />

our chemical series. We are currently focused on testing the in-vivo activity of the most advanced molecules in<br />

<strong>di</strong>fferent mouse tumour models.<br />

L-32<br />

93


94<br />

Notes


THE SUSTAINABILITY OF PHARMACEUTICALS IN THE 21 ST<br />

CENTURY: THE IDLE THOUGHTS OF AN IDLE FELLOW<br />

David J. Triggle<br />

State University of New York, Buffalo, NY USA<br />

“Now, La<strong>di</strong>es and Gentlemen, I may perhaps take the liberty of inviting you into the workshop of the<br />

chemotherapist”. So wrote Paul Ehrlich, a considerable optimist writing in 1913 on the eve of WW1 and<br />

mankind’s bloo<strong>di</strong>est century. Now almost a century later we may consider the progress of drug development and<br />

its future. The capacity to have an enduring flow of pharmaceuticals for the ills of the 21 st century will depend on<br />

science and in particular on the “new biology” based on the genomic <strong>di</strong>scoveries of the late 20 th century, but also<br />

and of equal importance, on science policy – will science endure as part of an independent and self-sustaining<br />

intellectual commons - on how much of the new biology will be private and corporate in nature? - - on economic<br />

policies – is health a public right or a private goods? – what are the effects of the increasing chemical<br />

environmental load on our health? – on health care policies – who and how will the 9.5 billion inhabitants of earth<br />

in 2050 have access to pharmaceuticals and health care? – how will it be paid for? - how will we grapple with the<br />

ethical and moral issues of the new biology – eternal life and happiness or a Pandora’s box of <strong>di</strong>sasters?<br />

References<br />

1. D. J. Triggle, Pharmaceuticals in Society, in, “Green and Sustainable Pharmacy”, Ed. K. Kummerer, Springer,<br />

Berlin, in press, 2010.<br />

2. D. J. Triggle, The chemist as astronaut: searching for biologically useful space in the chemical universe.<br />

Biochemical Pharmacology 78: 217-223, 2009.<br />

3. D. J. Triggle, Treating desires not <strong>di</strong>seases. Drug Discovery Today, 12: 161-166, 2006.<br />

L-33<br />

95


96<br />

Notes


SHORT COMMUNICATIONS<br />

97


98<br />

Bloom in “Castelluccio”<br />

“Green Italy” on Monti Sibillini<br />


WITHDRAWN<br />

S-01<br />

99


100<br />

SAR KNOWLEDGEBASE APPROACH FOR EXPLORING TARGET-<br />

LIGAND SELECTIVITY PATTERNS: APPLICATION TO KINASE<br />

TARGET FAMILY<br />

Frédéric Souvay, Ismail Ijjaali, Christelle Morlière, Elo<strong>di</strong>e Dubus, Olivier Barberan<br />

Aureus Pharma, 174 Quai de Jemmapes, 75010 Paris, FRANCE<br />

Protein kinases are important therapeutic targets for today’s drug <strong>di</strong>scovery as they are implicated in many<br />

<strong>di</strong>seases inclu<strong>di</strong>ng cancer and metabolic <strong>di</strong>sorders. AurSCOPE Kinase is one of Aureus Pharma’s knowledgebases<br />

developed to manage chemical and biological knowledge for this target family. Chemical structures of ligands as<br />

well as precise descriptions of their associated targets and all reported in vitro and in vivo pharmacological<br />

responses have been collected from literature (articles and patents). The scope of the present study is to explore the<br />

available structure-activity relationships (SAR) knowledge of kinase target family and identify related target-<br />

selectivity patterns as well as kinase ligands potency and specificity.<br />

S-02


IDENTIFICATION OF GPCR LIGANDS THROUGH DOCKING-BASED<br />

VIRTUAL SCREENING: NOT ONLY CRYSTAL STRUCTURES BUT<br />

ALSO HOMOLOGY MODELS.<br />

Stefano Costanzi 1 , Giulio Ferino 1 , Santiago Vilar 1 , Barkin Berk 1 , Stanislav Engel 2 , Irina G. Tikhonova 1 , T.<br />

Kendall Harden 4 , Kenneth A. Jacobson 3 , Marvin C. Gershengorn 2 , Clau<strong>di</strong>o Cavasotto 5<br />

1 Laboratory of Biological Modeling, 2 Clinical Endocrinology Branch and 3 Molecular Recognition Section, National Institutes<br />

101<br />

of Diabetes and Digestive and Kidney Diseases, NIH, DHHS. 4 Department of Pharmacology, School of Me<strong>di</strong>cine, University of<br />

North Carolina. 5 School of Health Information Sciences, University of Texas Health Science Center at Houston.<br />

Three-<strong>di</strong>mensional structures of target proteins are remarkably useful tools for the computer-assisted <strong>di</strong>scovery of<br />

novel lead ligands. Due to the paucity of crystal structures of G protein-coupled receptors (GPCRs), the scientific<br />

community has resorted to the construction of homology models which have been applied to the identification of<br />

lead ligands through virtual screening.<br />

Rhodopsin has been for years the only available template. However, recent breakthroughs in GPCR<br />

crystallography have led to the solution of the structures of a few ad<strong>di</strong>tional receptors. In light of these newly<br />

elucidated crystal structures, we have been able to produce a substantial amount of data to demonstrate that<br />

accurate models of GPCRs in complex with their ligands can indeed be constructed through homology modeling<br />

followed by fully flexible molecular docking. These results have been confirmed by our success in the first blind<br />

assessment of GPCR modeling and docking, organized in coor<strong>di</strong>nation with the solution of the X-ray structure of<br />

the adenosine A2A receptor (1).<br />

Using the β2-adrenergic receptor as a case study, we have investigated the applicability of crystal structures and<br />

homology models with <strong>di</strong>fferent levels of accuracy to the identification of GPCR ligands. In particular, our<br />

controlled docking-based virtual screening experiments have been very successful not only when applied to the<br />

experimental structure, but also with the homology models, yiel<strong>di</strong>ng a clearly delineated prioritization of a pool of<br />

known binders <strong>di</strong>spersed within a large set of decoy compounds. Remarkably, the retrieval yields can be further<br />

improved by generating an ensemble of alternative conformations of the receptor that accounts for its flexibility.<br />

These results are very much in line with our actual successes in the identification of lead compounds for several<br />

GPCRs, inclu<strong>di</strong>ng the thyrotropin releasing hormone (TRH) receptor, the free fatty acid receptor 1 (FFA1, or<br />

GPR40) and the nucleotide P2Y1 receptor, on the basis of virtual screenings conducted using their homology<br />

models.<br />

(1) Michino M. et al. Nat. Rev. Drug Discov. 2009, 8, 455-463.<br />

S-03


102<br />

FUNCTIONALLY-SELECTIVE SEROTONIN 5-HT2 GPCR DRUGS:<br />

5-HT2C AGONISTS WITH 5-HT2A/2B INVERSE AGONIST ACTIVITY<br />

FOR NEUROPSYCHIATRIC DISORDERS<br />

Raymond G. Booth<br />

Department of Me<strong>di</strong>cinal <strong>Chemistry</strong>, College of Pharmacy University of Florida, Gainesville, Florida 32610 USA<br />

Serotonin 5-HT2A, 5-HT2B, and 5-HT2C GPCRs signal primarily through Gαq to activate phospholipase C and<br />

formation of inositol phosphates and <strong>di</strong>acylglycerol second messengers. The human 5-HT2C receptor is found<br />

exclusively in the central nervous system and there is compelling evidence in humans and lab animals that 5-HT2C<br />

receptor activation is pharmacotherapeutic for severe neuropsychiatric <strong>di</strong>sorders such as schizophrenia, as well as,<br />

obsessive-compulsive behavioral <strong>di</strong>sorders such as over-eating and psychostimulant drug abuse. Meanwhile,<br />

5HT2A receptor activation produces hallucinogenic effects and 5-HT2B activation leads to heart valve damage and<br />

pulmonary hypertension, thus, there is zero tolerance for drug activation of these 5-HT2 subtypes. A 5-HT2C<br />

agonist drug that does not also activate 5-HT2A and/or 5-HT2B receptors has not been developed despite lucrative<br />

and beneficial therapeutic impact. Drug <strong>di</strong>scovery targeting the 5-HT2C receptor is challenging because its shares<br />

75% transmembrane sequence identity with 5-HT2A/2B. The highly conserved transmembrane domains and same<br />

second messenger signaling complicates development of ligands that activate 5-HT2C but not 5-HT2A or 5-HT2B<br />

receptors. Our me<strong>di</strong>cinal chemistry program incorporating site-<strong>di</strong>rected mutagenesis and molecular modeling<br />

stu<strong>di</strong>es reveals important <strong>di</strong>fferences in molecular determinants for activation of the 5-HT2 GPCR family<br />

members. We have developed several novel (4R)-substituted-(2S)-<strong>di</strong>methylaminotetralin derivatives that are full-<br />

efficacy agonists at human 5-HT2C receptors, but, inverse agonists at 5-HT2A/2B receptors expressed in clonal cells.<br />

Off-target screening of several analogs in<strong>di</strong>cates no significant affinity at 440 kinases and several-dozen GPCRs,<br />

ion channels, transporters, and signaling/metabolizing enzymes. Accor<strong>di</strong>ngly, preclinical stu<strong>di</strong>es in laboratory<br />

animals were conducted to assess pharmacotherapeutic activity and safety for 3 lead analogs administered by<br />

intraperitoneal injection. Using 2 <strong>di</strong>fferent rodent models of schizophrenia, potent and efficacious antipsychotic<br />

activity without weight-gain was demonstrated for 2 analogs. In mice, all 3 analogs demonstrated frank anorexic<br />

activity or no stimulation of food intake at doses that produce antipsychotic effects—no tolerance to the anorexic<br />

effects developed with chronic administration. Inhibition of psychostimulant-induced behavior was demonstrated<br />

for 2 analogs in rats and in vivo potency correlates with in vitro 5HT2A inverse agonist and 5-HT2C agonist<br />

potency. No adverse affects were apparent in the animal stu<strong>di</strong>es reported here and detailed toxicology and<br />

pharmacokinetic stu<strong>di</strong>es are in progress. Me<strong>di</strong>cinal chemistry, molecular modeling, and in vitro/in vivo<br />

pharmacology data will be presented at the Symposium.<br />

Research Fun<strong>di</strong>ng: United States National Institutes of Drug Abuse and Mental Health (RO1 DA023928 & MH081193)<br />

S-04


MEDICINAL CHEMISTRY OF NOVEL HISTAMINE H4 RECEPTOR<br />

ANTAGONISTS<br />

Rogier A Smits, Herman D. Lim, Gabriella Coruzzi, Maristella Adami, Enade Istyastono, Erik Haaksma, Obbe P.<br />

Zuiderveld, Bert D. Windhorst, Iwan J. P. de Esch, Rob Leurs<br />

Dept. Me<strong>di</strong>cinal <strong>Chemistry</strong>, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands<br />

103<br />

The histamine H4 receptor is a G protein-coupled receptor that, over the past few years, has been demonstrated to<br />

play an important role in inflammatory and immunomodulatory functions (1). Animal models show great potential<br />

for H4R ligands in the treatment of allergic rhinitis, asthma and pruritis, lea<strong>di</strong>ng to much interest in this receptor as<br />

a new drug target (2). In our search for new H4R ligands, we took a set of small heterocyclic fragments as a<br />

starting point for a me<strong>di</strong>cinal chemistry campaign. Using ligand based in silico modeling and parallel synthesis<br />

approaches, we developed several chemically <strong>di</strong>stinct ligand series with excellent affinity for the H4R. Several of<br />

the compounds showed inverse agonistic behavior and significant anti-inflammatory properties in vivo in the rat.<br />

(1) Smits, R.; de Esch, I. ; Leurs, R. Drug Disc. Today 2009, 15-16, 745-753.<br />

(2) Thurmond, R. ; Nat. Rev. Drug Discov. 2008, 7:41-53.<br />

S-05


104<br />

NOVEL α1-ADRENORECEPTOR ANTAGONISTS RELATED TO<br />

OPENPHENDIOXAN: BIOLOGICAL EVALUATION<br />

AND α1d COMPUTATIONAL STUDY<br />

Antonio Carrieri, a Alessandro Piergentili, b Fabio Del Bello, b Mario Giannella, b Maria Pigini, b Amedeo Leonar<strong>di</strong>, c<br />

Francesca Fanelli, d and Wilma Quaglia b<br />

a Dipartimento Farmaco-Chimico, <strong>Università</strong> <strong>di</strong> Bari, via Orabona 4, 70125 Bari, Italy<br />

b Scuola <strong>di</strong> Scienze del Farmaco e dei Prodotti della Salute, <strong>Università</strong> <strong>di</strong> Camerino, via S. Agostino 1, 62032 Camerino, Italy<br />

c Recordati S. p. A., Drug Discovery, via Civitali 1, 20148 Milano, Italy<br />

d Istituto Dulbecco Telethon e Dipartimento <strong>di</strong> Chimica, <strong>Università</strong> <strong>di</strong> Modena & Reggio Emilia,<br />

via Campi 183, 41100 Modena, Italy<br />

A series of novel openphen<strong>di</strong>oxan analogues were synthesized and tested at α1-adrenoreceptor (AR) subtypes by<br />

bin<strong>di</strong>ng and functional assays. The α1d-AR bin<strong>di</strong>ng profile was also examined by means of 2D, 3D-QSAR together<br />

with docking stu<strong>di</strong>es. Multiple regression analysis suggested the relevance of adequate number of heteroatoms in<br />

the whole molecule and of passive membrane <strong>di</strong>ffusion to enhance α1d-affinity. Docking simulations against a<br />

computational structural model of the biological target further proved this evidence and furnished support for<br />

chemiometric analysis, where polar, electrostatic, hydrophobic and shape effects of the ortho substituents in the<br />

phenoxy terminal, most likely governing ligand bin<strong>di</strong>ng, helped the depiction of pharmacophore hypothesis for the<br />

examined ligands data set.<br />

Thr249<br />

Ile248<br />

Gly247<br />

Asp176<br />

Phe384<br />

Phe365<br />

Phe364<br />

Tyr392<br />

Phe388<br />

Trp361<br />

S-06


PROTEIN KINASES: DOCKING AND HOMOLOGY MODELING<br />

RELIABILITY<br />

Tiziano Tuccinar<strong>di</strong>, 1,2 Adriano Martinelli 1<br />

1 Department of Pharmaceutical Science, University of Pisa, Italy. 2 Department of Biology, Sbarro Institute for Cancer<br />

Research and Molecular Me<strong>di</strong>cine, Temple University, Philadelphia, Pennsylvania 19122, USA<br />

105<br />

Protein kinases play a major role in eukaryotic signal transduction, via regulation of the phosphorylation states<br />

and, thus, cellular functions of substrate proteins. In order to design highly selective or particular multitarget<br />

kinase ligands, computational chemistry is a very helpful tool for pre<strong>di</strong>cting the geometry of kinase-ligand<br />

complexes (by means of docking software) and for provi<strong>di</strong>ng atomic-resolution models of kinases without an<br />

experimentally known 3D structure (by means of homology modeling techniques).<br />

With regards to the pre<strong>di</strong>ction of the geometry of kinase-ligand complexes, is it possible to measure the level of<br />

reliability of docking software to pre<strong>di</strong>ct the bin<strong>di</strong>ng pose of ATP bin<strong>di</strong>ng site ligands (ABSL)?<br />

In order to <strong>di</strong>scuss this question we retrieved from the PDB databank more than 700 X-ray structures of kinases<br />

complexed with ABSL and, using six <strong>di</strong>fferent docking software (for a total of 17 <strong>di</strong>fferent docking procedures),<br />

we applied two <strong>di</strong>fferent level of analysis: a) re-docking of the ligands into their co-crystallized kinases; b)<br />

extended cross-docking stu<strong>di</strong>es for all the kinases with more than five reported X-ray complexes.<br />

Re-docking calculations show that there are about sixty percent of possibilities to find a correct ligand pose in the<br />

best docking con<strong>di</strong>tions. The extended cross-docking stu<strong>di</strong>es suggest that the docking of an unknown ligand into a<br />

kinase has only 37% of possibilities to find a correct ligand pose. Furthermore a careful analysis of the cross-<br />

docking stu<strong>di</strong>es suggests that the docking success is correlated with the bin<strong>di</strong>ng site similarity and the latter is<br />

strongly affected by the similarity of the ligands. This fact determines that, taking into account only ligands with a<br />

good similarity, there are about 60% of possibilities to find a correct ligand pose even in the cross-docking results.<br />

In literature a large number of kinase 3D structures, developed by means of homology model (HM) techniques, are<br />

reported. Most of them have been obtained using as template the 3D structure possessing the highest sequence<br />

similarity with the target kinase. However, our data suggest that kinases adapt themselves for the interaction with<br />

ABSL and this adaptation appears to be fundamental for the docking pre<strong>di</strong>ction. On these bases, we have tested<br />

the hypothesis that better results might be obtained for docking a generic ligand X to a target kinase using a HM of<br />

the target created from a kinase template co-crystallized with a ligand similar to X, than from the crystal structure<br />

of the template with the highest sequence similarity with the target kinase. Our preliminary results suggest that<br />

since the kinase shape has a key role, the use of the template with the highest sequence similarity could not always<br />

be the best choice.<br />

S-07


106<br />

A NEGATIVE ALLOSTERIC MODULATOR OF α7 nAChRs<br />

Małgorzata Dukat, 1 Genevieve S. Alley, 1 Galya R. Abdrakhmanova, 2 Hernán A. Navarro 3<br />

1 Department of Me<strong>di</strong>cinal <strong>Chemistry</strong>, School of Pharmacy; 2 Department of Pharmacology & Toxicology, School of Me<strong>di</strong>cine;<br />

Virginia Commonwealth University, Richmond, VA 23298, USA; 3 Pharmacology and Toxicology, Discovery Sciences,<br />

Research Triangle Institute, Research Triangle Park, NC 27709, USA<br />

Historically our laboratory was and is interested in understan<strong>di</strong>ng and developing SAFiR and SAR for nAChR<br />

(nicotinic acetylcholine receptor) ligands. nAChRs belong to the Cys–loop super family of ligand gated ion<br />

channel receptors (LGICRs). Among them are 5-HT3, GABAA, GABAC, and glycine receptors. nAChRs are the<br />

oldest and most stu<strong>di</strong>ed receptors among the LGICRs. Neuronal nAChRs form functionally active heteromeric<br />

(e.g., α4β2) and homomeric (e.g., α7) receptors. α7 nAChRs are critical for learning and memory and have been<br />

implicated to play a role in Alzheimer’s <strong>di</strong>sease (AD) and other neurological <strong>di</strong>sorders. Interestingly, α7 nAChR-<br />

selective ligands, either agonist or antagonist, have demonstrated neuroprotective effects against β-amyloid1-42<br />

toxicity associated with AD. One plausible explanation is that these effects may be attributed to rapid and<br />

extensive desensitization of α7 nAChRs as with agonists and inhibition by antagonists.<br />

There is compelling evidence that MD-354 (m-chlorophenylguani<strong>di</strong>ne), an analog developed and synthesized in<br />

our laboratory, when co-administered with (-)nicotine might produce its biological effects through α7 nAChRs.<br />

MD-354 lacks affinity at the major population of neuronal nAChRs (i.e., α4β2) (1). In vivo rodent assays with (-<br />

)nicotine and MD-354, such as drug <strong>di</strong>scrimination, locomotor activity, and thermal nociceptive assays (i.e., hot-<br />

plate and tail-flick) (1) imply that MD-354 might behave as an α7nAChR antagonist. Consequently, MD-354 was<br />

examined for its ability to bind at α7 nAChRs. MD-354 produced < 50% inhibition at a concentration as high as<br />

100 µM. Lacking affinity for α7 nAChRs, a remaining possibility is that MD-354 is a negative allosteric<br />

modulator of α7 nAChRs. Indeed, MD-354 (IC50 = 7.98 µM) was demonstrated to allosterically antagonize the<br />

effect of ACh at α7 nAChRs. Moreover, MD-354 (doses 1 and 10 µM) was inactive at peripheral α3β4 nAChRs<br />

that are associated with undesirable side effects.<br />

Allosteric modulators are attractive but challenging targets in drug design. MD-354 is one of the first negative<br />

allosteric modulators of α7 nAChRs known today.<br />

(1) Dukat M, Wesolowska A, Young S, Bondareva T, Young R, Glennon RA. CPDD 2006 Annual Meeting, Scottsdale,<br />

Arizona. Abstract 202:51.<br />

S-08


CHEMOINFORMATICS APPROACHES FOR ANTI-DEMENTIA<br />

TRADITIONAL CHINESE MEDICINE RESEARCH AND<br />

IMPLICATIONS FOR FINDING POLYPHARMACOLOGY DRUGS<br />

Xue-Juan Li, De-Xin Kong, Hong-Yu Zhang*<br />

College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, P R China<br />

107<br />

Dementia, inclu<strong>di</strong>ng Alzheimer’s <strong>di</strong>sease (AD), is one of the most threatening <strong>di</strong>seases to the elderly population.<br />

However, there is no yet efficient therapeutic method to AD. It is of great interest to note that tra<strong>di</strong>tional Chinese<br />

me<strong>di</strong>cine (TCM) has accumulated some experiences in the treatment of dementia. TCM is one of the most<br />

potential sources of polypharmacology drugs. TCM featured in its combinatorial use of natural me<strong>di</strong>cines and its<br />

curative effects from integrative contributions of multi-components which especially have advantages in the<br />

treatment of multi-factor and complex <strong>di</strong>seases, such as dementia. Applying cheminformatics methods to calculate<br />

the similarity of hundreds of components which were identified from most prevalent herbs (Rhizoma Chuanxiong,<br />

Ra<strong>di</strong>x Salviae Miltiorrhizae, Ra<strong>di</strong>x Polygalae Tenuifoliae, and Rhizoma Acori Tatarinowii) in anti-dementia TCM<br />

formulae through searching tra<strong>di</strong>tional Chinese me<strong>di</strong>cine database (TCMD) with drug can<strong>di</strong>dates recorded in<br />

MDL drug data report (MDDR) database. It was found that some components (e.g., tetramethylpyrazine, 3-n-<br />

butylphthalide, miltirone, 9-cis, 12-cis-linoleic acid, baicalin) are associated with anti-dementia and have been<br />

recognized by modern me<strong>di</strong>cine. In ad<strong>di</strong>tion, Ra<strong>di</strong>x Polygalae Tenuifoliae contains 1-hydroxy-3,6,7-trimethoxy<br />

xanthone, which is annotated as an anti<strong>di</strong>abetic and an aldose reductase inhibitor in MDDR database and thus<br />

helpful to treating <strong>di</strong>abetes-related cognitive decrements. Therefore, it could be concluded that anti-dementia TCM<br />

formulae indeed contain some modern-me<strong>di</strong>cine-justified anti-dementia components, which are likely to act<br />

synergistically in the fight against dementia. These results have important implications for the rational design of<br />

anti-dementia polypharmacology drugs.<br />

S-09


108<br />

CURCUMIN ANALOGS AS MULTI-TARGET ANTIOXIDANTS: FOCUS<br />

ON MITOCHONDRIA<br />

Elena Simoni, a Michela Rosini, a Christian Bergamini, b Romana Fato, b Maria Laura Bolognesi, a Andrea Cavalli, a,c<br />

Roberto Motterlini, c Giorgio Lenaz, b Carlo Melchiorre. a<br />

a)Department of Pharmaceutical Science, University of Bologna , Via Belmeloro 6, I-40126 Bologna, Italy ; b) Department of<br />

Biochemistry, University of Bologna , Via Irnerio 48, I-40126 Bologna, Italy; c) Department of Drug Discovery and<br />

Development - Italian Institute of Technology, Via Morego 30, I-16163, Genova, Italy<br />

Many lines of evidence suggest that oxidative stress and mitochondria impairment have a central role in age-<br />

related neurodegenerative <strong>di</strong>seases such as Alzheimer’s <strong>di</strong>sease (AD). In ad<strong>di</strong>tion, the recent understan<strong>di</strong>ng that<br />

mitochondria are at the intersection of the life and death of a cell, particularly through the involvement of<br />

mitochondrial oxidative stress, has made mitochondria a promising target for drug <strong>di</strong>scovery and therapeutic<br />

interventions (1). Thus, considering the oxidative stress as a crucial event on the degenerative cascade, we<br />

designed a series of compounds presenting an antioxidant constrained curcumin-like moiety. Curcumin and its<br />

constrained analogs have currently received remarkable interest as they have a unique conjugated structure which<br />

shows a pleiotropic biological profile. Beside the well-known <strong>di</strong>rect antioxidant activity, curcumin <strong>di</strong>splays a wide<br />

range of properties inclu<strong>di</strong>ng anti-inflammatory and anti-amyloidogenic activities. Moreover, curcumin has<br />

recently been demonstrated to be neuroprotective through the induction of the antioxidant enzyme heme<br />

oxygenase-1, showing an ad<strong>di</strong>tional in<strong>di</strong>rect cytoprotective effect (2).<br />

On the basis of the consideration that mitochondria are a major source of reactive oxygen species and are<br />

particularly vulnerable to oxidative stress, it is plausible that antioxidants that alleviate mitochondrial dysfunction<br />

could be beneficial in AD (3). This prompted us to design mitochondria-targeted antioxidants by connecting<br />

curcumin analogs to <strong>di</strong>fferent polyamine chains. Polyamines might deliver the correspon<strong>di</strong>ng conjugates into the<br />

cell with active polyamine transporters and, acting as lipophilic cations (4), drive the antioxidant moiety<br />

selectively into mitochondria in a membrane potential-dependent manner across the inner membrane. In order to<br />

test if such compounds were preferentially taken up by mitochondria, a fluorescent probe was designed by<br />

combining the phenyl benzothiazole group with the polyamine residue.<br />

(1) Moreira, P. I.; Zhu, X.; Wang, X.; Lee, H.G.; Nunomura, A.; Petersen, R. B.; Perry, G.; Smith, M.A., Biochim. Biophys.<br />

Acta. 2010, 1802, 212-220.<br />

(2) Calabrese, V.; Cornelius, C.; Mancuso, C.; Barone, E.; Calafato, S.; Bates, T.; Rizzarelli, E.; Kostova, AT., Front. Biosci.<br />

2009, 14, 376-397.<br />

(3) Bolognesi, ML.; Matera, R.; Minarini, A.; Rosini, M.; Melchiorre C., Curr. Opin. Chem. Biol. 2009, 13, 1-6.<br />

(4) Murphy, M.P., Biochim. Biophys. Acta. 2008, 1777, 1028-1031.<br />

S-10


IDENTIFICATION OF A NOVEL OX1 ANTAGONIST SERIES BY<br />

HIGH-THROUGHPUT SCREENING AND SDM-DIRECTED<br />

HOMOLOGY MODELING OF OREXIN RECEPTORS<br />

*J. Bentley, *T. Fryatt, *E. Glenn, *D. Hallett, *A. Heifetz, † D. Manikowski, † R. Reifegerste, † X. Wang, † M.<br />

Slack, *D. Vaidya and *M. Whittaker<br />

*Evotec, 114 Milton Park, Abingdon, OX14 4SA, UK<br />

† Evotec AG, Schnackenburgallee 114, 22525 Hamburg, Germany<br />

109<br />

Clinical trials of dual OX1/2 receptor antagonists have shown statistically significant efficacy in increasing sleep<br />

efficiency, total sleep time and percentage of time spent in REM sleep in patients with insomnia. 1 Recent<br />

preclinical fin<strong>di</strong>ngs suggest that a selective OX2 antagonist may be preferred over a dual OX1/2 antagonist. 2<br />

Two high throughput screening campaigns of Evotec’s 250K compound library were conducted against both<br />

orexin receptors utilizing FLIPR-based assays in stably-transfected CHO cells. Molecular target engagement was<br />

confirmed using a non-ra<strong>di</strong>oactive GTP bin<strong>di</strong>ng assay. Cross-species selectivity was evaluated against rat OX1<br />

and rat OX2 receptors. One novel OX1 selective series and two novel OX2 selective series were confirmed. The<br />

novel OX1 selective series will be presented.<br />

Evotec has a developed a hierarchical protocol for GPCR modeling. Initial homology models of OX1 and OX2<br />

GPCR receptors were produced in order to suggest a set of potential key residues for site-<strong>di</strong>rected mutagenesis<br />

(SDM). The SDM study revealed that some of the residues are important to ligand bin<strong>di</strong>ng and others for receptor<br />

stability. Significant <strong>di</strong>fferences in the shift of potency of dual OX1/2 antagonists compared to OX1 selective<br />

antagonists were seen. This ad<strong>di</strong>tional SDM information was used to refine further the modeling process. Ab-<br />

initio modeling of TM4 and multiple kinks in TM6 of the GPCR model were required due to the significant<br />

<strong>di</strong>fferences between Orexin receptors and published 7-TM crystal structures.<br />

(1) Bisbare-Roch, C. et al., Nat. Med., 2007, 13, 150-155.<br />

(2) Dugovik, C. et al., J. Pharmacol. Exp. Ther., 2009, 330(1), 142-151.<br />

S-11


110<br />

SODIUM IONS TOGGLE THE ROTAMER SWITCH IN SODIUM-<br />

SENSITIVE DOPAMINERGIC G-PROTEIN COUPLED RECEPTORS<br />

Jana Selent a , Ferran Sanz b , Manuel Pastor a , and Gianni De Fabritiis c<br />

a Computer-Assisted Drug Design Laboratory, b Integrative Biome<strong>di</strong>cal Informatics Laboratory, c Computational Biochemistry<br />

and Biophysics Laboratory, GRIB, IMIM, Universitat Pompeu Fabra, C/ Dr. Aiguader 88, 08003, Barcelona, Spain<br />

G-protein coupled receptors, the largest family of proteins in the human genome, are involved in many signaling<br />

pathways regulated by orthosteric ligand bin<strong>di</strong>ng and allosteric modulation. Dopaminergic receptors which belong<br />

to the class A family of G-protein coupled receptors take part in the control of various neurological processes and<br />

are therefore important drug targets in the treatment of central nervous system <strong>di</strong>sorders (e.g. schizophrenia,<br />

Parkinson <strong>di</strong>sease). Recent stu<strong>di</strong>es in<strong>di</strong>cate that so<strong>di</strong>um ions allosterically modulate the dopaminergic D2 receptors<br />

affecting mainly agonist bin<strong>di</strong>ng at the orthosteric bin<strong>di</strong>ng site (1, 2). In an effort to understand the mechanisms of<br />

allosteric modulation of so<strong>di</strong>um ions for the dopaminergic D2 receptor, we performed microsecond scale all-atom<br />

molecular dynamics simulations to characterize so<strong>di</strong>um bin<strong>di</strong>ng and modulation. We found that so<strong>di</strong>um ions enter<br />

the receptor from the extracellular side and bind at the residue Asp2.50, an allosteric site deep inside the receptor.<br />

Most importantly, the presence of a so<strong>di</strong>um ion at this allosteric site provokes a conformational change of the<br />

rotamer toggle switch Trp6.48 which locks in a state identical to the inactive state of the crystallized human β2<br />

adrenergic receptor (3). This study provides novel insights into the effect of so<strong>di</strong>um ions on receptor conformation<br />

and its importance for the allosteric modulation of G-protein coupled receptors.<br />

(1) Ericksen, S.S.; Cummings D.F.; Weinstein H.; Schetz J.A. J. Pharmacol. Exp. Ther. 2009, 328, 40-54.<br />

(2) Schetz J.A. Mini. Rev. Med. Chem. 2005, 5, 555-61.<br />

(3) Selent J., Sanz F.; Pastor M.; De Fabritiis G. 2010, (submitted).<br />

S-12


POSTER SESSION<br />

111


112<br />

Pizzo del Diavolo<br />

Monte Bove


MOLECULAR MODELING ANALYSIS OF THE INTERACTIONS OF<br />

THE HUMAN ADENOSINE A2A RECEPTOR WITH ITS LIGANDS<br />

Barkin Berk 1 , Santiago Vilar Varela 1 , Kenneth A. Jacobson 2 , Stefano Costanzi 1<br />

1 Laboratory of Biological Modeling and 2 Molecular Recognition Section, National Institutes of Diabetes and Digestive and<br />

Kidney Diseases, NIH, DHHS,USA.<br />

113<br />

In 2008, the human adenosine A2A receptor and its moderately selective, high-affinity antagonist ligand,<br />

ZM241385 (4-2-[7-amino-2-(2-furyl)-1,2,4-triazolo[1,5-a][1,3,5]triazin-5-yl-amino]ethylphenol) joined the<br />

shortlist of GPCRs for which an X-ray crystallographic structure has been determined (1). Moreover, over the<br />

course of the years, many stu<strong>di</strong>es and reviews were published concerning its homology modeling, structure of<br />

bin<strong>di</strong>ng site, various synthesis of agonist /antagonists and their structure and activity relationships (2-3). All these<br />

data in<strong>di</strong>cate that typical antagonists and agonists of A2A AR bind with a mode similar to the one shown by<br />

ZM241385, forming a key H-bond with Asn253 in TM6 with their exocyclic NH groups. Ad<strong>di</strong>tionally, published<br />

docking stu<strong>di</strong>es suggest that the 2′- and 3′-OH groups of the ribose moiety, essential for agonistic activity, bind in<br />

proximity of Ser277 and His278 and are putatively involved in H-bon<strong>di</strong>ng interactions with these residues.<br />

To further elucidate the modes of bin<strong>di</strong>ng of ligands of the human A2A adenosine receptor and to establish the key<br />

<strong>di</strong>fferences between agonists and antagonists, in this study more than 200 ligands based on various chemical<br />

scaffolds were selected from many publications and subjected to molecular docking. The docking was performed<br />

using the bicyclic core of ZM241385 as a constraint and various criteria for the selection of the poses.<br />

The “protein-ligand interaction fingerprints” and “protein-ligand interaction fields” as implemented in the<br />

Molecular Operating Environment (MOE) were also used as tools to highlight the role of the various amino acids<br />

in the interaction with the ligands.<br />

(1) Jaakola VP. et al. Science. 2008 , 322, 1211–1217.<br />

(2) Cristalli G. et al. ChemMedChem. 2007, 2( 3), 260-281.<br />

(3) Ivanov AA. et al. J Med Chem. 2009, 52 (10), 3284-3292.<br />

P-01


114<br />

EFFECTS OF 5'-CHLORO-5'-DEOXY-N 6 -(±)-ENDO-NORBORNYL-<br />

ADENOSINE, A POTENT AND HIGHLY SELECTIVE A1 ADENOSINE<br />

RECEPTOR AGONIST, ON NEUROPATHIC PAIN-INDUCED<br />

BEHAVIOURAL AND MORPHOLOGICAL CHANGES IN SPINAL<br />

MICROGLIA<br />

Livio Luongo, a Loredana Cappellacci, b Catia Giordano, a Riccardo Petrelli, b Francesca Guida, a Palmarisa<br />

Franchetti, b Mario Grifantini b and Sabatino Maione a<br />

Department of Experimental Me<strong>di</strong>cine, The Second University of Naples, via Costantinopoli 16, 80138 Naples.<br />

School of Pharmacy, Me<strong>di</strong>cinal <strong>Chemistry</strong> Unit, University of Camerino, via S. Agostino 1, 62032 Camerino<br />

This study was undertaken in order to investigate the effect of chronic treatment with 5'-chloro-5'-deoxy-N 6 -(±)-<br />

endo-norbornyladenosine (5'Cl5'd-(±)-ENBA), 1 a potent and highly selective adenosine A1 receptor agonist, on<br />

thermal hyperalgesia and mechanical allodynia in a mouse model of neuropathic pain, the spared nerve injury<br />

(SNI). Chronic systemic administrations of 5'Cl5'd-(±)-ENBA (0.5 mg/kg, i.p once a day) reduced both thermal<br />

hyperalgesia and mechanical allodynia 3 and 7 days after SNI, in a way prevented by DPCPX (3 mg/kg, i.p.), a<br />

selective A1 receptor antagonist. SNI induced spinal changes on microglial activation ipsilaterally to the nerve<br />

injury. Moreover, 5'Cl5'd-(±)-ENBA significantly reduced microglial activation in vitro. In particular pre-<br />

incubation with 5'Cl5'd-(±)-ENBA prevented the microglial morphological changes induced by LPS, ATP,<br />

LPS+ATP challenges. Our results demonstrated an involvement of A1 receptors in the increase of nociceptive<br />

thresholds and in spinal changes occurred in neuropathic pain. In ad<strong>di</strong>tion, 5'Cl5'd-(±)-ENBA antiallodynic and<br />

antihyperalgesic effects could be me<strong>di</strong>ated by the A1 receptors expressed on microglial cells.<br />

(1) Franchetti, P.; Cappellacci, L.; Vita, P.; Petrelli, R.; Lavecchia, A.; Kachler, S.; Klotz, K.-N.; Marabese, I.; Luongo, L.;<br />

Maione, S.; Grifantini, M. J. Med. Chem. 2009, 52, 2393-2406.<br />

P-02


MOLECULAR MODELING STUDY ON POTENT AND SELECTIVE<br />

ADENOSINE A3 RECEPTOR AGONISTS<br />

Diego Dal Ben, Michela Buccioni, Dania Giacobbe, Catia Lambertucci, Gabriella Marucci, Deepak Shankar<br />

Panmand, Rosaria Volpini, Gloria Cristalli<br />

School of Pharmacy, Me<strong>di</strong>cinal <strong>Chemistry</strong> Unit, University of Camerino; Via S. Agostino 1, 62032 Camerino (MC), Italy<br />

115<br />

Adenosine A3 receptor (AA3R) is involved in a variety of key physiological processes such as release of<br />

inflammatory me<strong>di</strong>ators and inhibition of tumor necrosis factor-α production. Its activation is suggested to take<br />

part in immunosuppression and in the protection from brain and heart ischemia. Recent stu<strong>di</strong>es suggest that AA3R<br />

agonists could be employed as therapeutic agents for the treatment of rheumatoid arthritis, dry eye <strong>di</strong>sorders,<br />

asthma, as anti-inflammatory agents, and in cancer therapy as cytostatic and chemoprotective compounds (1).<br />

Hence, the design and synthesis of potent and selective AA3R agonists could help to provide tools for further<br />

characterization of the physiopathological role of this receptor and for the development of new drugs. Previously<br />

reported 2-(ar)alkynyl-N 6 -methoxyadenosine and correspon<strong>di</strong>ng MECA (5’-N-methylcarboxamidoadenosine)<br />

derivatives demonstrated to possess <strong>di</strong>fferent degrees of affinity and selectivity for the human AA3R (2). Basing<br />

on further previous stu<strong>di</strong>es demonstrating that also the introduction of a methyl group in N 6 -position of 2-<br />

phenylethynyladenosine favoured the interaction with the human AA3R (3), we designed and synthesised 2-<br />

aralkynyl-N 6 -methylMECA analogues with the aim at improving AA3R affinity and selectivity. Biological<br />

evaluation of the compounds proved that the new nucleosides behave as full AA3R agonists, possessing sub-<br />

nanomolar AA3R affinity and remarkable selectivity versus the other adenosine receptor (AR) subtypes, hence<br />

resulting to be among the most potent and selective AA3R ligands reported so far (4). In this study, we made an<br />

attempt to get a rationalization of the high affinities and selectivities of these molecules for the human AA3R, by<br />

using AR structural models based on the AA2AR crystal structure and molecular docking analysis. Post-docking<br />

analysis allowed to assess the ability of modeling tools in pre<strong>di</strong>cting AA3R affinity and in evaluating the influence<br />

of substituents on affinity and selectivity.<br />

References:<br />

(1) Cristalli, G.; Volpini, R. Adenosine <strong>Receptor</strong>s: Curr. Top. Med. Chem. 2003, 3, 355-469.<br />

(2) Volpini, R.; Dal Ben, D.; Lambertucci, C.; Taffi, S.; Vittori, S.; Klotz, K.-N.; Cristalli, G. J. Med. Chem. 2007, 50, 1222-<br />

1230.<br />

(3) Volpini, R.; Costanzi, S.; Lambertucci, C.; Taffi, S.; Vittori, S.; Klotz, K.-N.; Cristalli, G. J. Med. Chem. 2002, 45, 3271-<br />

3279.<br />

(4) Volpini, R.; Buccioni, M.; Dal Ben, D.; Lambertucci, C.; Lammi, C.; Marucci, G.; Ramadori, A. T.; Klotz, K.-N.; Cristalli,<br />

G. J. Med. Chem. 2009, 52, 7897-7900.<br />

P-03


116<br />

SYNTHESIS AND BIOLOGICAL EVALUATUION OF POTENT<br />

LIGANDS FOR THE PUTATIVE GUANOSINE RECEPTOR<br />

Ajiroghene Thomas, Michela Buccioni, Diego Dal Ben, Catia Lambertucci, Carmen Lammi, Gabriella Marucci,<br />

Rosaria Volpini, Gloria Cristalli<br />

School of Pharmacy, Me<strong>di</strong>cinal <strong>Chemistry</strong> Unit, University of Camerino, via S. Agostino, 1, 62032 Camerino, Italy.<br />

A variety of cells releases guanosine and its nucleotides under physiological and pathological con<strong>di</strong>tions. The<br />

amount of guanine-based purines released from astrocytes is fivefold much greater than that of their adenine-based<br />

counterparts (1). There are extensive evidences that extracellular guanosine exerts a plethora of biological effects<br />

inclu<strong>di</strong>ng trophic modulations in the nervous system, promotion of cell proliferation and <strong>di</strong>fferentiation,<br />

neuroprotective actions, inhibition of glutamatergic transmission, and antiapoptotic effects (2).<br />

Specific bin<strong>di</strong>ng sites for [ 3 H]-guanosine, which are not recognized by other purinergic receptor ligans, were<br />

detected on membrane preparations from rat brain (3). These fin<strong>di</strong>ngs supported the hypothesis of the existence of<br />

a specific G protein-coupled receptor (GPCR) for guanosine.<br />

On this basis, various ligands were designed and synthesized for the further characterization of this elusive bin<strong>di</strong>ng<br />

site. In particular, the synthesis of new ligands involved mo<strong>di</strong>fications of guanosine at the 6- and 5-positions.<br />

These molecules were tested through an innovative Eu-GTP bin<strong>di</strong>ng assay, which, by using the time-resolved<br />

fluorescence technique, allows to assess the coupling of a receptor to a G protein.<br />

The tested compounds seems to activate a GPCR with <strong>di</strong>fferent potencies, in some case even higher than that of<br />

guanosine itself.<br />

References:<br />

(1) Ciccarelli, R.; Di Iorio, P.; Giuliani, P.; D’Alimonte, I; Ballerini, P.; Caciagli, F.; Rathbone, M.P. Glia 1999, 25, 1, 93-98.<br />

(2) Traversa, U.; Bombi, G.; Di Iorio, P.; Ciccarelli, R.; Werstiuk, E. S.; Rathbone, M. P. Br. J. Pharmacol. 2002, 135, 4, 969-<br />

976.<br />

(3) Traversa, U.; Bombi, G.; Camaioni, E.; Macchiarulo, A.; Costantino, G.; Palmieri, C.; Caciagli, F.; Pellicciari, R. Bioorg.<br />

Med. Chem. 2003, 11, 24, 5417-5425.<br />

P-04


SYNTHESIS OF NOVEL LIGANDS FOR P2X3 RECEPTOR<br />

Deepak Shankar Panmand, 1 Catia Lambertucci, 1 Diego Dal Ben, 1 Gabriella Marucci, 1 Rosaria Volpini, 1<br />

Andrea Nistri, 2 Gloria Cristalli 1<br />

1 School of Pharmacy, Me<strong>di</strong>cinal <strong>Chemistry</strong> Unit, University of Camerino, I-62032 Camerino, Italy; 2 Sector of Neurobiology<br />

and Italian Institute of Technology Unit, International School for Advanced <strong>Stu<strong>di</strong></strong>es (SISSA), Via Beirut 2/4, 34014 Trieste,<br />

Italy<br />

117<br />

Purinergic receptors are involved in various physiological and pathological con<strong>di</strong>tions like brain trauma ischemia,<br />

and neurodegenerative <strong>di</strong>seases. They are classified into P1 and P2 receptors activated by adenosine and several<br />

nucleotides, respectively. Among P2 receptors the P2X subtypes are ligand gated ion channels while P2Y subtypes<br />

are G-protein coupled receptors. P2X3 subtype is activated by adenosine-5’-triphosphate (ATP) and it plays an<br />

important role in neuropathic and chronic pain pathways. Hence, ligands able to block these receptors could be<br />

useful for the treatment of chronic pain con<strong>di</strong>tions and migraine (1).<br />

Recently, a number of purine acyclic-nucleotides have been synthesized and proved to behave as partial agonists<br />

of P2X3 receptors (2). Furthermore, since non-nucleotide <strong>di</strong>ammino-pyrimi<strong>di</strong>ne derivatives were reported to block<br />

P2X3 receptors with high potency (3), a new series of P2X3 ligands was designed taking into account the structure<br />

of the acyclic nucleotides and molecular modeling data obtained by comparing pyrimi<strong>di</strong>ne and purine scaffolds.<br />

Hence, purine derivatives bearing a substituted benzyl chain in 9-position were synthesized and evaluated on<br />

HEK293 cells expressing rat P2X3 receptors using patch clamp technique.<br />

Results demonstrated that the new compounds behave as P2X3 receptor antagonists, endowed with IC50 activity in<br />

the micromolar range.<br />

References:<br />

(1) Burnstock G. Pharmacol. Rev. 2006, 58, 58–86.<br />

(2) Volpini, R.; Mishra, R. C.; Kachare, D. D.; Dal Ben, D.; Lambertucci, C.; Antonini, I.; Vittori, S.; Marucci, G.; Sokolova,<br />

E.; Nistri, A.; Cristalli, G. J. Med. Chem. 2009, 52, 4596–4603.<br />

(3) Carter, D. S.; Alam, M.; Cai, H.; Dillon, M. P.; Ford, A. P. D. W.; Gever, J. R.; Jahangir, A.; Lin, C.; Moore, A. G.;<br />

Wagner, P. J.; Zhai, Y. Bioorg. Med. Chem. Lett. 2009, 19, 1628-1631.<br />

P-05


118<br />

ALLOSTERIC ACTIVITY OF POTENT CIRAZOLINE ANALOGUES<br />

TOWARDS ADRENERGIC AND IMIDAZOLINE RECEPTORS<br />

Marialessandra Contino, a Nicola A. Colabufo, a Roberto Perrone, a Maria Pigini, b Francesca Ghelfi, b Russell<br />

Thomas c and Antonio Carrieri a<br />

a Dipartimento Farmaco-Chimico, <strong>Università</strong> <strong>di</strong> Bari, via Orabona 4, 70125 Bari, Italy<br />

b Scuola <strong>di</strong> Scienze del Farmaco e dei Prodotti della Salute, <strong>Università</strong> <strong>di</strong> Camerino, via S. Agostino 1, 62032 Camerino, Italy<br />

c Siena Biotech S.p.A., Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy<br />

Imidazoline receptors (I2R) are involved in the modulation of some behavioral states such as depression, anxiety<br />

and in vivo alterated receptor density in<strong>di</strong>cate their role in the modulation of catecholamines release in the CNS<br />

(1). The presence of these receptors has been also demonstrated in gastric and intestinal tissues. Imidazoline-like<br />

structures, such as cirazoline, have been characterized as modulators of cholinergic motor function in guinea-pig<br />

ileum by a <strong>di</strong>rect interaction with presynaptic α2D-adrenoceptors while an involvement of presynaptic imidazoline<br />

receptors was not reported (2). Moreover, an involvement of α1-adrenoceptors in me<strong>di</strong>ating relaxation of<br />

carbachol-evoked contraction in rat ileum has been showed (3) and an interaction between α1 adrenoceptors<br />

activation and I2R in the same organ has been hypothesized (4). Cirazoline and some structurally related<br />

compounds, previously characterized as potent α2-adrenoceptor ligands (5), are here stu<strong>di</strong>ed in isolated guinea pig<br />

ileum for determining I2, α1 post-junctional and α2 pre-junctional activities, and their pharmacological profiles,<br />

endowing <strong>di</strong>fferent (ant)agonsim together with allosterism, are presented.<br />

X<br />

O<br />

R<br />

NH<br />

N<br />

R X<br />

Cirazoline H cyclopropyl<br />

1 CH3 m-NO2-C6H4<br />

2 CH3 C6H5<br />

(1) Nutt, D. J.; French, N.; Handley, S.; Hudson, A.; Husbands, S.; Jackson, H.; Jordan, S.; Lalies, M. D.; Lewis, J.; Lione, L.;<br />

Mallard, N.; Pratt, J. In The Imidazoline <strong>Receptor</strong>. Pharmacology, Functions, Ligands, and Relevance to Biology and<br />

Me<strong>di</strong>cine; Reis, D.J.; Bousquet, P.; Parini, A.; Eds.; The New York Academy of Sciences: New York, 1995, Vol. 763, 125-139.<br />

(2) Colucci, R.; Blan<strong>di</strong>zzi, C.; Carignani, D.; Placanica, G.; Lazzieri, G.; Del Tacca, M. Effects of imidazoline derivatives on<br />

cholinergic motility in guineapig ileum: involvement of presynaptic alpha2-adrenoceptors or imidazoline receptors? Naunyn-<br />

Schmiedeberg’s Arch. Phrmacol. 1998, 357, 682-691.<br />

(3) Liu, L.; Coupar, I. M. Characterisation of pre- and postsynaptic-adrenoceptors in a modulation of rat ileum longitu<strong>di</strong>nal and<br />

circular muscle activities. Naunyn Schmiedeberg’s Arch Pharmacol. 1997, 356, 248-225.<br />

(4) MacInnes, N.; Handley, S. L. Potential serotonergic and noradrenergic involvement in the <strong>di</strong>scriminative stimulus effects of<br />

the selective imidazoline I 2-site ligand 2-BFI. Pharmacol. Biochem. Behav. 2003, 73, 427-433.<br />

(5) Crassous, P. A.; Car<strong>di</strong>naletti, C.; Carrieri, A.; Bruni, B.; Di Vaira, M.; Gentili, F.; Ghelfi, F.; Giannella, M.; Paris, H.;<br />

Piergentili, A.; Quaglia, W.; Schaak, S.; Vesprini, C.; Pigini, M. α 2-adrenoreceptors profile modulation. 3. (R)-(+)-mnitrobiphenyline,<br />

a new efficient and α 2C-subtype selective agonist. J. Med. Chem. 2007, 50, 3964-3968.<br />

P-06


ENANTIOSELECTIVE INTERACTIONS TO IMPROVE ADRENERGIC<br />

α2C-AGONISM/α2A-ANTAGONISM<br />

Fabio Del Bello, a Laura Mattioli, a Aniket Farande, a Francesca Ghelfi, a Mario Giannella, a Alessandro Piergentili, a<br />

Wilma Quaglia, a Marina Perfumi, a Russell Thomas, b Carla Marchioro, c Michele Dal Cin, c Maria Pigini a<br />

119<br />

a Scuola <strong>di</strong> Scienze del Farmaco e dei Prodotti della Salute, <strong>Università</strong> <strong>di</strong> Camerino, via S. Agostino, 1, 62032 Camerino, Italy<br />

b Siena Biotech S.p.A., Strada del Petriccio e Belriguardo, 35, 53100 Siena, Italy<br />

c GlaxoSmithKline Me<strong>di</strong>cine Research Center, via Fleming, 4, 37135 Verona, Italy<br />

Recently (1) we demonstrated that compounds 1 and 2 behaved as potent and selective α2C-AR agonists and<br />

effective α2A-AR antagonists. It is known that in clinical pain management, α2C selective agonists devoid of the<br />

sedative side effects associated with α2A-AR stimulation might represent alone or in combination with opioid<br />

analgesics an improvement over current therapies with cloni<strong>di</strong>ne-like drugs. Therefore, 1 and 2 co-administered<br />

with morphine were tested using the mouse tail-flick test. A very low dose (0.05 mg/kg) of both compounds<br />

caused a significant increase in morphine analgesia comparable to that obtained with a 0.5 mg/kg dose of<br />

cloni<strong>di</strong>ne. In ad<strong>di</strong>tion, 1 and 2 were devoid of sedative side effects. This promising behaviour is being<br />

strengthened by the observation that 1 and 2 are endowed with good chemical stability and exhibit high passive<br />

<strong>di</strong>ffusion through an artificial phospholipid membrane (PAMPA), which could permit them to interact with their<br />

target at a non-surface exposed bin<strong>di</strong>ng site. The presence of a stereocentre in 1 and 2 prompted us to investigate<br />

the stereochemical requirements for a possible α2C-AR agonism/α2A-AR antagonism improvement. Therefore, the<br />

novel R and S enantiomers of 1 and 2 were prepared and their biological profiles were determined by bin<strong>di</strong>ng and<br />

functional assays. Moreover, their effects on morphine analgesia modulation were evaluated. In ad<strong>di</strong>tion, for<br />

racemic compound 1 and its correspon<strong>di</strong>ng enantiomers (R)-(-)-1 and (S)-(+)-1 the study was also extended to the<br />

acquisition and expression of morphine tolerance and dependence.<br />

(±)-1<br />

(S)-(+)-1<br />

(R)-(−)-1<br />

(1) Car<strong>di</strong>naletti, C.; Mattioli, L.; Ghelfi, F.; Del Bello, F.; Giannella, M.; Buzzone, A.; Paris, H.; Perfumi, M.;<br />

Piergentili, A.; Quaglia, W.; Pigini, M. J. Med. Chem. 2009, 52, 7319-7322.<br />

P-07<br />

(±)-2<br />

(S)-(+)-2<br />

(R)-(−)-2


120<br />

NOVEL MUSCARINIC ANTAGONISTS DESIGNED ON THE 1,4-<br />

DIOXANE SCAFFOLD<br />

Fabio Del Bello, a Aniket Farande, a Yogita Farande, a Mario Giannella, a Maria Pigini, a Alessandro Piergentili, a<br />

Wilma Quaglia, a Marta Nesi, b Rosanna Matucci b<br />

a Scuola <strong>di</strong> Scienze del Farmaco e dei Prodotti della Salute, <strong>Università</strong> <strong>di</strong> Camerino, Via S. Agostino 1, 62032 Camerino, Italy<br />

b Dipartimento <strong>di</strong> Farmacologia, <strong>Università</strong> <strong>di</strong> Firenze, Viale G. Pieraccini 6, 50139 Firenze, Italy<br />

The five subtypes of muscarinic receptors (M1-M5) are widely <strong>di</strong>stributed in multiple organs and tissues and are<br />

critical to the maintenance of central and peripheral cholinergic neurotrasmission. Smooth muscle expresses<br />

several muscarinic receptor subtypes, the most important being M2 and M3 subtypes from a functional point of<br />

view. Although the M2 muscarinic subtypes are predominant, the smaller population of M3 subtypes appears to be<br />

the most functionally important for treating various me<strong>di</strong>cal con<strong>di</strong>tions associated with improper smooth muscle<br />

function, such as overactive bladder (OAB), chronic obstructive pulmonary <strong>di</strong>sease (COPD), and pain-<br />

predominant irritable bowel syndrome (IBS). Due to the wide <strong>di</strong>stribution of muscarinic receptors in the body,<br />

significant systemic exposure to muscarinic antagonists is associated with effects such as dry mouth, constipation,<br />

mydriasis, urinary retention and tachycar<strong>di</strong>a. Therefore, the <strong>di</strong>scovery of novel muscarinic antagonists showing<br />

target organ selectivity would be very useful (1).<br />

The (1,4-<strong>di</strong>oxan-4-yl)-N,N,N-trimethylmethanaminium io<strong>di</strong>de nucleus has proved to be a suitable scaffold for its<br />

ability to give potent muscarinic agonists (1) or antagonists (2), depen<strong>di</strong>ng on the size of the substituent in position<br />

6 (methyl or bulkier groups, respectively) (2). Therefore, we designed, prepared and stu<strong>di</strong>ed, at all five muscarinic<br />

receptor subtypes, analogs obtained by replacing the hydrophobic portion in position 6 of 2 with groups of<br />

<strong>di</strong>fferent nature and bulk. The pharmacological profile of the novel compounds was evaluated by receptor bin<strong>di</strong>ng<br />

assays on chinese hamster ovary (CHO) cells, expressing the five human muscarinic receptor subtypes (hM1-hM5).<br />

From preliminary results, compound 3 showed at all subtypes nM affinity values, which are comparable to those<br />

of some compounds used in therapy for the treatment of OAB.<br />

H 3C<br />

O<br />

O<br />

(1) Peretto, I.; Petrillo, P.; Imbimbo, B. P. Med. Res. Rev. 2009, 29, 867-902.<br />

(2) Piergentili, A.; Quaglia, W.; Giannella, M.; Del Bello, F.; Bruni, B.; Buccioni, M.; Carrieri, A.; Ciattini, S. Bioorg. Med.<br />

Chem. 2007, 15, 886-896.<br />

N + (CH 3) 3 I -<br />

1 2<br />

O<br />

O<br />

P-08<br />

N + (CH 3) 3 I -<br />

O<br />

O<br />

3<br />

N + (CH 3) 3 I -


SYNTHESIS, BIOLOGICAL EVALUATION AND DOCKING STUDIES<br />

OF NR2B/NMDA RECEPTOR ANTAGONISTS<br />

Rosaria Gitto, a Laura De Luca, a Stefania Ferro, a Maria Rosa Buemi, a Francesca Scicchitano, b Giovambattista De<br />

Sarro, b Lara Costa, c Lucia Ciranna c and Alba Chimirri a<br />

a Dipartimento Farmaco-Chimico, <strong>Università</strong> <strong>di</strong> Messina, Viale Annunziata, 98168 Messina, Italy<br />

b Dipartimento <strong>di</strong> Me<strong>di</strong>cina Sperimentale e Clinica, <strong>Università</strong> Magna Græcia, Germaneto, 88100 Catanzaro, Italy<br />

c Dipartimento <strong>di</strong> Scienze Fisiologiche, <strong>Università</strong> <strong>di</strong> Catania, Viale Andrea Doria 6, 95125 Catania, Italy.<br />

121<br />

NMDA receptors (NMDARs) are involved in many physiological processes, such as neuronal development,<br />

learning and memory but also play important roles in pathological states of CNS, inclu<strong>di</strong>ng strokes, seizures, and<br />

pain. NMDARs are receptor complexes containing NR1 and NR2 subunits and, more rarely, NR3 subunit. NR2<br />

subunits exist as four subtypes, each encoded by a <strong>di</strong>stinct gene and named NR2A-D. It is well-known that the<br />

noncompetitive NR2B-antagonists, such as ifenpro<strong>di</strong>l (1) and Ro 25-6981 (2), have good potential as<br />

neuroprotective agents and have been shown to bind at the so-called NR2B ifenpro<strong>di</strong>l site. In previous stu<strong>di</strong>es we<br />

reported a combined ligand- and target-based approach which led to the identification of new selective ligands<br />

containing an indole scaffold (1-2). The most active compounds (e.g. 3) proved to prevent au<strong>di</strong>ogenic seizures in<br />

DBA/2 mice and reduce NMDA receptor-me<strong>di</strong>ated current in patch clamp experiments.<br />

N<br />

OH<br />

OH<br />

1, ifenpro<strong>di</strong>l 2, Ro 25-6981<br />

N<br />

OH<br />

OH<br />

R<br />

O<br />

N<br />

H<br />

3, R = H, OMe, OH<br />

In attempt to obtain new insights into ifenpro<strong>di</strong>l-like ligands we synthesized new indole derivatives and screened<br />

their receptor affinity. We also tested the anticonvulsant properties in DBA/2 mice and evaluated the functional<br />

activity by electrophysiological experiments. Moreover, docking experiments were carried out to rationalize the<br />

obtained biological results.<br />

(1) Gitto, R.; De Luca, L.; Ferro, S.; Occhiuto, F.; Samperi, S.; De Sarro, G.; Russo, E.; Ciranna, L.; Costa, L. Chimirri, A.<br />

ChemMedChem 2008, 3, 1539-1548.<br />

(2) Gitto, R.; De Luca, L.; Ferro, S.; Citraro, R.; De Sarro, G.; Costa, L.; Ciranna, L. Chimirri, A. Bioorg. Med. Chem. 2009,<br />

17, 1640-1647.<br />

Research supported by MiUR (PRIN 2008, Grant No 20085HR5JK_002)<br />

P-09<br />

N


122<br />

SYNTHESIS OF NEUROPEPTIDE S RECEPTOR LIGANDS<br />

Dania Giacobbe, 1 Diego Dal Ben, 1 Catia Lambertucci, 1 Gabriella Marucci, 1 Girolamo Calò, 2 Rosaria Volpini, 1<br />

Gloria Cristalli 1<br />

1 School of Pharmacy, Me<strong>di</strong>cinal <strong>Chemistry</strong> Unit, University of Camerino Via S. Agostino, 62032 Camerino, Italy; 2<br />

Department of Experimental and Clinical Me<strong>di</strong>cine, Section of Pharmacology, University of Ferrara, via Fossato <strong>di</strong> Mortara,<br />

44100 Ferrara, Italy<br />

Neuropeptide S (NPS) is a recently identified transmitter constituted by 20 amino acids and it is involved in the<br />

modulation of anxiety, arousal, food intake, and insomnia. NPS activates a recently dehorphanized G protein-<br />

coupled receptor (NPSR) and the NPSR is <strong>di</strong>stributed in many brain areas with high expression levels in cortex,<br />

hypothalamus, amygdala and multiple midline thalamic nuclei. Many of these areas have been functionally<br />

associated with arousal and processing of emotional behaviour and, on this basis, NPSR ligands could have<br />

therapeutic potential (1).<br />

Up to date, limited information is available about nonpeptide NPSR ligands, as only one series of bicyclic<br />

piperazine derivatives has been published in this regard (2). However these molecules have been only partially<br />

characterized, hence the synthesis of selected 1,1-<strong>di</strong>phenyltetrahydro-1H-oxazolo[3,4-a]pyrazin-3(5H)-one<br />

derivatives, bearing <strong>di</strong>fferent substituents in 7-position, was undertaken aimed at defining the structural figures<br />

relevant for NPSR antagonist activity.<br />

The synthesized compounds were tested in vitro in a calcium mobilization assay, performed using HEK293 cells<br />

expressing the recombinant mouse NPS receptor (HEK293mNPSR).<br />

Tested compounds showed to inhibit NPSR with KB activity in the nanomolar range and, in particular, it has been<br />

demonstrated that the CH2 linker in the side chain of the reference compound SHA 68 is not crucial for the<br />

potency.<br />

References:<br />

(1) Xu, Y. L.; Reinscheid, R. K.; Huitron-Resen<strong>di</strong>z, S.; Clark, S. D.; Wang, Z.; Lin, S. H.; Brucher, F. A.; Zeng, J.; Ly, N. K.;<br />

Henriksen, S. J.; de Lecea, L.; Civelli, O. Neuron. 2004, 43, 487-497.<br />

(2) Yanan Z.; Brian P. G.; Hernán A. N.; Scott P. R. Bioorg. Med. Chem. Lett. 2008, 18, 4064-4067.<br />

P-10


123<br />

OVERACTIVE BLADDER (OAB): A NEW THERAPEUTIC APPROACH.<br />

1-PIPERAZINYLCARBOXAMIDES AND ANALOGUES AS<br />

ALLOSTERIC ANTAGONISTS AT MGLU5 RECEPTOR<br />

Carlo Riva, Carlo De Toma, Matteo Longhi, Ilaria Bettinelli, Katia Karamfilova, Davide Graziani, Gianni Motta,<br />

Elena Poggesi, Amedeo Leonar<strong>di</strong>.<br />

Drug Discovery – Recordati SpA<br />

Overactive Bladder (OAB) is, in a societal perspective, a very onerous pathology, taking into account both <strong>di</strong>rect<br />

and in<strong>di</strong>rect costs 1 . Thus, looking for new pharmacological treatments of OAB, as a replacement of antimuscarinic<br />

drugs, which are not devoid of important side effects, could be an important issue.<br />

MGlu5 receptors have been shown to play a role in the voi<strong>di</strong>ng reflex and bladder nociception. Experimental data<br />

suggest that mGlu5 receptors play facilitator roles in the processing of afferent input from the urinary bladder, and<br />

that central rather than peripheral mGlu5 receptors appear to be responsible 2 .<br />

On these bases a new project was started, looking for mGluR5 allosteric antagonists (inverse agonist) by a<br />

multi<strong>di</strong>sciplinary approach, using as a starting point the structures of MTEP and MPEP (Figure 1).<br />

.<br />

Figure 1<br />

A series of 1-piperazinylpropargylamides and closed analogues was synthesized with the aim of defining the main<br />

features of the pharmacophore and the peculiar chemical groups, which could afford affinity for the mGlu5<br />

receptor and selectivity vs. the mGlu1 receptor.<br />

(1) Ganz, M.L.; Smalarz, A.M.; Krupski, T.L., Anger, J.T., Hu, J.C.; Wittrup-Jensen, K.U.; Pashos, C.L., Urology, 2009, Dec<br />

24 in print – available on-line.<br />

(2) Hu, Y.; Dong, L.; Sun, B.; Guillon, M.A.; Burbach, L.R.; Nunn, P.A.; Liu, X.; Vilenski, O.; Ford, A.P.D.W.; Zhong, Y.;<br />

Rong, W., Neuroscience Letters, 2009, 450 (1), 12-17.<br />

P-11


124<br />

NOVEL BIS(7)-TACRINE DERIVATIVES AS MULTITARGET<br />

LIGANDS: FOCUS ON ANTI-CHOLINESTERASE AND ANTI-<br />

AMYLOID ACTIVITIES<br />

Maria Laura Bolognesi, Manuela Bartolini, Francesca Mancini, Gianpaolo Chiriano, Luisa Ceccarini, Michela<br />

Rosini, Andrea Milelli, Vincenzo Tumiatti, Vincenza Andrisano, Carlo Melchiorre<br />

Department of Pharmaceutical Science, University of Bologna , Via Belmeloro 6, 40126 Bologna, Italy<br />

A vast array of drug <strong>di</strong>scovery research focuses on the development of innovative drugs targeting Alzheimer's<br />

<strong>di</strong>sease (AD) and related dementias. However, despite several lead can<strong>di</strong>dates have progressed to AD pre-clinical<br />

testing in the last decade, to date none has been successful in late-stages clinical trials. Beta-secretase (BACE-1)<br />

inhibition has been a strategy actively pursued, although progress in moving inhibitors to the clinic has been slow,<br />

partly as a consequence of its aspartic proteinase character, which hampers the development of potent, selective<br />

and brain-permeable compounds. At present, however, AD pharmacotherapy still relies on acetylcholinesterase<br />

inhibitors donepezil, rivastigmine and galantamine. In this scenario, we and others envisaged the development of<br />

Multi-Target Directed Ligands as an innovative and effective strategy to face the multifactorial etiopathogenesis of<br />

AD (1). This strategy, embo<strong>di</strong>ed on single chemical entities able to simultaneously modulate multiple targets<br />

involved in the neurodegenerative cascade, has proven particularly fruitful in the last couple of years and allowed<br />

to <strong>di</strong>scover several promising anti-Alzheimer's drug can<strong>di</strong>dates.<br />

Starting from this consideration, we have developed a new series of <strong>di</strong>meric ligands derived from the lead<br />

can<strong>di</strong>date bis(7)-tacrine, for which a multifunctional profile has been <strong>di</strong>sclosed (2). The effect of replacing the<br />

heptamethylene chain connecting the two monomeric tacrine units with polyaromatic spacers was evaluated at<br />

three validated drug <strong>di</strong>scovery targets, namely cholinesterases (acetyl- and butyrylcholinesterase) and amyloid<br />

formation (BACE-1) and aggregation.<br />

(1) Cavalli, A.; Bolognesi, M. L.; Minarini, A.; Rosini, M.; Tumiatti, V.; Recanatini, M.; Melchiorre, C. J. Med. Chem. 2008,<br />

51, 347-372.<br />

(2) Li, W.; Mak, M.; Jiang, H.; Wang, Q.; Pang, Y.; Chen, K.; Han, Y. Neorotherapeutics 2009, 6, 187-201.<br />

P-12


DISCOVERY OF NEW POTENTIAL γ-SECRETASE MODULATORS<br />

FOR THE TREATMENT OF ALZHEIMER’S DISEASE<br />

Dario Ambrosini*, Gianfabio Giorgioni*, Piera Sozio, § Laura S.Cerasa, § Antonio Di Stefano §<br />

Amelia Catal<strong>di</strong>, ç Susi Zara. ç<br />

*Scuola <strong>di</strong> Scienze del Farmaco e dei Prodotti della Salute, via S. Agostino 1, 62032, Camerino, Italy.<br />

§ Dipartimento <strong>di</strong> Scienze del Farmaco, <strong>Università</strong> “G. D’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy.<br />

ç Dipartimento <strong>di</strong> Biomorfologia, <strong>Università</strong> “G. D’Annunzio”, Via dei Vestini 31, 66100 Chieti, Italy.<br />

125<br />

The presence of Aβ-peptide containing plaques in the brain, is a hallmark of Alzheimer’s <strong>di</strong>sease (AD). Several<br />

stu<strong>di</strong>es have suggested a pathogenic role in AD for Aβ42, the longer form of the two Aβ-peptides, which has a<br />

strongest tendency to deposit insoluble plaques in the AD brain. Aβ peptides are produced from β-Amyloid<br />

Precursor Protein (APP) through the sequential cleavage by two proteases, namely β-secretase and γ-secretase.<br />

Firstly, β-secretase produces a C-terminal fragment of APP (CTFβ), then γ-secretase cleaves the CTFβ to produce<br />

Aβ peptides of <strong>di</strong>fferent lengths. 1<br />

Preventing the formation or intracellular deposition of Aβ42-peptide represents a strategy to stop or slow down the<br />

progression of the AD. At this purpose, inhibition of γ-secretase has proven to be an interesting target for drug<br />

<strong>di</strong>scovery. 2 One of the first compounds which efficaciously inhibited Aβ production in animal models of AD<br />

blocking γ-secretase activity was LY411575. 3 It has been demonstrated that inhibition of γ-secretase reduce Aβ in<br />

a murine model of AD, but has potentially undesirable biological effects, most likely due to of the inhibition of<br />

Notch cleavage, a transmembrane receptor involved in regulating cell-fate decision. 4 Thus, selective inhibition of<br />

Aβ42 production seems to be a promising alternative to the complete inhibition of γ-secretase activity. The pursuit<br />

of such a result was made possible by the surprising fin<strong>di</strong>ng that certain non-steroidal anti-inflammatory drugs<br />

(NSAIDs), inclu<strong>di</strong>ng flubiprofen, <strong>di</strong>splay preferential Aβ42-lowering activity without affecting Notch cleavage. 5<br />

The proposed mechanism for this activity is an allosteric modulation of γ-secretase activity, the enzyme<br />

responsible for the formation of Aβ.<br />

Starting from these data we proposed the synthesis of new LY411575 analogues that were designed to function as<br />

potential γ-secretase modulators able to penetrate the blood-brain barrier, the synthesis and preliminary<br />

pharmacological evaluation will be <strong>di</strong>scussed.<br />

(1) Olson, R.E. et al. Curr. Topics Med. Chem., 2008, 8, 17.<br />

(2) a) Churcher I. et al. Curr. Pharm. Des., 2005, 11, 3363; b) Kornilova A.Y., et al. Annu. Rep. Med. Chem., 2003, 38, 41; (c)<br />

John V. et al. J. Med. Chem., 2003, 46, 4625; (d) Josien H. Curr. Opin. Drug Discov. Devel., 2002, 5, 513.<br />

(3) a)Wu, J.; et al. Int. Patent Appl. WO 9828268, 1998. (b) Au<strong>di</strong>a, J. E.; et al. Int. Patent Appl. WO 2000019210, 2000.<br />

(4) Wong G.T. et al. J. Biol. Chem., 279, 2004, 12876–12882.<br />

(5) Czirr E. et al. Neurodegenerative Diseases, 2006, 3, 298.<br />

P-13


126<br />

WITHDRAWN<br />

P-14


PUTATIVE GUANOSINE RECEPTOR IDENTIFICATION IN RAT<br />

BRAIN BY THE INNOVATIVE EU EU-GTP GTP BINDING ASSAY<br />

Michela Buccioni, , Diego Dal Ben, Catia Lambertucci, Carmen Lammi, Gabriella Marucci, Ajiroghene Thomas,<br />

School of Pharmacy, Me<strong>di</strong>cinal <strong>Chemistry</strong> Unit, University of Camerino, 62032 Camerino, Italy Italy.<br />

The purine nucleoside guanosine anosine possesses many trophic effects, inclu<strong>di</strong>ng promotion, <strong>di</strong>vision, and growth of<br />

astrocytes and other neuron-like like cells. This nucleoside seems to act as signalling molecule via the activation of<br />

specific membrane receptors. 1 Competition stu<strong>di</strong>es, perfor performed med by Traversa et al. on rat brain membranes,<br />

confirmed that [ 3 H]-guanosine guanosine site was <strong>di</strong>stinct from the well characterized ATP and adenosine bin<strong>di</strong>ng sites. 2<br />

Starting from these observations and aimed at developing a new assay that allows to evaluate the po potency of<br />

ligands at the putative guanosine receptor, Eu Eu-GTP GTP assay was performed in rat brain. This technique, using the<br />

principle of the [ 35 S]GTPgammaS bin<strong>di</strong>ng assay, replaces the ra<strong>di</strong>oactive material with Eu Eu-GTP and exploits the<br />

unique fluorescence properties ties of Europium lanthanide chelate.<br />

Furthermore, since potent and selective ligands are highly needed for the new putative guanosine receptor<br />

characterization, new molecules were synthesized and tested using Eu Eu-GTP assay.<br />

The results showed that some new compounds are able to activate the putative guanosine receptor with a potency<br />

higher than that of guanosine itself. On the other hand, this functional assay allows to assume the presence in rat<br />

brain of a GPCR activated by guanosine.<br />

References:<br />

(1) Caciagli, F.; Illes, P. Purinergic Signal. 2005, 1, 297-298.<br />

(2) Traversa, U.; Bombi, G.; Camaioni Camaioni, E.; Macchiarulo, A.; Costantino, G.; Palmieri, C.; Caciagli Caciagli, F.; Pellicciari, R. Bioorg.<br />

Med. Chem. 2003, 11(24), 5417-5425. 5425.<br />

RosariaVolpini, Gloria Cristalli<br />

P-15<br />

127


128<br />

PARALLEL SYNTHESIS AND EVALUATION OF 2,5-DIAMINO-1,4-<br />

BENZOQUINONES AS A NOVEL CLASS OF BIFUNCTIONAL ANTI-<br />

PRION COMPOUNDS<br />

Salvatore Bongarzone, 1, 2 Hoang Ngoc Ai Tran, 3 Andrea Cavalli, 4,5 Marinella Roberti, 4 Paolo Carloni, 1,2,6,7<br />

Giuseppe Legname, 2,3 and Maria Laura Bolognesi 4<br />

1 Statistical and Biological Physics Sector, International School for Advanced <strong>Stu<strong>di</strong></strong>es (SISSA), 34151 Trieste, Italy.<br />

2 SISSA-Unit, Italian Institute of Technology, 34151 Trieste, Italy.<br />

3 Neurobiology Sector, International School for Advanced <strong>Stu<strong>di</strong></strong>es (SISSA), 34151 Trieste, Italy.<br />

4 Department of Pharmaceutical Sciences, Alma Mater <strong>Stu<strong>di</strong></strong>orum, University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy.<br />

5 Department of Drug Discovery and Development, Italian Institute of Technology, 16163 Genova, Italy.<br />

6 CNR-INFM-DEMOCRITOS Modeling Center for Research in Atomistic Simulation, 34151 Trieste, Italy.<br />

7 German Research School for Simulation Sciences GmbH, Forschungzentrum Jülich GmbH - RWTH Aachen University.<br />

Prion <strong>di</strong>seases, also known as transmissible spongiform encephalopathies (TSE), are fatal neurodegenerative<br />

<strong>di</strong>sorders of the central nervous system. The main molecular mechanism underlining TSE is based on the aberrant<br />

misfol<strong>di</strong>ng of the cellular form of the prion protein (PrP C ) into its pathological counterpart denominated PrP Sc . To<br />

date there are no identified therapies. One therapeutic strategy against this <strong>di</strong>sease is focused on the stabilization of<br />

the PrP C in order to prevent its conversion to PrP Sc (1). PrP Sc is involved in fibrillation processes and thus play a<br />

pivotal role in the pathogenesis of TSEs. Crucial elements me<strong>di</strong>ating fibrillation event are protein-protein<br />

interactions (PPIs). Although me<strong>di</strong>cinal chemistry approaches considering PPIs as drug target are daunting<br />

challenge, recent stu<strong>di</strong>es showed that small molecules bearing homo<strong>di</strong>meric structure properly spaced have shown<br />

possible therapeutic effect (2). Buil<strong>di</strong>ng on the bivalent approach, we have recently reported that 2,5-<strong>di</strong>amino-1,4benzoquinone<br />

(BQ) linked with two phenyl rings by a spacer <strong>di</strong>splayed remarkable anti-prion activity in a cellular<br />

model (EC50 = 0.87 µM) (3). This allowed us to propose that the planar BQ scaffold might be considered as a<br />

privileged motif in modulating PPIs and as a promising central core in the search for bifunctional anti-prion<br />

compounds. Starting from these results, a small library featured a BQ nucleus as central core, with two spacers in<br />

position 2 and 5 connected to two terminal moieties was designed. Designed entries were made by parallel<br />

synthesis, and evaluated against prion infection. Some of them have shown inhibition against PrP Sc aggregation in<br />

ScGT1 cell line. The most promising anti-prion compounds (quinoline derivatives) have shown low toxicity and<br />

capability to inhibit prion fibril formation in vitro.<br />

PrP C PrP Sc fibril of PrP Sc<br />

O O<br />

H<br />

Cl N<br />

O<br />

H H<br />

O<br />

N Cl<br />

H<br />

O O<br />

EC50= 0.87 µM<br />

(1) Caughey, B.; Baron, G. S. Nature 2006, 443, 803-810<br />

(2) May, B. C. H.; Fafarman, A. T.; Hong, S. B.; Rogers, M.; Deady, L. W.; Prusiner, S. B.; Cohen, F.; .Proc. Natl. Acad. Sci. U. S. A. 2003,<br />

100, 3416-3421.<br />

P-16<br />

N<br />

Cl<br />

H<br />

N<br />

N<br />

H<br />

O<br />

O<br />

H<br />

N<br />

N<br />

H<br />

N<br />

Cl


AMINOPYRIMIDINES AS INHIBITORS OF THE WNT SIGNALLING<br />

PATHWAY<br />

Fabio Del Bello, a Aniket Farande, a Mario Giannella, a Alessandro Piergentili, a Wilma Quaglia, a Arianna Nencini, b<br />

Russell Thomas, b Maurizio Varrone, b Tiziana Benicchi, b Giovanni Benedetti, b Alessandra De Robertis, b<br />

Silvia Valensin b<br />

129<br />

a Scuola <strong>di</strong> Scienze del Farmaco e dei Prodotti della Salute, <strong>Università</strong> <strong>di</strong> Camerino, via S. Agostino 1, 62032 Camerino, Italy<br />

b Siena Biotech S.p.A., Strada del Petriccio e Belriguardo 35, 53100 Siena, Italy<br />

The aberrant activation of the Wnt signalling pathway is believed to be a major driving force in a broad range of<br />

pathological con<strong>di</strong>tions and is common in many types of human tumors. Inhibition of the pathway has been shown<br />

to block tumor growth giving great therapeutic potential to drugs targeting this pathway. 1<br />

In a program aimed at the identification of small molecules inhibitors of the Wnt pathway for the treatment of<br />

glioblastoma (GBM) we identify a series of aminopyri<strong>di</strong>nes which specifically inhibit the pathway without<br />

affecting cell viability. This series of compounds has been expanded with the synthesis of a focused set of<br />

aminopyrimi<strong>di</strong>nes with two points of <strong>di</strong>versity. All compounds have been tested for their activity on the Wnt<br />

pathway using DBTRG cells stably transfected with TCF-Luciferase and TA-Renilla. The TCF-Luc reporter gene<br />

specifically responds to Wnt stimuli while the Renilla readout is Wnt independent and can be used as cell viability<br />

readout. In ad<strong>di</strong>tion, in order to identify potential ADME liabilities, all compounds were tested for their in vitro<br />

solubility, metabolic stability and permeability.<br />

(1) Barker, N.; Clevers, H. Nat. Rev. Drug Discov. 2006, 5, 997-1014.<br />

This work was supported by the Monte dei Paschi <strong>di</strong> Siena Foundation Award<br />

P-17


130<br />

INSECTICIDAL HETEROLIGNANS – IN VIVO SAR STUDY OF<br />

POTENT TUBULINE POLYMERIZATION INHIBITORS WITH<br />

ACTIVITY AGAINST CHEWING PESTS<br />

Jens Frackenpohl a , Isabelle Adelt b , Horst Antonicek b , Christian Arnold b , Nicole Blaha a , Sabine<br />

Hohmann a , Elisabeth Peschel a , Thomas Schenke b , Robert Velten b , Hans-Christoph Weiss c<br />

a Bayer CropScience AG, Industriepark Höchst, G836, D-65926 Frankfurt a. Main<br />

b Bayer CropScience AG, Alfred-Nobel-Strasse 50, D-40789 Monheim<br />

c Currenta GmbH, Chemiepark Leverkusen, Q18, D-51368 Leverkusen<br />

Starting from natural product podophyllotoxin substituted heterolignans were identified with promising<br />

insecticidal in vivo activity. The impact of substitution in each segment of the core structure was<br />

investigated in a detailed in vivo-SAR study, and variation of substituents in both aromatic moieties<br />

afforded promising derivatives with broad insecticidal activity against lepidopteran and coleopteran<br />

species. In vitro measurements supported by modeling stu<strong>di</strong>es in<strong>di</strong>cate that heterolignans investigated in<br />

our study act as potent tubuline polymerization inhibitors interacting with the colchicine-bin<strong>di</strong>ng site.<br />

Various unprecedented substituents have been introduced to fully explore the structure-insecticidal<br />

activity relationship. Thus, insect specific structure-activity effects were observed showing that the<br />

insecticidal SAR described herein <strong>di</strong>ffers from reported cytotoxicity stu<strong>di</strong>es.<br />

O<br />

O<br />

A<br />

O<br />

OH<br />

H<br />

B C O<br />

D<br />

O<br />

H<br />

O<br />

O<br />

Podophyllotoxin Deoxypodophyllotoxin<br />

[1] J. Frackenpohl et al., WO 05097802, 2005.<br />

[2] J. Frackenpohl et al., Bioorg. Med. Chem., 2009, 17, 4160-4184.<br />

O<br />

O<br />

O<br />

O<br />

H<br />

H<br />

O<br />

O<br />

O<br />

O<br />

O<br />

P-18<br />

6<br />

5<br />

7 8<br />

O<br />

H<br />

N 4<br />

O<br />

9<br />

3<br />

O<br />

1<br />

O<br />

O<br />

O<br />

O<br />

O<br />

H<br />

N<br />

O<br />

O<br />

O


TARGETING PROTEIN PROTEIN-PROTEIN PROTEIN INTERACTIONS FOR NOVEL<br />

ANTICANCER THERAPIES<br />

THERAPIES: : MOLECULAR DYNAMICS OF SMALL<br />

MOLECULES INHIBITING THE p53-MDM2 MDM2 COMPLEX.<br />

Nicola Giacchè Giacchè, Antonio Macchiarulo, Roberto Pellicciari.<br />

Dipartimento <strong>di</strong> Chimica e Tecnologia del Farmaco, <strong>Università</strong> <strong>di</strong> Perugia, via del Liceo 1, 06123 Perugia.<br />

MDM2 is an oncogenic protein that regulates the activity and stability of p53. The evidence that more than 50% of<br />

human cancers show altered regulations of p53 and overexpressed levels of MDM2, has fu fuelled the search for new<br />

therapeutic agents that could rescue p53 from the inhibition of MDM2. 1-2 Accor<strong>di</strong>ngly, during the last decade,<br />

<strong>di</strong>verse small molecules have been <strong>di</strong>sclosed as able to <strong>di</strong>srupt the interaction between MDM2 and p53. On this<br />

basis, experimental imental and computational approaches have been developed to study the interaction of small molecules<br />

with the p53 bin<strong>di</strong>ng cleft of MDM2, aimed at facilitating the bench to bedside translation of new potent and<br />

selective chemical entities as novel anticance anticancer therapies. 3<br />

In this framework, as continuation of our efforts in the field, 4-5 we report the results of molecular dynamic<br />

simulations carried out to study the conformational transitions of MDM2 in response to small molecule bin<strong>di</strong>ng.<br />

Our study points out a con<strong>di</strong>tional active role of the NN-terminal<br />

region (1-25 25 residues) of MDM2 in the molecular<br />

recognition of inhibitors, that is dependent on the specific chemical class of the ligand. The results obtained will<br />

prove useful to drive me<strong>di</strong>cinal chemistry in de designing signing new MDM2 inhibitors with improved potency and<br />

selectivity.<br />

(1) Vogelstein, B.; Lane, D.; Levine, AJ. Nature 2000, 408, 307-310; 310; (2) Toledo, F.; Whal, GM. Nat. Rev. Cancer 2006, 6,<br />

909-923; (3) Patel, S.; Player, MR. Expert Opin. Investig. Drugs 2008, 17, 1865-1882; 1882; (4) Macchiarulo, A.; Giacchè, N.;<br />

Carotti, A.; Baroni, M.; Cruciani, G.; Pellicciari, R. J. Chem. Inf. Model. 2008, 48, 1999-2009; 2009; (5) Carotti, A.; Macchiarulo, A.;<br />

Giacchè, N.; Pellicciari, R. Proteins 2009 2009, 77, 524-535.<br />

Acknowledgements:<br />

This work was supported by the European Union. FP6 PRIORITY LSH LSH-2005-2.2.0-8: Small-ligand ligand libraries: improved tools<br />

for exploration and prospective anti-tumour tumour therapy. DePPICT Project (Designing Therapeutic Protein Protein-Protein Inhibitors for<br />

Brain n Cancer Treatments) Contract number: LSHC LSHC-CT-2007- 037834 (http://www.deppict.eu/home.jsp)<br />

P-19<br />

131


132<br />

WITHDRAWN<br />

P-20


DESIGN AND SYNTHESIS OF Smo ANTAGONISTS FOR TREATMENT<br />

OF CANCER<br />

133<br />

Annette Bakker, Marta Bellini, Giovanni Benedetti, Chiara Caramelli, Pietro Ferruzzi, Simone Galeazzi, Giacomo<br />

Minetto, Gal.la Pericot Mohr, Andrea Nuzzi, Alessandro Panico, Francesco Russo and Russell Thomas<br />

Me<strong>di</strong>cinal <strong>Chemistry</strong> Department, Siena Biotech S.p.A., Strada del Petriccio e Belriguardo, 35. 53100, Siena, Italy<br />

gminetto@sienabiotech.it.<br />

The Hedgehog pathway plays an important role in the embryonic patterning and development of many tissues and<br />

somatic structures as well as maintaining and repairing mature tissues in adults. Uncontrolled activation of this<br />

pathway has been implicated in several cancers inclu<strong>di</strong>ng glioblastoma, melanoma, basal cell carcinoma and<br />

breast, lung, prostate and pancreatic cancers. The aberrant activation of Hedgehog signaling in several cancers has<br />

made it an attractive target for anticancer drug <strong>di</strong>scovery (1-2).<br />

Most of the me<strong>di</strong>cinal chemistry efforts have been <strong>di</strong>rected towards targeting Smo, a transmembrane protein that<br />

plays a central role in modulating Gli transcription factors and initiating transcription of Hh target genes. A<br />

number of Smo inhibitors have been reported and they can be classified as cyclopamine analogues or synthetic<br />

small molecule antagonists (3).<br />

Different classes of molecules have been designed in Siena Biotech as Smo inhibitors for treatment of<br />

glioblastoma. Great attention has been put towards the physicochemical properties of such molecules in order to<br />

facilitate their subsequent optimization.<br />

P-21


134<br />

DESIGN, SYNTHESIS, AND BIOLOGICAL EVALUATION OF<br />

NAPHTHALENE DIIMIDES DERIVATIVES AS ANTICANCER AGENTS<br />

A. Milelli 1 , A. Minarini 1 , C. Melchiorre 1 , M. Micco 1 , E. Simoni 1 , G. Zuccari 1 , L. Raffaghello 3 , C. Stefanelli 2 ,<br />

M. Zini 2 , V. Tumiatti 1<br />

1 Department of Pharmaceutical Sciences, Via Belmeloro 6, 2 Department of Biochemistry<br />

“G. Moruzzi”, Via Irnerio 48, Alma Mater <strong>Stu<strong>di</strong></strong>orum, University of Bologna, 40126 Bologna, Italy<br />

3 Laboratory of Oncology, Giannina Gaslini Institute, Genova, Italy.<br />

The search for novel chemotherapeutic agents and approaches to cancer treatment is an active research field<br />

stimulated by the <strong>di</strong>scovery of new biological targets and by the possibility of obtaining new drugs without serious<br />

and undesirable side effects. Several anticancer agents have been developed so far based on <strong>di</strong>fferent mechanism<br />

of action; despite this, intercalator agents still remain an interesting class of molecules to focus on. In particular, in<br />

the literature there are several examples of Naphthalimmide and 1,4,5,8-Naphtalentetracaboxylic <strong>di</strong>imide<br />

derivatives as intercalating and anticancer agents. It is well-known that, at physiological pH, protonated<br />

polyamines interact strongly with the phosphate residues of DNA. Therefore, the inclusion of these basic<br />

functionalities, on an intercalating moiety, may improve the interaction with DNA structure.<br />

Recently, we reported on the design and synthesis of substituted Naphtalen(<strong>di</strong>)immides as anticancer agents (1).<br />

Among the several developed compounds, 1 and 2 were the most interesting derivatives showing the ability to<br />

bind DNA, trigger caspase activation, cause accumulation of p53 protein, down-regulate the survival kinase AKT,<br />

cause a decrease of ERK1/2 and, inhibit ERK’s phosphorylation.<br />

OCH 3<br />

H<br />

N<br />

O<br />

N<br />

n<br />

O<br />

1: n = 1<br />

2: n = 2<br />

The aim of the present work is to further investigate the promising biological profile of 1 and 2, and the role of the<br />

2-methoxy substituents on the benzyl moieties. For this purpose several analogues, with <strong>di</strong>fferent substituents on<br />

the two aromatic rings, were synthesized and evaluated for their antiproliferative activity.<br />

This research was supported by grants from MUR, the University of Bologna, and Polo Scientifico-Didattico <strong>di</strong> Rimini. We<br />

thank the National Cancer Institute for the anticancer assays.<br />

(1) Tumiatti, V., Milelli, A., Minarini, A., Micco, M., Gasperi Campani, A., Roncuzzi, L., Baiocchi, D., Marinello, J.,<br />

Capranico, G., Zini, M., Stefanelli, C., Melchiorre C. J. Med. Chem. 2009, 52, 7873-7.<br />

O<br />

N<br />

O<br />

P-22<br />

n<br />

H<br />

N<br />

OCH 3


SULFONATE BILE ACID DERIVATIVES AS POTENT AND<br />

SELECTIVE MODULATORS OF TGR5, A NEW TARGET FOR<br />

DIABESITY<br />

Paola Sabbatini, 1 Antimo Gioiello, 1 Antonio Macchiarulo, 1 Emiliano Rosatelli, 1 Gianni Rizzo, 2 Luciano Adorini, 2<br />

Johan Auwerx, 3 Charles Thomas, 3 Roberto Pellicciari. 1<br />

1) <strong>Università</strong> <strong>degli</strong> <strong>Stu<strong>di</strong></strong> <strong>di</strong> Perugia, Dipartimento <strong>di</strong> Chimica e Tecnologia del Farmaco, Via del Liceo, 1, 06100 Perugia,<br />

Italy. 2) Intercept Italia, Via P. Togliatti, 22,06073 Corciano, Italy. 3) Institut Clinique de la Souris, 67404 Illkirch, France.<br />

135<br />

TGR5 is a bile acid (BA) membrane receptor, G-protein coupled to the adenylate cyclase. On the basis of<br />

sequence similarity stu<strong>di</strong>es, it belongs to the group of MECA (melanocortin, endothelial, cannabinoid and<br />

adenosine) receptors, inside the rhodopsine-like family of GPCRs. Recent years have witnessed a growing interest<br />

in TGR5 as an attractive target for obesity and type-2-<strong>di</strong>abetes, due to its roles in the regulation of glucose<br />

metabolism and energy homeostasis. 1 These effects are linked to the ability of the receptor to stimulate type 2<br />

iodothyronine deio<strong>di</strong>nase (D2) activity in brown a<strong>di</strong>pose tissue (BAT) and muscle, as well as induce the release of<br />

glucagon-like peptide 1 (GLP-1) in enteroendocrine cells. 1<br />

In this scenario, we have been committed toward the identification of novel BA derivatives as potent and selective<br />

agonists for TGR5. Our efforts have hitherto resulted in the <strong>di</strong>sclosure of S-EMCA (INT-777), a compound that<br />

has been instrumental to provide the first proof-of-concept that the pharmacological modulation of TGR5 may<br />

indeed represent a novel viable strategy for the treatment of type 2 <strong>di</strong>abetes. 2,3 On the basis of our preliminary<br />

QSAR study, 4 and as a continuation of our work in the field, we have further investigated the effect of<br />

mo<strong>di</strong>fications on the BA side chain. In particular, in this communication we report the design, synthesis and<br />

preliminary biological appraisals of a new series of BA derivatives, bearing bioisosteric substitution of the<br />

carboxylic group with sulfonate moieties.<br />

References:<br />

(1) Thomas, C.; Pellicciari, R.; Pruzanski, M.; Auwerx, J. Schoonjans, K. Nat. Rev. Drug Disc. 2008, 7(8), 678-693.<br />

(2) Pellicciari, R.; Gioiello, A.; Macchiarulo, A.; Thomas, C.; Rosatelli, E.; Natalini, B.; Sardella, R.; Pruzanski, M.; Roda,<br />

A.; Pastorini, E.; Schoonjans, K.; Auwerx, J. J. Med. Chem. 2009, 52 (24), 7958–7961.<br />

(3) Thomas, C.; Gioiello, A.; Noriega, L.; Strehle1, A.; Oury1, J.; Rizzo, G.; Macchiarulo, A.; Yamamoto, H.; Mataki, C.;<br />

Pruzanski, M.; Pellicciari, R.; Auwerx, J.; Schoonjans, K. Cell Met. 2009, 10(3), 162-164.<br />

(4) Macchiarulo, A.; Gioiello, A.; Thomas, C.; Massarotti, A.; Nuti, R.; Rosatelli, E.; Sabbatini, P.; Schoonjans, K.;<br />

Auwerx, J.; Pellicciari, R. J. Chem. Inf. Model. 2008, 48 (9), 1792–1801.<br />

P-23


136<br />

DIKETOPIPERAZINE-BASED LIGANDS OF PRION PROTEIN<br />

Maria Laura Bolognesi, b Matteo Staderini, a Hoang Ngoc Ai Tran, d Alessandra Monaco, a Salvatore Bongarzone, ce<br />

Xevi Biarnés, ce Pilar López-Alvarado, a Nieves Cabezas, a Paolo Carloni, ce J. Carlos Menéndez, a and Giuseppe<br />

Legname de<br />

aDepartamento de Química Orgánica y Farmacéutica, Facultad de Farmacia, Universidad Complutense, 28040 Madrid,<br />

Spain. bDepartment of Pharmaceutical Sciences, Alma Mater <strong>Stu<strong>di</strong></strong>orum, University of Bologna, Via Belmeloro 6, 40126<br />

Bologna, Italy. cStatistical and Biological Physics Sector, dNeurobiology Sector, International School for Advanced <strong>Stu<strong>di</strong></strong>es<br />

(SISSA), eSISSA-Unit, Italian Institute of Technology, 34014 Trieste, Italy.<br />

Prion <strong>di</strong>seases, or transmissible spongiform encephalopathies (TSEs) are a family of invariably fatal<br />

neurodegenerative <strong>di</strong>sorders for which no effective curative therapy currently exists. A common feature of TSEs is<br />

the deposition of insoluble aggregates of <strong>di</strong>sease-associated prion protein (PrP Sc ), the post-translationally refolded<br />

and partially protease-resistant isoform of normal cellular prion protein (PrP C ).(1)<br />

The interactions between proteins are important for many biological functions and prion <strong>di</strong>seases have been linked<br />

to protein fol<strong>di</strong>ng problems which lead to the build-up of insoluble protein plaques in the brain or other organs.<br />

The ability to design novel ligands that inhibit such interactions remains one of the major challenges in<br />

contemporary biome<strong>di</strong>cal science.(2)<br />

We aimed to identify anti-prion lead compounds with unprecedented molecular frameworks. As the result of the<br />

structural similarity of <strong>di</strong>ketopiperazines (DPKs) to peptides, their appearance in biologically active natural<br />

products has inspired the use of DKPs to circumvent the limitations of peptides. We propose DKPs as modulators<br />

of critical protein-protein interactions in prion <strong>di</strong>sease, inhibiting the conversion of PrP C to PrP Sc .<br />

For the design of our compounds, we noticed that most anti-prion molecules possess a symmetrical bifunctional<br />

structure, consisting of two moieties joined via an appropriate spacer. Since we consider the DKP nucleus as a<br />

privileged structure in the modulation of protein-protein interactions, we have synthesized some <strong>di</strong>meric<br />

compounds by the three-component double condensation of 1,4-<strong>di</strong>acetyl-2,5-piperazine<strong>di</strong>one with aromatic<br />

aldehydes. To study the importance, if any, of an appropriate planar conformation some monoreduced and a<br />

saturated derivative were also synthesized.<br />

A cell screening assay was used to test anti-prion activity across the synthesized compounds. The ability to reduce<br />

PrP Sc concentrations in scrapie-infected mouse hypothalamus (ScGT1) cells was determined from Western blot<br />

densitometry of the PK-resistant PrP Sc . We report the identification of a novel bifunctional DKP derivative, which<br />

exhibited activity in the low micromolar range against prion replication in ScGT1 cells, while showing low<br />

toxicity.<br />

References<br />

(1) Hu, W.; Kieseier, B.; Frohman, E.; Eagar, T. N.; Rosenberg, R. N.;Hartung, H.-P.; St€uve, O. Prion proteins: physiological<br />

functions and role in neurological <strong>di</strong>sorders. J. Neurol. Sci. 2008, 264, 1–8.<br />

(2) Breinbauer, R.;, Vetter, I. R;. Waldmann, H.. From protein domains to drug can<strong>di</strong>dates-natural products as gui<strong>di</strong>ng<br />

principles in the design and synthesis of compound libraries. Angew. Chem. Int. Ed. 2002, 41, 2878 – 2890.<br />

P-24


SOUNDS OF SILENCE: INNOVATIVE APPROACH FOR THE<br />

IDENTIFICATION OF NOVEL GPCR-MODULATOR CHEMICAL<br />

ENTITIES<br />

Stephan Schann, Stanislas Mayer, Mélanie Frauli, Christel Franchet, Pascal Neuville<br />

Domain Therapeutics, BioParc, Boulevard Sebastien Brant, F-67400 Illkirch, France<br />

137<br />

Allosteric modulators (AMs) represent an emerging and promising approach for the regulation of G-protein<br />

coupled receptors (GPCRs), the best family of drug targets with nearly half of the current market for therapeutic<br />

agents. AM’s use is associated with major advantages such as subtype selectivity, saturable effects, mimicking of<br />

physiological response and more tractable chemistry. To date, two GPCR AMs have entered the market:<br />

Cinacalcet, a positive allosteric modulator (PAM) of calcium sensing receptor for the treatment of<br />

hyperparathyroi<strong>di</strong>sm and Maraviroc, a negative allosteric modulator (NAM) of CCR5 for the treatment of HIV. 1,2<br />

This relatively low number can be explained by the lack of technology specifically de<strong>di</strong>cated for their <strong>di</strong>scovery.<br />

The standard process used in the Pharmaceutical industry consists in performing an HTS screening with a<br />

functional assay using specific con<strong>di</strong>tions and a single readout. This process undersamples the chemical space<br />

surroun<strong>di</strong>ng potential therapeutic can<strong>di</strong>date for a chosen GPCR. 3 At Domain Therapeutics we are using a <strong>di</strong>fferent<br />

strategy consisting in identifying first a broad set of GPCR binders using a FRET-based bin<strong>di</strong>ng assay 4 and then<br />

characterizing these binders with multiple functional test para<strong>di</strong>gm to make sure no active AM is missed. This<br />

process enables detection of PAMs, NAMs but also silent allosteric modulators (SAMs), which are binders devoid<br />

of any functional activity. These SAMs constitute a class of molecules not detectable with functional assay and<br />

that represent a unique source of chemical <strong>di</strong>versity.<br />

References:<br />

(1) Conn, PJ.; Christopoulos, A.; Lindsley, CW. Nat. Rev. Drug Disco. 2009, 8, 41-54.<br />

(2) Bridges, TM.; Lindsley, CW. ACS Chem. Biol. 2008, 3(9), 530-41.<br />

(3) Leach, K.; Sexton, PM.; Christopoulos, A. TiPS 2007, 28(8), 382-9.<br />

(4) Tahtaoui, C.; Guillier, F.; Klotz, P.; Galzi, JL. ; Hibert, M.; Ilien, B. J. Med. Chem. 2005, 48, 7847-59.<br />

P-25


138<br />

LIST of PARTICIPANTS


Allegretti Marcello Italy 43<br />

Ambrosini Dario Italy 125<br />

Angeli Piero Italy<br />

Baringhaus Karl-Heinz Germany 63<br />

Bentley Jonathan UK 109<br />

Berk Barkin Turkey 101, 113<br />

Bolognesi Maria Laura Italy 25, 108, 124, 128, 136<br />

Bongarzone Salvatore Italy 128, 136<br />

Booth Raymond G USA 102<br />

Borza István Hungary<br />

Boss Christoph Switzerland 85<br />

Bouvier Michel Canada 39<br />

Bovino Clara Italy<br />

Brandt Wolfgang Germany<br />

Buccioni Michela Italy 115, 116, 127<br />

Cano Montserrat Spain<br />

Cappellacci Loredana Italy 114<br />

Carrieri Antonio Italy 104, 118<br />

Congreve Miles S UK 61<br />

Costanzi Stefano USA 101, 113<br />

Cristalli Gloria Italy 115, 116, 117, 122, 127<br />

Dal Ben Diego Italy 115, 116, 117, 122, 127<br />

De Amici Marco Italy 69<br />

Del Bello Fabio Italy 104, 119, 120, 129<br />

Di Fabio Romano Italy 79, 81<br />

Donati Daniele Italy 22<br />

Dukat Malgorzata USA 106<br />

Enzensperger Christoph Germany 51<br />

Farande Aniket Italy 119, 120, 129<br />

Farande Yogita Italy 120<br />

Farouz Francine USA 87<br />

Forlani Roberto Italy<br />

139


140<br />

Frackenpohl Jens Germany 130<br />

Froestl Wolfgang F Switzerland<br />

Gaviraghi Giovanni Italy 93<br />

Ghelfi Francesca Italy 118, 119<br />

Ghiron Chiara Italy 35<br />

Giacchè Nicola Italy 131<br />

Giacobbe Dania Italy 115, 122<br />

Giannella Mario Italy 104, 119, 120, 129<br />

Giar<strong>di</strong>na Giuseppe Italy<br />

Giar<strong>di</strong>nà Dario Italy<br />

Giorgioni Gianfabio Italy 125<br />

Gitto Rosaria Italy 121<br />

Glennon Richard A USA 24<br />

Goller Andreas H Germany<br />

Gualtieri Fulvio Italy<br />

Hacksell Uli USA 17<br />

Hubbard Roderick E UK 31<br />

Ijjaali Ismail France 100<br />

Kerns Edward H USA 33<br />

Lankau Hans J Germany<br />

Lange Udo E W Germany<br />

Leeson Paul D UK 37<br />

Leurs Rob The Netherlands 21, 45, 103<br />

Li Xue-Juan P.R. China 107<br />

Makriyannis Alexandros USA 27, 53<br />

Marchioro Carla Italy<br />

Martinelli Adriano Italy 23<br />

Mason Jonathan S Denmark 71<br />

Melchiorre Carlo Italy 25, 108, 124, 134<br />

Merlo Pich Emilio Italy 79<br />

Milelli Andrea Italy 124, 134


Minetto Giacomo Italy 133<br />

Mogemark Mickael Sweden<br />

Mohr Klaus Germany 69<br />

Morphy Richard UK 73<br />

Mosti Luisa Italy<br />

Moureau Florence Belgium<br />

Neumeyer John L USA<br />

Nilsson Karolina Sweden<br />

Ofner Silvio Switzerland<br />

Panmand Deepak S Italy 115, 117<br />

Pegoraro Stefano Germany<br />

Pellicciari Roberto Italy 49, 131, 135<br />

Piergentili Alessandro Italy 104, 119, 120, 129<br />

Pigini Maria Italy 104, 118, 119, 120<br />

Piomelli Daniele USA 55<br />

Porter Roderick Italy 81<br />

Portoghese Philip S USA 59<br />

Quaglia Wilma Italy 104, 119, 120, 129<br />

Reid Mark UK<br />

Riva Carlo Italy 123<br />

Roecker Anthony J USA 83<br />

Ronzoni Silvano Italy<br />

Rosini Michela Italy 108, 124<br />

Sabbatini Paola Italy 135<br />

Saunders John USA<br />

Schann Stephan France 47, 137<br />

Schaus John M USA<br />

Schell Peter Sweden<br />

Schoen Uwe Germany<br />

Schulte Marvin K USA 67<br />

Selent Jana Spain 110<br />

Simoni Elena Italy 108, 134<br />

141


142<br />

Smit Martine J The Netherlands 45<br />

Smits Rogier A The Netherlands 103<br />

Staderini Matteo Spain 136<br />

Svenningsson Per J L Sweden 65<br />

Szentirmay Eva Hungary<br />

Thomas Ajiroghene Italy 116, 127<br />

Thomas Russell J Italy 20, 91, 118, 119, 129, 133<br />

Timmerman Henk The Netherlands 26<br />

Triggle David J USA 95<br />

Tuccinar<strong>di</strong> Tiziano Italy 105<br />

Tunici Patrizia Italy 89<br />

Van der Schyf C. (Neels) J USA 75<br />

Weber Lutz Germany<br />

You<strong>di</strong>m Moussa B H Israel 77<br />

Zaman Guido J R The Netherlands 41


ACKNOWLEDGEMENTS<br />

info@coneroazzurro.it<br />

www.coneroazzurro.it<br />

143


144<br />

Farmacia Angelini<br />

Dr. Daniele Maria<br />

Miips Informati tiica<br />

Comune <strong>di</strong> Camerino<br />

Comune <strong>di</strong> Matelica<br />

Associazione<br />

Corsa alla Spada<br />

ARTELITO TELITO<br />

SOLUZIONI LITOGRAFICHE RAFICHE<br />

University of Camerino<br />

Photos by Maurizio Bran<strong>di</strong> and Pierluigi Lana<br />

Comune <strong>di</strong> Castelraimondo<br />

Comunità Montana<br />

<strong>di</strong> San Severino Marche<br />

illuminazione<br />

Fondaziion one<br />

MA..SO..GI.. ..BA..

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!